Vous êtes sur la page 1sur 188

Volume 142

Number 3

August 6, 2010

www.cell.com

Noncoding RNAs
Orchestrate p53
Integrated solutions from BD

TE D Y ZE CE
LA AN AL DU
O XP N RO

IS

A
E

P
SYSTEMATIC
EXCELLENCE
SM

Powerful platforms and flexible


solutions that enable your work to flow.
From isolation through production, today Systematic ExcellenceSM is our commitment to
BD Biosciences delivers high quality products and deliver both best-in-class and highly integrated
services, meeting your needs at every step in the products and services to help you reduce risk and
cell-based workflow. to ensure superior quality in the production of
clinical-grade applications.
As research moves toward clinical trials and
ultimately new cell therapies, you can rely on BD Systematic ExcellenceSM: Powering the
BD’s proven experience in delivering quality GMP next generation of tools and systems to
products and services as we deliver the next transform disease management in the next
generation of tools and systems to help you trans- decade.
form disease management in the next decade.
Visit us at bdbiosciences.com/stemcells.
BD Biosciences
For Research Use Only. Not for use in diagnostic or therapeutic procedures. 2350 Qume Drive
BD, BD Logo and all other trademarks are property of Becton, Dickinson and Company. © 2010 BD San Jose, CA 95131
23-11458-00 bdbiosciences.com
FlowCytomix ™
5 T helper cell profiles in 1 kit!
FlowCytomix™ are bead-based immunoassays for
multiple analyte detection on the flow cytometer.
• Measure up to 20 analytes simultaneously from
Th1 only 25 µl of sample
APC
• Choose pre-configured panels or create your
own custom combinations
Th2 • Easy-to-use analysis software included
• For use on most flow cytometers, including
single laser instruments
Naїve
Th
Th1/ Th2/ Th9/ Th17/ Th22
Th9

Multiple Analyte Detection Kits!


Th17 NEW! Human Th Differentiation 13plex
Cat. No. BMS817FF
(IFN-у, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12 (p70),
IL-13, IL-17A, IL-22, TNF-α)
Th22

NEW! Mouse Differentiation 13plex


Cat. No. BMS822FF
(IFN-у, IL-1α, IL-2, IL-4, IL-5, IL-6, IL-10, IL-13, IL-17A,
IL-21, IL-22, IL-27, TNF-α)

Visit www.eBioscience.com
and search FlowCytomix.

www.eBioscience.com ORDER 888.999.1371 • TECH 888.810.6168 • INQUIRIES info@eBioscience.com


Twin Guard Series™ with
-86 Dual˚Cool Technology

Nothing Beats 100% Protection.


Except 200% Protection.
If you’re preserving the work of a lifetime, this -86°C freezer is for you.
-86˚C SANYO introduces Twin Guard Series™, the most reliable dual, redundant autocascade
freezer for the most critical -86°C ultra-low applications ever. The V.I.P.™ insulated
MDF-U500VXC is powered by two completely separate -86 Dual˚Cool refrigeration
systems, each built to run efficiently – alone or even better together in
energy-saving EcoMode™ – to safely preserve whatever you put inside.
A B
Learn more. Visit www.twinguardseries.com or call 800-858-8442.

Pictured: The 18.3 cu.ft. V.I.P.™ insulated MDF-U500VXC. Includes integrated LCD performance monitor and digital controller
for comprehensive system management, data logging, remote communications, alarms, predictive performance and
validation. Maintenance free, filterless design.

FREEER
OF F A LUE ]
3 ,7 9 5 V
[$
! Limited Time Free Offer. ($3,795 Value)
For a third level of security during brief power outages SANYO offers a free,
built-in Liquid N2 or Liquid CO2 back-up system with the purchase of any
MDF-U500VXC now through September 30, 2010.
www.twinguardseries.com
GREEN
product

©2010 Sanyo Biomedical


Editor Editorial Board Douglas Green Dana Pe’er
Emilie Marcus Abul Abbas Leonard Guarente Kathrin Plath
C. David Allis Taekjip Ha Carol Prives
Senior Deputy Editor
Geneviève Almouzni Daniel Haber Klaus Rajewsky
Elena Porro
Uri Alon Ulrike Heberlein Venki Ramakrishnan
Scientific Editors Angelika Amon Nobutaka Hirokawa Rama Ranganathan
Karen Carniol Johan Auwerx Mark Hochstrasser Anne Ridley
Connie M. Lee Richard Axel Arthur Horwich James Roberts
Fabiola Rivas Cori Bargmann Tony Hunter Alexander Rudensky
Lara Szewczak Konrad Basler James Hurley Helen Saibil
Bonnie Bassler Richard Hynes Joshua Sanes
Leading Edge Editor
David Baulcombe Thomas Jessell Randy Schekman
Orla M. Smith
Jeffrey Benovic Narry Kim Ueli Schibler
Associate Editor Carolyn Bertozzi Mary-Claire King Joseph Schlessinger
Robert Kruger Wendy Bickmore David Kingsley Hans Schöler
Elizabeth Blackburn Frank Kirchhoff Trina Schroer
Assistant Editor Joan Brugge Richard Kolodner Geraldine Seydoux
Michaeleen Doucleff Lewis Cantley John Kuriyan Kevan Shokat
Senior Managing Editor Joanne Chory Robert Lamb Pamela Sklar
Meredith Adinolfi David Clapham Mark Lemmon Nahum Sonenberg
Andrew Clark Beth Levine James Spudich
Deputy Managing Editor Hans Clevers Wendell Lim Paul Sternberg
Jackie M. Smith Stephen Cohen Jennifer Lippincott-Schwartz Bruce Stillman
Illustration and Pascale Cossart Dan Littman Azim Surani
Design Supervisor Jeff Dangl Richard Losick Keiji Tanaka
Nathan E. Bress Ted Dawson Scott Lowe Craig Thompson
Pier Paolo di Fiore Tom Maniatis Robert Tjian
Lead Illustrator Marileen Dogterom Matthias Mann Jürg Tschopp
Andrew A. Tang Julian Downward Kelsey Martin Ulrich von Andrian
Bruce Edgar Joan Massagué Gerhard Wagner
Illustrators
Steve Elledge Iain Mattaj Detlef Weigel
Yvonne Blanco
Anne Ephrussi Satyajit Mayor Alan Weiner
Kate Mahan
Ronald Evans Ruslan Medzhitov Jonathan Weissman
Production Staff Witold Filipowicz Craig Mello Matthew Welch
Reyna Clancy Marco Foiani Tom Misteli Tian Xu
Elaine Fuchs Tim Mitchison Shinya Yamanaka
Editorial Assistant
Yukiko Goda Alex Mogilner Marino Zerial
Mary Beth O’Leary
Stephen Goff Paul Nurse Xiaowei Zhuang
Joe Goldstein Roy Parker Huda Zoghbi

Cell Office
Cell, Cell Press, 600 Technology Square, 5th Floor, Cambridge, Massachusetts 02139
Phone: (+1) 617 661 7057, Fax: (+1) 617 661 7061, E-mail: celleditor@cell.com
Online Publication: http://www.cell.com

Cell (ISSN 0092-8674) is published biweekly by Cell Press, 600 Technology Square, 5th Floor, Cambridge, Massachusetts 02139. The institutional subscription rate for
2010 is $1,360 (US and Canada) or $1,532 (elsewhere). The individual subscription rate is $202 (US and Canada) or $305 (elsewhere). The individual copy price is $50.
Periodicals postage paid at Boston, Massachusetts and additional mailing offices. Postmaster: send address changes to Elsevier Customer Service Americas,
Cell Press Journals, 11830 Westline Industrial Drive, St. Louis, MO 63146, USA.
The paper used in this publication meets the requirments of ANSI/NISO Z39.48-1992 (Permanence of Paper). Printed by Dartmouth Printing Company, Hanover, NH.
CIM-Plate 16
E-Plate 16

xCELLigence RTCA DP System

Leave endpoint assays behind


and move to real time
CIM-Plate 16 Monitor cell invasion and migration continuously in
real time. Go beyond the limits of endpoint assays by using
Lid ——————————————————————
the xCELLigence RTCA DP Instrument and CIM-Plate 16
for label-free measurement of physiologically relevant cell
Upper
Chamber responses.
(cells) ——————————————————
Current Assays*
Membrane
with
pores ——————————————
Sensor on
bottom
————————————————————————— surface

———————————————— Lower Chamber


(media, chemoattractant)

* Boyden Chamber, transwell


CIM-Plate 16 for cell invasion and migration analysis on
the xCELLigence RTCA DP System.
For more information, visit www.xcelligence.roche.com

For life science research only.


Not for use in diagnostic procedures. Roche Diagnostics Corporation
XCELLIGENCE is a trademark of Roche. CIM-PLATE, E-PLATE and ACEA BIOSCIENCES Roche Applied Science
are registered trademarks of ACEA Biosciences, Inc. in the U.S. Indianapolis, Indiana
© 2010 Roche Diagnostics. All rights reserved.
Cell Press Display Advertising
Midwest/Southeast/Eastern Canada:
President & CEO Inez Herrero, ph: 585 678 4395; fax: 585 678 4722;
Lynne Herndon e-mail: i.herrero@elsevier.com

Editor in Chief, Vice President of Content Development Mid-Atlantic/New England: Vikki Macomber, ph: 508 928
Emilie Marcus 1255; fax: 508 928 1256; e-mail: v.macomber@elsevier.com

Vice President of Marketing and Publishing West Coast/Western Canada/Asia: Lynne Stickrod, ph: 415
Els Bosma 931 9782; fax: 415 520 6940; e-mail: l.stickrod@elsevier.com

Vice President of Web Development and Operations UK/Europe: James Kenney, ph: +44 20 7424 4216; fax: +44 18
Keith Wollman 6585 3136; e-mail: j.kenney@elsevier.com

Director of Marketing Classified Advertising


Jonathan Atkinson United States and Canada:
Gordon Sheffield, Key Account Manager, ph: 617 386 2189; fax:
Production Supervisor
617 397 2805; e-mail: g.sheffield@elsevier.com
Meredith Adinolfi
UK, Europe, and Asia:
Operations Supervisor
Sabrina Dodge, Key Account Manager, ph: +44 20 7424 4997;
Daniel Fauxsmith
fax: +44 18 6585 3136; e-mail: s.dodge@elsevier.com
Press Officer
Cathleen Genova

ª2010 Elsevier Inc. All rights reserved. or ideas contained in the material herein. Because of rapid advances in
This journal and the individual contributions contained in it are protected the medical sciences, in particular, independent verification of diagnoses
under copyright by Elsevier Inc., and the following terms and conditions and drug dosages should be made. Although all advertising material is
apply to their use: expected to conform to ethical (medical) standards, inclusion in this
Photocopying: publication does not constitute a guarantee or endorsement of the quality
Single photocopies of single articles may be made for personal use as al- or value of such product or of the claims made of it by its manufacturer.
lowed by national copyright laws. Permission of the Publisher and payment Reprints:
of a fee are required for all other photocopying, including multiple or system- Article reprints are available through Cell’s reprint service; for informa-
atic copying, copying for advertising or promotional purposes, resale, and tion, contact Nicholas Pavlow (e-mail: n.pavlow@elsevier.com; ph: (+1)
all forms of document delivery. Special rates are available for educational 212 633 3960).
institutions that wish to make photocopies for nonprofit educational class- Subscription Orders and Inquiries:
room use. For information on how to seek permission, visit www.elsevier. Mail, fax, or e-mail address changes to Elsevier Customer Service Amer-
com/permissions or call (+44) 1865 843830 (UK) / (+1) 215 239 3804 (US). icas, allowing 4–6 weeks for processing. Lost or damaged issues will be
Permissions: replaced, subject to availability, if Cell Press is notified within the claim
For information on how to seek permission, visit www.elsevier.com/ period (US and airmail delivery: 3 months from issue date; surface deliv-
permissions or call (+44) 1865 843830 (UK) / (+1) 215 239 3804 (US). ery: 4 months from issue date). Periodical delivery in the US can take up
Derivative Works: to 3 weeks. Airmail delivery can take 2–4 weeks.
Subscribers may reproduce tables of contents or prepare lists of articles The price of a single copy of Cell is $50 (excluding special issues). All
including summaries for internal circulation within their institutions. orders must be prepaid and in writing. Please include the volume and is-
Permission of the Publisher is required for resale or distribution outside sue number, payment (check or credit card, MasterCard, Visa, or Amer-
the institution. Permission of the Publisher is required for all other deriv- ican Express only), and a delivery address. Allow 4–6 weeks for delivery.
ative works, including compilations and translations (please consult Mailing address: Elsevier Customer Service Americas, Cell Press
www.elsevier.com/permissions). Journals, 11830 Westline Industrial Drive, St. Louis, MO 63146,
USA. Toll-free phone within USA/Canada: 866 314 2355; phone for
Electronic Storage or Usage:
outside US/Canada: (+1) 314 453 7038; fax: (+1) 314 523 5170; e-mail:
Permission of the Publisher is required to store or use electronically any
subs@cell.com; internet: www.cellpress.com or <www.cell.com>.
material contained in this journal, including any article or part of an article
(please consult www.elsevier.com/permissions). Except as outlined Funding Body Agreements and Policies:
above, no part of this publication may be reproduced, stored in a retrieval Elsevier has established agreements and developed policies to allow au-
system, or transmitted in any form or by any means, electronic, mechan- thors whose articles appear in journals published by Elsevier to comply
ical, photocopying, recording, or otherwise, without prior written permis- with potential manuscript archiving requirements as specified as condi-
sion of the Publisher. tions of their grant awards. To learn more about existing agreements and
Notice: policies, visit www.cellpress.com.
No responsibility is assumed by the Publisher for any injury and/or damage Guide for Authors:
to persons or property as a matter of products liability, negligence, or other- For a full and complete guide for authors, please go to www.cell.com/
wise, or from any use or operation of any methods, products, instructions, authors.
REMARKABLY FAST

Phusion® High-Fidelity DNA Polymerase


With Phusion® High-Fidelity DNA Polymerase, there is no need to compromise any aspect of your PCR performance.
A superior choice for cloning, this recombinant polymerase has an error rate 50-fold lower than Taq DNA Polymerase,
combining extreme precision with unparalleled speed and robustness.

Advantages:
Not all PCR polymerases are created equal
• Extreme fidelity • High yield
• High speed • Specificity
• Robustness

Go to www.neb.com/phusion to find out


how Phusion High-Fidelity DNA Polymerase can
improve your PCR performance.

Produced by Distributed by
Amplification of a 3.8 kb fragment from the human beta globin gene clearly illustrates the
extreme speed and robustness offered by using Phusion DNA Polymerase. Reactions were
performed according to the suppliers’ recommendations using varying extension times
(shown above gel).

Phusion is a registered trademark of Finnzymes Oy.

DNA AMPLIFICATION PROTEIN EXPRESSION GENE EXPRESSION


CLONING & MAPPING RNA ANALYSIS
& PCR & ANALYSIS & CELLULAR ANALYSIS
www.neb.com
Leading Edge
Cell Volume 142 Number 3, August 6, 2010

IN THIS ISSUE
STEM CELL SELECT

ANALYSIS
347 Stitching Together Cross-border Research C. Macilwain

CORRESPONDENCE
350 Successes of Genome-wide Association Studies R.J. Klein, X. Xu, S. Mukherjee,
J. Willis, and J. Hayes
351 Strategies for Genetic Studies of Complex Diseases K. Wang, M. Bucan, S.F.A. Grant,
G. Schellenberg, and H. Hakonarson

353 Response: Why It Is Time to Sequence J. McClellan and M.C. King

PREVIEWS
356 Fyn-Tau-Amyloid: A Toxic Triad C. Haass and E. Mandelkow

358 Noncoding RNAs: The Missing ‘‘Linc’’ A.M. Barsotti and C. Prives
in p53-Mediated Repression
360 Stem Cells and DNA Damage: Persist or Perish? A.A. Lane and D.T. Scadden

362 Mitochondrial Matrix Reloaded with RNA T. Endo, K. Yamano, and T. Yoshihisa

MINIREVIEW

364 A MAP for Bundling Microtubules C.E. Walczak and S.L. Shaw

PERSPECTIVE
368 The Pioneer Round of Translation: L.E. Maquat, W.-Y. Tarn, and O. Isken
Features and Functions

SNAPSHOT

496 Nonhomologous DNA End Joining (NHEJ) M.R. Lieber and T.E. Wilson
www.phosphosite.org

A Comprehensive Online
Protein Modification Resource
provided by Cell Signaling Technology with grant support from the NIH

Cell Signaling Technology® and PhosphoSitePlus® are trademarks of Cell Signaling Technology, Inc.
© 2010 Cell Signaling Technology, Inc.
PhosphoSitePlus® v3.0 (PSP) is an open web • Expansive and continuously curated content
resource that integrates encyclopedic information • Molecular rendering to visualize the location of modification sites
on experimentally determined protein modification
• On-the-fly generation of kinase substrate sequence logos
sites, upstream and downstream regulation of
• Browsing of high-throughput content by disease, cell line, and tissue
these modifications, and powerful analytical tools
for investigating the structural and biological • New search interfaces that retrieve modification sites and proteins by
significance of protein modifications. subcellular locations, sequence and motifs, domains, responsiveness
to treatments, disease, tissue, and cell type

Unparalleled product quality, validation and technical support.


www.cellsignal.com
Orders (toll-free) 1-877-616-2355 | Technical support (toll-free) 1-877-678-8324 support@cellsignal.com | Inquiries info@cellsignal.com | Environmental Commitment eco.cellsignal.com
Articles
Cell Volume 142 Number 3, August 6, 2010

375 Direct Reprogramming of Fibroblasts M. Ieda, J.-D. Fu, P. Delgado-Olguin,


into Functional Cardiomyocytes V. Vedantham, Y. Hayashi, B.G. Bruneau,
by Defined Factors and D. Srivastava

387 Dendritic Function of Tau Mediates L.M. Ittner, Y.D. Ke, F. Delerue, M. Bi, A. Gladbach,
Amyloid-b Toxicity in €
J. van Eersel, H. Wolfing, B.C. Chieng, M.D.J. Christie,
Alzheimer’s Disease Mouse Models I.A. Napier, A. Eckert, M. Staufenbiel, E. Hardeman,

and J. Gotz

398 Single-Stranded DNA Transposition B. Ton-Hoang, C. Pasternak, P. Siguier,


Is Coupled to Host Replication C. Guynet, A.B. Hickman, F. Dyda, S. Sommer,
and M. Chandler

409 A Large Intergenic Noncoding RNA M. Huarte, M. Guttman, D. Feldser, M. Garber,


Induced by p53 Mediates Global Gene M.J. Koziol, D. Kenzelmann-Broz, A.M. Khalil,
Repression in the p53 Response O. Zuk, I. Amit, M. Rabani, L.D. Attardi, A. Regev,
E.S. Lander, T. Jacks, and J.L. Rinn

420 A Minimal Midzone Protein Module P. Bieling, I.A. Telley, and T. Surrey
Controls Formation and Length of
Antiparallel Microtubule Overlaps

433 Insights into Antiparallel Microtubule R. Subramanian, E.M. Wilson-Kubalek, C.P. Arthur,
Crosslinking by PRC1, a Conserved M.J. Bick, E.A. Campbell, S.A. Darst, R.A. Milligan,
Nonmotor Microtubule Binding Protein and T.M. Kapoor

444 Aurora Kinases and Protein Phosphatase 1 Y. Kim, A.J. Holland, W. Lan, and D.W. Cleveland
Mediate Chromosome Congression
through Regulation of CENP-E

456 PNPASE Regulates RNA Import G. Wang, H.-W. Chen, Y. Oktay, J. Zhang, E.L. Allen,
into Mitochondria G.M. Smith, K.C. Fan, J.S. Hong, S.W. French,
J.M. McCaffery, R.N. Lightowlers, H.C. Morse III,
C.M. Koehler, and M.A. Teitell

468 Plzf Regulates Germline Progenitor R.M. Hobbs, M. Seandel, I. Falciatori, S. Rafii,
Self-Renewal by Opposing mTORC1 and P.P. Pandolfi

(continued)
Working with stem cells ?

Stem Cell Promotion !


www.promocell.com/promotion

Go for PromoCell !
Your partner for stem cell cultivation
 Mesenchymal Stem Cells & Pericytes
CD34+ & CD133+ Progenitor Cells

 CD14+ Monocytes new
www.promocell.com

PromoCell GmbH
North America: 1 - 866 - 251 - 2860 (toll free)
Deutschland: 0800 - 776 66 23 (gebührenfrei)
United Kingdom: 0800 - 96 03 33 (toll free)
France: 0800 90 93 32 (ligne verte)
Other countries: +49 6221 - 649 34 0
480 Myc-Nick: A Cytoplasmic Cleavage M. Conacci-Sorrell, C. Ngouenet, and R.N. Eisenman
Product of Myc that Promotes a-Tubulin
Acetylation and Cell Differentiation

ERRATUM

494 SIRT1 Suppresses b-Amyloid Production G. Donmez, D. Wang, D.E. Cohen, and L. Guarente
by Activating the a-Secretase Gene ADAM10

ANNOUNCEMENTS

POSITIONS AVAILABLE

On the cover: When p53 is active in cells, it, like an orchestra conductor, coordinates the
activation and repression of many cellular players. Among these are long intergenic noncod-
ing RNAs (lincRNAs) that contribute to the balanced modulation of transcription in the p53
response. Huarte et al. (pp. 409–419) show that lincRNA-p21, represented as a baton, is a di-
rect transcriptional target of p53 and is necessary for the repression of many target genes,
particularly those involved in apoptosis downstream of p53 activation. Artwork by Lauren
Solomon, John Rinn, Maite Huarte, and Sigrid Hart, Broad Institute; adapted from artwork
by iStockphoto.
R&D Systems Tools for Cell Biology Research™

Proteins
R&D Systems Quantikine® ELISAs
Antibodies

ELISAs
The Most Referenced Immunoassays
A direct measure of product quality is the frequency of citations in the scientific literature. R&D Systems has more
Assay Services
than 20 years of experience designing, testing, and optimizing the most cited ELISA kits in the world. Find out why
MultiAnalyte Profiling scientists trust R&D Systems ELISAs more than any other brand.

Activity Assays R&D Systems is the Most Referenced ELISA Manufacturer NEW Quantikine ELISA Kits
 α1-Acid Glycoprotein
Stem Cells  Angiopoietin-like 3
 Cathepsin V
ELISpot Kits  Clusterin
17.3%  Dkk-1
R&D Systems  EGF R/ErbB1
Flow Cytometry 41.8% 7.9%  EG-VEGF/PK1
 Fetuin A
Cell Selection 3.7%  FGF-21
 Galectin-3
2.5%  Gas 6
2.1%  GDF-15
1.6%  IL-17A/F Heterodimer
1.4%
 IL-19
 Lipocalin-2/NGAL
 MBL
 Proprotein Convertase 9/PCSK9
Approximately 42% of Referenced Immunoassays are Developed and Manufactured by  Periostin/OSF-2
R&D Systems. A survey of 860 manuscripts from 44 journals was conducted to compare the  Progranulin
number of citations specifying the use of R&D Systems ELISAs to the number citing ELISAs from  ST2/IL-1 R4
other commercial sources. A total of 433 ELISA citations referencing immunoassays from 66  Thrombomodulin/CD141
different vendors were identified in the survey.  Tie-1
 TIM-1/KIM-1

For more information visit our website at www.RnDSystems.com/go/ELISA

For research use only. Not for use in diagnostic procedures.

R&D Systems, Inc. www.RnDSystems.com

R&D Systems Europe, Ltd. www.RnDSystems.co.uk

R&D Systems China Co., Ltd. www.RnDSystemsChina.com.cn


Leading Edge

In This Issue

Reprogramming’s Got the Beat


PAGE 375
Reprogramming of fibroblasts to induced pluripotent stem cells suggested that a somatic cell could be reprogrammed into
alternative fates. Ieda et al. now report that a combination of three developmental transcription factors, Gata4, Mef2c, and
Tbx5, rapidly and efficiently reprograms postnatal cardiac or dermal fibroblasts directly into cardiomyocyte-like cells without
passing through a cardiac progenitor state. Thus, reprogramming of endogenous or explanted fibroblasts might provide
a source of cardiomyocytes for regenerative approaches.

Fyn-ally, the Missing Link between Tau and Ab


PAGE 387
The peptide amyloid-b (Ab) and the tau protein are both found in toxic aggre-
gates in the brains of Alzheimer’s disease (AD) patients. In this issue, Ittner
et al. reveal a mechanism by which tau may collaborate with Ab in mediating
AD pathogenesis. The authors show that tau, generally thought of as an axonal
protein, targets Fyn kinase to dendritic spines, where it phosphorylates NMDA
receptors. This leads to increased excitotoxicity, which boosts the toxic effects
of Ab on neurons. Disruption of tau-mediated targeting of Fyn in a mouse model
of AD decreases excitotoxicity via NMDA receptors and ameliorates other path-
ological features associated with Ab.

p53 Gets Linc-ed In


PAGE 409
Mammalian genomes encode numerous noncoding RNAs, including a class of large intergenic noncoding RNAs (lincRNAs)
associated with p53. In this issue, Huarte et al. show that lincRNA-p21 is a direct p53 transcriptional target that also mediates
p53-dependent transcriptional repression through its physical association with hnRNP-K. These results reveal a new
aspect to the p53 transcriptional response and suggest that lincRNAs may serve as key regulatory hubs in transcriptional
pathways.

Lagging Strand Takes the Lead in Transposition


PAGE 398
For most transposons, excision and insertion require double-stranded
templates. In one recently identified family of bacterial insertion sequences
(IS200/IS605), however, the cutting and pasting is done with only a single-
stranded DNA segment. Ton-Hoang et al. now show that these single strands
are preferentially excised and inserted from the lagging strand of replicating
DNA, coupling transposition to replication fork passage. In addition to identifi-
cation of a novel transposition pathway, the unique excision and insertion prop-
erties of the IS200/IS605 family may make them useful tools for probing the
in vivo structures of ssDNA segments.

A-PRC-iating Midzone Dynamics


PAGE 420 and PAGE 433
During cell division, microtubules are arranged in the mitotic spindle to segre-
gate the duplicated chromosomes. In anaphase, tightly bundled antiparallel microtubules in the spindle center form the mid-
zone structure. Using an in vitro reconstitution approach, Bieling et al. show how PRC1, an antiparallel microtubule bundler,
and the processive motor kinesin-4 form the minimal module needed to dynamically organize the core structure of the verte-
brate midzone. Related work from Subramanian et al. provides detailed insight into how PRC1 functions. Using structural and
biophysical approaches, they find that PRC1 dimers contain both structured and unstructured domains that crosslink anti-
parallel microtubules, which allow the protein to track along the overlap at the midzone without substantially resisting relative
filament sliding.

Cell 142, August 6, 2010 ª2010 Elsevier Inc. 335


We set out to bring
Attune
you superior data

ACOUSTIC FOCUSING

from a flow cytometer.


CYTOMETER

Greg Kaduchak, PhD


Director
Instrumentation & Engineering
Flow Cytometry Systems

Mike Ward, PhD


Associate Director
Instrumentation & Engineering
Flow Cytometry Systems

We succeeded…
and changed traditional flow cytometry in the process.
The new Applied Biosystems® Attune™ Acoustic Focusing Cytometer is unlike traditional
flow cytometers. It uses high-frequency sound waves to precisely focus cells before they
pass through the point of interrogation. Now get:
• Ease of use—without sacrificing complete control of your run

• Better clarity—by providing amazing separation of signal peaks

• Higher sample rates—faster rare-event detection

Find out how acoustics is revolutionizing cytometry. Visit us at


www.appliedbiosystems.com/attune.

For research, forensic or paternity use only. Not intended for any animal or human therapeutic or diagnostic use. The trademarks mentioned herein are the property of Life Technologies Corporation or their respective owners. © 2010 Life Technologies Corporation.
Chromosomes Turn on a Phosphate
PAGE 444
Proper orientation of chromosomes in the mitotic spindle ensures accurate
segregation and guards against aneuploidy, a common feature of human
cancers. In this issue, Kim at al. demonstrate that CENP-E, a kinetochore motor
protein, is phosphorylated at a single site by Aurora kinases at the spindle poles
to promote chromosome congression. CENP-E is subsequently dephosphory-
lated by PP1 at the outer kinetochore, enabling bistable orientation. These find-
ings explain how spatially regulated phosphorylation can control chromosome
movements during mitosis.

Protecting the Family Jewels


PAGE 468
The mTORC1-signaling pathway is a critical regulator of cell growth. Aberrant
mTORC1 activation is associated with stem cell depletion through poorly char-
acterized targets. Now, Hobbs et al. demonstrate that Plzf, a transcription factor implicated in germline maintenance,
opposes mTORC1 pathway activity in spermatogonial progenitor cells (SPCs). Further, they show that elevated mTORC1
activity inhibits response of SPCs to GDNF, a niche-derived growth factor required for self-renewal. This study provides
important insight into mechanisms of germline maintenance and defines a model by which mTORC1 activity is detrimental
to stem cell function.

RNA Takes the Autobahn to the Mitochondria


PAGE 456
RNA import into mammalian mitochondria is poorly understood compared
with protein import. Here, Wang and colleagues report the identification of
a mammalian mitochondrial RNA import factor, the enzyme PNPASE. They
find a 20 nucleotide stem-loop RNA structure that can mediate PNPASE-
dependent mitochondrial RNA import. PNPASE-dependent imported RNAs
regulate processing of long mitochondrial RNA transcripts and the translation
of electron transport chain proteins for respiration. The study identifies a
component of the mammalian mitochondrial RNA import pathway and a poten-
tial new approach for selectively targeting RNAs to mitochondria.

Mini-Myc Goes Nonnuclear


PAGE 480
Myc family proteins are nuclear transcriptional regulators that control cell
growth, proliferation, and apoptosis. Here, Conacci-Sorrell et al. report the characterization of Myc-nick, a truncated form
of Myc generated by calpain cleavage of full-length Myc. Myc-nick lacks nuclear localization and DNA binding regions and
is predominantly cytoplasmic. Myc-nick interacts with microtubules and the GCN5 acetyltransferase to promote a-tubulin
acetylation, changes in cell morphology, and terminal differentiation. Proteolytic cleavage may provide a functional switch
from the nuclear, proproliferation form of Myc to a cytoplasmic, prodifferentiation form.

Cell 142, August 6, 2010 ª2010 Elsevier Inc. 337


#ALLåFORå0APERS
3UBMITåYOURåMANUSCRIPTåTODAY
)MPACT¬
&ACTOR


3OURCEåå4HOMSONå2EUTERSå*OURNALå#ITATIONå2EPORTS

4OPICSåCOVEREDåINCLUDE å
¬#HANNELS ¬2ECEPTORS ¬AND¬%LECTRICAL¬3IGNALING ¬3PECTROSCOPY ¬)MAGING ¬/THER¬4ECHNIQUES
¬0ROTEINS¬ ¬-USCLE¬AND¬#ONTRACTILITY
¬"IOPHYSICAL¬4HEORY¬AND¬-ODELING¬ ¬3UPRAMOLECULAR¬!SSEMBLIES
¬#ELL¬"IOPHYSICS ¬"IOENERGETICS
¬0HOTOBIOPHYSICS ¬.UCLEIC¬!CIDS¬
¬-EMBRANES¬ ¬%LECTROPHYSIOLOGY

HTTPSUBMITBIOPHYSJORG
Leading Edge

Stem Cell Select

Lineage commitment requires stem cells to loosen their grip on pluripotency, sequentially picking and
choosing among competing genetic programs to realize specific cell fates. New findings described in this
issue’s Stem Cell Select address the impact of history and cellular memory on differentiation events and
reveal critical molecular mediators of stem cell reprogramming and pluripotency.

A Remembrance of Tissues Past


According to two recent reports (Kim et al., 2010; Polo et al., 2010),
how you achieve stem cell pluripotency has unexpected conse-
quences. Kim et al. show that reprogrammed pluripotent cells from
mice created by somatic cell nuclear transfer differ from those
created through the expression of transcription factor cocktails.
Their findings suggest that cells reprogrammed by somatic cell
nuclear transfer bear greater similarity to embryonic stem cells
than induced pluripotent stem (iPS) cells reprogrammed with defined
factors, that is, at least prior to extensive passaging. Delving into the
molecular basis for this difference reveals that iPS cells retain
residual DNA methylation signatures reflecting their cell type of
Osteogenic colonies from fibroblast-derived
induced pluripotent stem cells stained with alizarin
origin. Similar observations are made by Polo et al. in their examina-
red to detect calcium deposits. Image courtesy of tion of the gene expression patterns and epigenetic marks of
K. Kim. a collection of iPS cells derived from different mouse tissues. The
two reports show that iPS cells retain a form of epigenetic memory
that makes it possible to identify which tissue iPS cells come from and, more importantly, biases their return
to that tissue type upon differentiation. Kim et al. show that further interventions, such as treatment of iPS cells
with chromatin-modifying agents or repeating the cycle of differentiation and reprogramming, alter the impact of
this epigenetic memory, whereas Polo et al. report that the epigenetic memory is dissipated by continuous
passaging of the cells in culture. These findings highlight the notion that pluripotency is not a singular condition
but a diverse spectrum of states. They also suggest that a cell’s history is not easily erased, bringing to mind the
well-known quote from William Faulkner: ‘‘The past is never dead. It’s not even past.’’
K. Kim et al. (2010). Nature. Published online July 19, 2010. 10.1038/nature09342.
J. M. Polo et al. (2010). Nat. Biotechnol. Published online July 19, 2010. 10.1038/nbt1667.

DNA Repair Gives Germ Cells a Fresh Start


DNA methylation is one of the cell’s most stable epigenetic marks.
Hajkova et al. (2010) now provide evidence that base excision repair
(BER) is actively engaged in the removal of these marks in mouse
primordial germ cells (PGCs). On their journey to totipotency,
PGCs go through a dramatic transformation in their chromatin land-
scape, with a major reorganization in their nuclear architecture and
changes in histone and DNA modifications. In cells undergoing these
reprogramming events, the authors observe an activation of BER
pathways, coincident with the removal of methylated cytosines. Chromatin-bound XRCC1 (green) in the male
The activation of BER is also temporally linked to the occurrence pronucleus of the zygote (shown) and in
of single-stranded DNA (ssDNA) breaks, a step in BER. Consistent primordial germ cells suggests the presence of
with a direct role of BER in DNA demethylation, inhibitors of BER single-stranded DNA breaks at the time of DNA
demethylation. Image courtesy of P. Hajkova.
interfere with the removal of methyl-cytosine from the paternal
pronucleus of the zygote, another setting in which active DNA demethylation is reported to occur. In relation
to the recent papers by Kim et al. (2010) and Polo et al. (2010) discussed above, future work may assess whether
promotion of this BER pathway could be used to augment existing methods for the generation of induced plurip-
otent stem cells.
P. Hajkova et al. (2010). Science 329, 78–81.

Cell 142, August 6, 2010 ª2010 Elsevier Inc. 339


,OOKING¬FOR¬WHAT¬#ELL¬
HAS¬PUBLISHED¬IN¬¬
%VOLUTION¬LATELY

TION
LIANå%VOLU
O å6 IS IO N åI Nå-AMMA å   å PPån
å
D A P TS åT PR å 
Tå3PECIES
å!  å!
Rå#ELLS å#ELL å
Få 2 O D å0 H OTORECEPTO REMERå4 å'UCKå* å*OFFEå" " E TW E E N å4WOå9EAS
åO å# RILITYå
RCHITECTURE å+ÙSEMå
3 å0EICHLå, å(YBRIDå3TE
.UCLEARå! - å,ANCTØTå# N O M ESå#AUSES  åPPån R
YS IN Gå Lå ' E ESISå-OTO
3OLOVEIå) å+
RE HONDRIA ECå å EDå#YTOKIN
. U C LE A Rå ANDå-ITOC EU å* 9 å# ELL åå$
A å# O N S E RV
BILITYåOFå å9ANGå39 å,
LLSå$EPRIV
EDåOFå
)NCOMPATI
åå
å#HANGå.( DELå#7 å,Iå2
å# HO Uå *9 å#HEONGå, O LU TI O N åO Få9EASTå#E G ( AM PTONå+ å3EI
,EEå( 9 TIVEå% V ! å3TAEH LIN
APIDå!DAP Nå+ å0ERERAå
P LO ID Yå 5 NDERLIESå2 å.OLLå! å4RIMBLEå2 å7ALTO Gå9EAST +2 åå
!NE U å&LEHARTYå" O N åINå"UDDIN '2 å&OSTERå
2ANCATIå' å0
AV ELKA å.
å  n  IK E å# O O P E RA TI
# å, AT GÎå*0 å&INKå
 å PP IOlLM , Nå! å0REVO
ST å-
å.OVå å Tå$RIVESå" Så-$ å*ANSE
#ELL å E A RD å'ENEåTHA å9ANå# å6INCE
N å" QUENCING
RE E GN INIå3
å6ARIABLEå' HETå. å"EAUVAISå! å'UA
DA
UGHPUTå3E å3IEBAUERå- å"URBANO
å
&,/åISåA RA å- å0 OC å  n  åB Yå ( IG H 4 H RO
FERå+
å# AL DA   å PP
ETERM IN E D EL å5 å0Rß åå
3MUKALLAå3 å.OVå
 å
EQUENCEå$ å4 å3TENZ å0ÊÊBOå3
å+*åå#ELL å *- å-ARICIC å* å3LATKINå-
6ERSTREPEN H O N D RI A Lå 'ENOMEå3 HLERå# å-EYERå- å'OODå å, AA KK ON ENå, å+ELSO
RTALå-ITOC NSONå0, å5 | ) å7IKSTRÙ
Må-
TEå.EANDE RAUSEå* å"RIGGSå!7 å*OH OVICå | Nå: å'US
ICå
!å#OMPLE å+ RA JK | $ å+UCA ON
CEå%VOLUTI
Så ! 3 å0 å"
ALASPINA å- å2UDAN
'REENå2% å-
OT HB ER Gå *- å%GHOLM O N å# O D IN G 3EQUEN
(! å2ONAN
å- å2 n NSTRAINTå
 åPPå MINANTå#O
#ELLå
å!UGå å
IS FO LD IN GåASåAå$O ROSOPHILA

N D U C E D å0 ROTEINå-  å PP ån IM O RP H IC å4RAITSåINå$
TION ) å*ULå å
 UALLYå$
-ISTRANSLA #/å#ELL å OLLINGå3EX ån
M ON Då $! å7ILKEå N E TI C å3 W ITCHå#ONTR å!UGå å åPP ,,å&AMILY

+./8"%
$RU M Få A å' E å# EL L å
V O LU TI O N åO å# AR RO LLå 3 "
E å0 LA N Tå
å% ! NåOFåTH
LATIONåAND ERNERå4 å'OMPELå. å+OP

4HEå2EGU Då%VOLUTIO
4- å3 EL EG UEå*% å7
TE RO D IM E RIZATIONåAN
7ILLIAMSå INå(E n
MEOPROTE  åPPå
E X U A Lå / RI GINSåOFå(O 5å#ELL åå-AYå å
%ARLYå3 OODENOUGHå
å( å*OOå3 å'
,EEå*( å,IN

,OOKå!GAIN
$ISCOVERå-ORE
WWWCELLCOM
A Chronicle of Differentiation Foretold
How do events in embryonic stem cells set the stage for tissue-specific expression later in development? Liber
et al. (2010) follow the molecular events at an enhancer specific for pre-B cell differentiation to show how it is
primed in embryonic stem cells (ESCs) for activation at a later stage. The core of the pathway uncovered involves
a handover between the transcription factor Sox2, which binds the l5-VpreB1 enhancer in ESCs, and Sox4,
which binds the same region in pro-B cells. In ESCs, Sox2 promotes histone 3 lysine 4 di- and trimethylation,
which are activating histone marks, and modulates the recruitment of the Foxd3, a factor that maintains the
enhancer and the surrounding regions in a repressed state. At the pro-B cell stage, the Sox and Fox binding sites
cooperate (the former with Sox4 bound) to fully activate transcription, thereby enhancing expression of l5,
a protein that acts as a critical surrogate for the immunoglobulin light chain during B cell differentiation. The
model proposed by the authors is that factors in ESCs establish active epigenetic marks that then cooperate
with tissue-specific factors to drive transcription during differentiation. Future work is likely to address whether
ESC factors have a more general role in gene priming in this and other lineage commitment events.
D. Liber et al. (2010). Cell Stem Cell 7, 114–126.

Holding Back a Natural Killer Instinct


Becoming a T cell from a hematopoietic progenitor means avoiding
the temptations of other possibilities along the way, including B cell,
macrophage, dendritic cell, and natural killer (NK) cell fates. Three
recent papers reveal a transcription factor needed for T cell precur-
sors to take the final step of commitment (Ikawa et al., 2010; Li et al.,
2010a; Li et al., 2010b). They show that in the absence of this factor,
Bcl11b, would-be T cells can instead be redirected to a natural killer
cell fate. NK cells are lymphocytes that directly kill cells recognized
T cell lineage commitment depends on the tran- as non-self. Li et al. (2010a) demonstrate that Bcl11b is needed
scription factor Bcl11b. Image courtesy of L. Li both for T lineage commitment and to limit self-renewal, as it
and E.V. Rothenberg. promotes the downregulation of both NK cell genes and regulatory
genes that are characteristic of stem and progenitor cells. Future
work may explore which of the regulated genes are direct targets of Bcl11b. Potential functional consequences
of reprogramming T cell progenitors are explored by Li et al. (2010b), who show that even mature T cells can be
converted to NK cells by the loss of Bcl11b, and these reprogrammed cells share with native NK cells the
capacity to hinder tumor establishment in a mouse model of lung metastasis. The findings of Ikawa et al. may
serve as a starting point for efforts to identify the in vivo signals that direct this lineage decision or that maintain
progenitors in a multipotent state. They show in culture that an arrest in T cell development can be promoted by
interleukin-7 (IL-7), thereby maintaining their myeloid and natural killer cell potential, due to failure to induce
Bcl11b. A topic for future examination is to determine how signaling by IL-7 or other cytokines intersects with
Bcl11b at the decision point for T cell commitment.
T. Ikawa et al. (2010). Science 329, 93–96.
L. Li et al. (2010a). Science 329, 89–93.
P. Li et al. (2010b). Science 329, 85–89.

Robert P. Kruger

Cell 142, August 6, 2010 ª2010 Elsevier Inc. 341


“As financial resources become more scarce,
it is more critical to identify research and
researchers who are the most productive and
on the right track.”
Peter Brimblecombe
Professor, Atmospheric Chemistry
School of Environmental Sciences
University of East Anglia, UK

How does your


institution measure up?
Scopus is the optimal data source for research performance measurement.
No other database has so much breadth of content covering so many authors.

With Scopus you can identify authors’ papers, track Now it’s easy to:
their citations and analyze their influence using the • Evaluate and prioritize resource allocation by
Scopus h-index. And, to evaluate the performance of departments or fields
journals, research projects and groups of researchers • Make informed decisions about tenure and promotion
you can measure the performance of a specified • Promote your institution for funding and recruitment
collection of articles.

www.scopus.com
015.A1.0101.F.US © 2009 Eppendorf AG.
Background: electroporated L-mouse cells.
 Electroporation of
mammalian cells

 Electroporation
of bacteria

 Electroporation
of yeasts

 Cell fusion

Outstanding
transfection
rates!

One electroporator– many applications.


Our Multiporator ® is a compact, flexible system A safe and easy method for performing cell fusion:
designed for the electroporation of eukaryotic cells,  Microfusion chamber for viewing and optimization
bacteria and other microorganisms, as well as the of fusion parameters
electrofusion of cells.  Helix chamber for larger cell volumes
 Applications include: monoclonal antibodies, plant
Unique features include Soft Pulse microsecond pulses, protoplast fusion, tumor research, cancer vaccines,
patented* electronically regulated pulses, a gentle cloning mammals
hypoosmolar buffer system and a choice of
electroporation and electrofusion options. For more information visit www.eppendorf.com

*U.S. Pat. 6,008,038

www.eppendorf.com • Email: info@eppendorf.com • Application hotline: 516-515-2258


In the U.S.: Eppendorf North America, Inc. 800-645-3050 • In Canada: Eppendorf Canada Ltd. 800-263-8715
¬,OOK¬!GAIN

$ISCOVER¬-ORE
s¬!CCESS¬TO¬THE¬¬#ELL¬0RESS¬PRIMARY¬RESEARCH¬JOURNALS¬AND¬
¬4RENDS¬REVIEWS¬TITLES ¬ALL¬ON¬THE¬SAME¬PLATFORM
s¬)MPROVED ¬MORE¬ROBUST¬ARTICLE¬AND¬AUTHOR¬SEARCH
s¬6IDEO ¬ANIMATIONS¬AND¬SOUND¬lLES
s¬%ASY¬TO¬NAVIGATE¬HOME¬PAGE ¬ARTICLES¬PAGES¬AND¬ARCHIVE

WWWCELLCOM
Next-gen GWAS.
NOW.
It’s a content revolution.
Up to 50% more coverage of common and rare
variants than all other arrays. Maximum power for
any population.

The Omni family of microarrays can propel


your studies into true next-gen GWAS. With
a clearly defined product path to the future.
Immediate utility. Future flexibility.

Get on the path to next-gen GWAS.

Now is the time. Get started at


www.illumina.com/GWAS
Expand your stem cell library and save
today on the latest books on stem cells
and regenerative medicine

Stem Cells Stem Cell Anthology


Scientific Facts and Fiction From Stem Cell Biology, Tissue Engineering,
Christine Mummery, Ian Wilmut, Anja Van de Stolpe Regenerative Medicine, Cloning and Stem
and Bernard Roelen
Cell Methods
November 2010 | 400 pages | Paperback | Bruce M. Carlson
$79.95 | €57.95 | £48.99 | October 2009 | 450 pp. | Hardback | $150.00 |
ISBN: 9780123815354 €100.00 | £95.00 |AU$222.00 |
ISBN: 9780123756824

Principles of Regenerative
Medicine, 2nd Edition Essential Stem Cell Methods
Anthony Atala and Robert Lanza A Volume in the Reliable Lab Solutions Series
November 2010 | 1400 pages | Hardback | Robert Lanza and Irina Klimanskaya
$199.95 | €143.00 | £125.00 | April 2009 | 628 pp. | Paperback | $75.00 | €50.95
ISBN: 9780123814227 | £45.99 |AU$111.00 | ISBN: 9780123750617

Heart Development and Tissue Engineering


Regeneration, 2-Volume Set Clemens van Blitterswijk, Peter Thomsen, Jeffrey Hubbell,
Ranieri Cancedda, Anders Lindahl Sahlgrenska,
Nadia Rosenthal and Richard P. Harvey
Jerome Sohier and David F. Williams
June 2010 | 1072 pp. | Hardback
| $199.95 | €143.00 | £125.00 | March 2008 | 760 pp. | Hardback | $115.00 |
AU$296.00 | ISBN: 9780123813329 €76.95 | £69.99 |AU$170.00 |
ISBN: 9780123708694

Essentials of Stem Cell


Biology, 2nd Edition Human Stem Cell Manual
Robert Lanza, Roger Pedersen, John Gearhart, E. A Laboratory Guide
Donnall Thomas, Brigid Hogan, James Thomson, Jeanne F. Loring, Robin L. Wesselschmidt and Philip H. Schwartz
Douglas Melton and Sir Ian Wilmut June 2007 | 488 pp. | Spiral bound | $88.95 |
June 2009 | 600 pp. | Hardback | $199.95 €59.95 | £53.99 |AU$132.00 |
| €134.00 | £125.00 | AU$302.00 | ISBN: ISBN: 9780123704658
9780123747297

Handbook of Stem Cells


Foundations of 2-Volume Set with CD-ROM Vol. 1–2
Regenerative Medicine Vol. 1 – Embryonic Stem Cells
Clinical and Therapeutic Applications Vol. 2 – Adult & Fetal Stem Cells
Anthony Atala, Robert Lanza, James Thomson and Robert Lanza, Roger Pedersen, Helen Blau, E. Donnall Thomas,
Robert Nerem John Gearhart, James Thomson, Brigid Hogan, Catherine
Verfaillie, Douglas Melton, Irving Weissman, Malcolm Moore
September 2009 | 750 pp. | Hardback |
and Michael West
$99.95 | €66.95 | £60.99|AU$148.00 |
ISBN: 9780123750853 September 2004 | 1,760 pp. | Hardback | $566.00
| €380.00 | £345.00 | AU$817.00 |
ISBN: 9780124366435

Cell Stem Cell subscribers save 25% on their book order


Secure ordering online at elsevierdirect.com
Enter promo code 28024 at check out
Prices and publication dates subject to change without notice.
Leading Edge

Analysis

Stitching Together Cross-border Research


European research has been hampered by fragmented national research programs. Is joint
programming the answer? Colin Macilwain investigates.

The nations of the European Union (EU) In the last 2 years, however, a new project would focus on neurodegenerative
spent €80 billion ($100 billion) last year on fix has been proposed for the problem: disease research, which is particularly
public nonmilitary research and develop- “joint programming” between national weak and fragmented in Europe (Figure
ment, yet European science still seems research agencies. The idea is to get 1). The European Commission’s research
to have a quality gap compared with the interested nations to band together directorate estimates that US spending in
US. For example, the EU produces 33% of and agree on a detailed strategy for a this area ($856 million, or €527 million, in
research papers published annually world- given research field and then pick-and- 2007) is almost ten times that of Europe
wide but garners only 34% of citations, choose which elements of that strategy ($93 million, or €57 million).
compared with the US, which publishes to collaborate on. “Joint programming Alzheimer’s disease researcher Bart
29% of papers but earns 41% of citations is critical to the future, but it is still in De Strooper of KU Leuven in Belgium
(http://www.nsf.gov/statistics/seind08/). gestation,” says Frank Gannon, former agrees that neurodegenerative dis-
Policymakers believe that one reason for director of the European Molecular Biol- ease research is lagging in Europe. “My
this quality shortfall is the fragmentation ogy Organization and current member impression is that in the United States,
of research spending in Europe. Accord- of the European Research Area Board, much more of a vision has been devel-
ing to the European Commission based which advises the European Commis- oped with regard to problems of aging,
in Brussels, 85% of public research funds sion. “From my point of view, it is cur- and Alzheimer’s in particular,” he says,
in Europe are distributed through sepa- rently the most crucial, single thing that pointing to collaborations such as the
rate national programs run by the EU’s 27 we have to put right.” Alzheimer’s Disease Neuroimaging Ini-
member states. Many think that the way to Last December, the Council of Ministers tiative (http://www.loni.ucla.edu/ADNI/),
get more bang for their euro is to tie these representing the EU member states con- which is supported by several institutes
national activities more closely together. firmed that the first joint programming pilot of the National Institutes of Health (NIH)
Multiple attempts to unite different
European research programs have
failed, however. The €7 billion that the
European Commission allocates for
research annually through its Frame-
work Programme is supposed to nur-
ture cross-border collaborations but
does so one project at a time. And
other efforts—including the long-
established European Cooperation
in Science and Technology (COST)
scheme and plans in the Framework
6 Programme (which ran from 2002
to 2006) for “integrated projects” and
“networks of excellence”—tried and
failed to link the national research pro-
grams together. All of these efforts have
foundered on a mixture of bureaucracy,
nationalism, inertia, and the reluctance
of top researchers, who are able to get
funding in their own countries, to get
involved. “It is very difficult for member
states to come together on a common
basis,” concedes Enda Connolly, chief
executive of Ireland’s Health Research
Board, which distributes €40 million Figure 1. Europe’s Research Landscape
annually for biomedical research in Ire- Brain disease research in Europe, including the study of neurodegenerative diseases such as Alzheimer’s
and Parkinson’s, is weakly coordinated across Europe and receives less investment compared with the
land. “They are all locked into their own US. (x axis, degree of coordination; y axis, spending in Europe relative to the US). Source: European
programs.” Commission, 2008 (ec.europa.eu/research/press/2008/pdf/com_2008_468_en.pdf).

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  347


and the private sector. “We in Europe a common view in Europe of what we markers, developing new therapeutics,
look to the US and are happy if some of need to do in neurodegenerative dis- and infrastructure for large clinical trials.
us are incorporated in their initiatives,” he ease research.” “They could get going by the end of the
says. “We should be much more active Most of the research supported will year,” he says, adding that the initiative
on the international scene.” be in basic neurobiology, including is moving quickly. “To get something
The idea of a combined approach sequencing the complete genomes of from concept to practical action in three
to boost neurodegenerative disease patients to find risk genes, the devel- years is really new in European research
research was first proposed in 2008, opment and standardization of disease policy.”
when France held the rotating presi- biomarkers, and developing better ani- De Strooper adds that as a researcher
dency of the EU and President Nicolas mal models of these diseases. “The big in Belgium, he sees advantages in col-
Sarkozy sought to push aging issues problem is that we don’t have a pipeline laborating with larger countries—France,
higher up Europe’s research agenda. for new therapies, so we need to better Germany, and the UK—that are starting
In July of that year, the European Com- understand the fundamental pathophys- initiatives in neurodegenerative disease
mission issued a paper advocating joint iology of these diseases,” says Rob research. He says that he hopes the
programming as a generic approach to Buckle, program manager for neurosci- Joint Programme will help researchers to
improving coordination among national ences and mental health at the UK Medi- do animal modeling and drug screening,
research bodies. Meanwhile, medical cal Research Council (MRC). “This is an obtain microRNA profiles of patients,
research agencies in France, Germany, opportunity to do things in a different and do deep sequencing and annota-
and the UK were already seeking ways way in Europe.” Buckle says he hopes tion of expression profiles for Alzheim-
to strengthen their respective involve- that the Joint Programme will do work er’s and other diseases. Alzheimer’s
ment in an area of biology where the US that could be relevant to the treatment of researcher and geneticist John Hardy
has a pronounced lead (http://www.mrc. several neurological diseases, including says he hopes that the program can
ac.uk/Utilities/Documentrecord/index. Parkinson’s and motor neuron disease, help researchers to find better biomark-
htm?d=MRC004898). as well as Alzheimer’s. “Mechanistically, ers for neurodegenerative diseases and
Philippe Amouyel, an epidemiologist there’s a lot of overlap between these to use rapid, full-genome sequencing of
at the University of Lille in France, led disorders,” he says. patients to find the risk genes for them.
a working group established in 2008 to Gasser, a neurologist who studies But he admits that it is too early to know
look into the idea of joint programming Parkinson’s disease, says the scien- how it will unfold. “You get involved in the
in the discipline and is now chair of the tific advisory board will confer broadly process, but you never know if there’s
board of the EU Joint Programme for before publishing its research plan in going to be a good outcome,” he says.
Neurodegenerative Disease. (Connolly summer 2011. Three meetings early next “There is a genuine need for it. But you
also sits on the initiative’s five-person year, involving interested biologists, cli- do worry that the optimism behind the
management board.) Initially supported nicians, and social scientists, respec- programme will end up clashing with the
by INSERM, the French biomedical tively, will help the process along. Gas- hard reality of budget cuts.”
research agency, the working group now ser adds that there is a “huge political Although the joint programming pilot
has a small €2 million European Com- will” to make the Joint Programme work has been broadly welcomed, many
mission grant to cover its administration but notes that its directed approach “will experienced researchers and research
costs. never displace bottom-up, undirected administrators have questioned whether
The Joint Programme has appointed basic research that can give us com- the joint programming approach has
a fifteen-member scientific advisory pletely new insights.” enough backing or momentum to have
board, which met in Stockholm in April There are already some examples much impact on fragmented European
and will now draw up a 5 year strate- of the kinds of collaboration that might research. Critics charge that the overall
gic plan for neurodegenerative disease proceed under the Joint Programme. On approach is ill-defined, inadequately pro-
research in Europe. The board is made June 29th, for example, the UK MRC, moted, and—most of all—underfinanced.
up of five social scientists, five clini- the German Centre for Neurodegenera- Physicist and European Research Advi-
cians, and five biologists: Jesus Avila tive Diseases (DZNE), and the Canadian sory Board member Jerzy Langer of the
of the University of Madrid, Bart De Institutes of Health Research (CIHR) Polish Academy of Sciences says he
Strooper, John Hardy of University Col- announced a £3 million ($4.6 million) col- fears that it won’t get much further than
lege, London, Leszek Kaczmarek of the laboration on methods, technologies, a long line of prior Commission efforts to
Polish Academy of Sciences and the and data sharing in neurodegenerative get national research agencies to work
chairman, Thomas Gasser of the Uni- disease research. (Canada’s involvement more closely together. “I’m not saying
versity of Tubingen. There is no budget reflects the MRC’s desire to cooperate it is wrong,” he says. “But any initiative
yet to implement the plan, but officials with partners outside Europe who have that starts from the bureaucracy tends to
involved in the discussions say that it is relevant expertise.) And Amouyel says go nowhere.” Langer contends that joint
likely to involve an investment of about that some test projects may go ahead programming still has little constituency
€200 million over 5 years, mainly from under the Joint Programme before the among researchers. “The key question,”
national funding bodies. “For the very strategic plan’s completion, in areas such he says, “is when, and in what capacity,
first time,” says Amouyel, “we’ll have as genomics, the standardization of bio- real researchers get involved.”

348  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


“There is serious added value to be together at the ministerial level to iden- Amouyel, “because you have to allocate
achieved if we can use joint program- tify jointly areas where public research resources more efficiently.” And in fields
ming to get national funding used more can contribute to tackling Europe’s such as neurodegenerative disease,
effectively,” says Ian Halliday, president major societal problems,” said Maire where even the largest national research
of the European Science Foundation in Geoghegan-Quinn, the newly appointed agencies cannot cover every aspect, the
Strasbourg. “But politically and organi- European Commissioner for research, joint programming approach may help.
zationally, this is not going to be easy.” in a statement. “It is precisely because Yet the odds remain stacked against
And observers of the joint programming it is underpinned by a high-level, strate- it making much difference: EU member
pilot say that the MRC and INSERM are gic and structured process—and most states control five-sixths of Europe’s
already frustrated by the need to accom- importantly of all, by real political will— research budget, and they want to spend
modate participation by the 24 EU nations that joint programming is a very big step it on their own scientists. A 2009 study
who have signed up for the pilot—many forward.” by Eurohorcs, the federation of national
of whom cannot contribute much in the The Commission’s approach to the research councils, found that 14 out of
way of cutting-edge neurobiology. Some idea is finely nuanced, however, because 32 of the national agencies surveyed had
suggest that participants will have to if joint programming is seen as a Com- legal prohibitions on supporting work
learn to do what physicists have done at mission project, member states will outside their borders (http://www.era.
CERN, the successful European particle view it as a means of getting Commis- gv.at/attach/EUROHORCs.pdf). At the
physics center in Switzerland, and find sion money. So Commission officials are larger agencies, around 5% of the budget
ways of accommodating partners who orchestrating it from behind the scenes, is usually devoted to collaborations with
are less scientifically advanced. hoping that national governments will European partners (some small agen-
Buckle denies that this is a problem take the actions needed to provide cies, such as the Foundation for Polish
for the MRC, arguing that the Joint Pro- money and drive it forward. But there are Science and the Greek National Hellenic
gramme for Neurodegenerative Disease hints that large-scale Commission fund- Research Foundation, spend proportion-
will be an “umbrella” for the sharing of ing to support joint programming could ally more). The perceived political draw-
information and will allow “different con- become available under the next phase backs of spending dwindling research
stellations” of nations to work together of the Framework Programme, FP8, funds “abroad” remain daunting, however.
on different areas of interest. “Everyone which starts in 2014. And there is a strong imperative within
involved will be signed up to the same Governments are certainly watching national research agencies to hold on to
top-level objectives,” he says, “but the pilot with interest. In April, the Com- control of what they have. “There’s always
everyone will have different interpreta- mission recommended three further going to be people at national agencies
tions of how to best achieve them.” pilots—in food security, healthy diets, who fear that they could lose out” from
Backers of joint programming say it and cultural heritage conservation. In joint programming, agrees Connolly. “But
can work because it is the EU member tough economic times, at least in theory, there’s very strong commitment at the
states, through the Council of Ministers, joint programming could help to reduce political level, and in the European Com-
who have endorsed the approach and duplication enabling research funds to mission, to making this happen. Member
have pledged to see it through. “For the be spent more efficiently. “In a way, a states are coming to see that they can’t
first time, member states are coming shrinking budget is an opportunity,” says do everything on their own.”

Colin Macilwain
Edinburgh, UK
DOI 10.1016/j.cell.2010.07.031

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  349


Leading Edge

Correspondence

Successes of Genome-wide The fact that most SNPs identified by


GWAS do not lie in coding regions or other
known regulatory elements is expected
Association Studies from the study design and is not evidence
of false positives. The assumption under-
lying the design of GWAS and choice of
In a recent Essay in Cell, McClellan and such instances, for example, the TCF2 genotyped SNPs is that the true functional
King argue that genomic resequencing (or HNF1B) gene where alternate alleles allele will be nearby and correlated with
rather than genome-wide association are risk factors for type 2 diabetes and the initial SNP through linkage disequi-
studies (GWAS) will be necessary to prostate cancer (Gudmundsson et al., librium. When one considers linkage dis-
understand the genetic basis of common 2007). More generally, several loci where equilibrium, there is an observed excess of
disease (McClellan and King, 2010). Like alleles have opposite effects on the risk of GWAS hits that influence promoter regions
the authors, we too are excited about the developing type 1 diabetes and Crohn’s or change the protein-coding sequence
potential for emerging sequencing tech- disease have been reported (Wang et al., of a gene and a relative paucity of hits in
nologies to facilitate discoveries that 2010), and it is likely that more examples intergenic regions (Hindorff et al., 2009).
explain the missing heritability of com- of balancing selection are yet to be dis- Moreover, many disease-associated SNPs
mon diseases. However, we disagree covered. identified by GWAS are located in genes or
with the implication that GWAS have not The argument that common pathogenic pathways previously known or suspected
been successful to date. Instead, we variants must have withstood selective to play a role in disease etiology. Recent
propose that insofar as the goal of these pressure throughout human history is GWAS for Alzheimer’s disease, Crohn’s
studies is to understand the etiology of predicated on the assumption that mod- disease, type 1 diabetes, and type 2 dia-
heritable diseases, GWAS have provided ern humans developed in the same envi- betes have rediscovered SNP associations
numerous tantalizing clues for us biolo- ronment that we exist in today. However, previously reported from candidate gene
gists to decipher. Rather than disprove due to the rapid acceleration of human studies. The demonstration that GWAS
the common disease/common variant development in the recent evolution- can identify common disease suscepti-
hypothesis, we find that results from ary timeframe, numerous environmental bility variants provides a positive control
GWAS support the contention that com- changes have occurred that may impact (Hindorff et al., 2009). More generally, the
mon polymorphisms do directly contrib- the risk of complex common diseases. functional pathways of GWAS-identified
ute to disease risk, validating the linkage For instance, variants that were ben- genes often make sense; for instance,
disequilibrium-based GWAS approach eficial in the past may well have turned numerous inflammatory genes have been
for helping to identify variants underlying against their carriers as human lifestyles implicated by GWAS in inflammatory bowel
disease. Although we do not dismiss the changed. The “thrifty gene hypothesis” disease (Hindorff et al., 2009). We have
likelihood that rare variants also contrib- suggests that variants that predispose found a similar concordance between the
ute to common diseases, we expect that to type 2 diabetes and obesity may have literature and GWAS hits in our own work.
whole-genome sequencing approaches conferred a selective advantage in times In a GWAS for age-related macular degen-
will show that the full spectrum of alleles, of famine (Neel, 1962). However, in devel- eration, an SNP in complement factor H
from rare to common, play important oped countries, where food tends to be strongly associates with disease risk; this
roles in disease etiology. Here, we argue in overabundance, type 2 diabetes and is consistent with previous suggestions
that the existence of common disease- obesity have become common diseases. that the complement pathway plays a role
causing polymorphisms is not inconsis- Furthermore, existing neutral variation in disease etiology (Klein et al., 2005). Sim-
tent with population genetic theory and may manifest positive or negative effects ilarly, using GWAS, we identified a germ-
that actual results from GWAS suggest as new environmental modifiers come into line variant in the intron of the JAK2 gene
that the reported associations represent play. A set of single-nucleotide polymor- that is associated with myeloproliferative
real biology rather than false positives. phisms (SNPs) associated with lung can- neoplasms; JAK2 is known to harbor acti-
The contention that deleterious alleles cer and located at a locus encoding the vating oncogenic somatic mutations in this
that cause human diseases are common nicotinic acetylcholine receptor appears disease (Kilpivaara et al., 2009). One would
in the population may seem paradoxical, to have a stronger effect on lung cancer not expect such correlations between
but several mechanisms can explain how risk in smokers born long ago relative to GWAS findings, genic regions, and known
such pathogenic alleles can overcome those born more recently (Landi et al., disease biology if these findings were ran-
negative selective pressure. First, accu- 2009); this effect has been attributed to domly distributed false positives due to
mulating evidence demonstrates that changes in the composition of cigarettes population stratification or other causes.
there is balancing selection in which a over time. This demonstrates that recent As these associations are likely to be real,
certain allele confers susceptibility to one environmental changes can alter the the most logical and parsimonious expla-
disease while simultaneously conferring disease-influencing effect of a common nation is that, in general, GWAS success-
protection from another. The best known variant. These examples are likely to be fully identify disease-associated variants
example is heterozygosity for sickle cell only the tip of the iceberg of phenotypic and that variants found through GWAS tag
anemia, which affords protection against effects modulated by gene-environment regions important for the biology of these
malaria. GWAS have identified other interactions. diseases.

350  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


In light of this, it is likely that GWAS hits ined to see such a correlation. Although References
found in intergenic regions far from known further work is necessary to uncover the
genes are true associations whose biol- elusive mechanism by which the SNP Gaulton, K.J., Nammo, T., Pasquali, L., Simon,
J.M., Giresi, P.G., Fogarty, M.P., Panhuis, T.M.,
ogy is not yet understood, rather than confers risk, we propose that the existing
Mieczkowski, P., Secchi, A., Bosco, D., et al.
false positives. The human genome is evidence supports rather than refutes this (2010). Nat. Genet. 42, 255–259.
incompletely annotated. Regions where SNP as a true cancer risk allele. Another
GWAS associations have been found, but example is a non-protein-coding region Gudmundsson, J., Sulem, P., Steinthorsdottir, V.,
Bergthorsson, J.T., Thorleifsson, G., Manolescu,
no known genes are located, could easily of chromosome 9q21 in which SNPs
A., Rafnar, T., Gudbjartsson, D., Agnarsson,
harbor unidentified new genes or regula- have been robustly associated with arte- B.A., Baker, A., et al. (2007). Nat. Genet. 39,
tory elements. For instance, the authors rial disease. A recent paper reported that 977–983.
point to the colon and prostate cancer risk targeted deletion of an orthologous region
in mouse interferes with cis-regulation of Hindorff, L.A., Sethupathy, P., Junkins, H.A., Ra-
SNP rs693267 located 335 kb upstream
mos, E.M., Mehta, J.P., Collins, F.S., and Mano-
from the MYC gene on chromosome 8q24 nearby genes (Cdkn2a/Cdkn2b) and may lio, T.A. (2009). Proc. Natl. Acad. Sci. USA 106,
(McClellan and King, 2010). This locus has influence vascular cell proliferation (Visel 9362–9367.
been shown to physically interact with et al., 2010). As a third example, an intronic
MYC and is associated with enhanced type 2 diabetes risk SNP (rs7903146) was Kilpivaara, O., Mukherjee, S., Schram, A.M.,
Wadleigh, M., Mullally, A., Ebert, B.L., Bass, A.,
Wnt signaling. Therefore, although the biol- recently found to overlap with a region of Marubayashi, S., Heguy, A., Garcia-Manero, G.,
ogy of this locus is not fully understood, islet cell-selective chromatin, and the two et al. (2009). Nat. Genet. 41, 455–459.
it suggests a paradigm where intergenic alleles of rs7903146 correlate with the
disease-associated SNPs alter enhancer open/closed chromatin state of the region Klein, R.J., Zeiss, C., Chew, E.Y., Tsai, J.Y.,
Sackler, R.S., Haynes, C., Henning, A.K., San-
elements, either directly or through linkage (Gaulton et al., 2010). Thus, understanding
giovanni, J.P., Mane, S.M., Mayne, S.T., et al.
disequilibrium, and therefore cause differ- the mechanisms by which GWAS loci con- (2005). Science 308, 385–389.
ential regulation of disease-related genes. tribute to disease will require considerable
This observation leads to a broader effort and time. We take this not as a sign Landi, M.T., Chatterjee, N., Yu, K., Goldin, L.R.,
Goldstein, A.M., Rotunno, M., Mirabello, L., Ja-
point: A lack of biological understanding that the common disease-common vari-
cobs, K., Wheeler, W., Yeager, M., et al. (2009).
of how these disease-associated vari- ant model has failed but rather that a chal- Am. J. Hum. Genet. 85, 679–691.
ants are pathogenic does not mean that lenge exists for the scientific community—
there is no biology to discover. Although a challenge that must be addressed with McClellan, J., and King, M.C. (2010). Cell 141,
210–217.
our understanding of the mechanisms both traditional experimental genetics and
by which disease risk loci contribute to innovative new approaches. Neel, J.V. (1962). Am. J. Hum. Genet. 14,
pathogenesis currently lags behind the 353–362.
pace at which new loci are discovered,
Robert J. Klein,1,* Xing Xu,1 Semanti Visel, A., Zhu, Y., May, D., Afzal, V., Gong, E.,
promising stories continue to emerge. To Attanasio, C., Blow, M.J., Cohen, J.C., Rubin,
Mukherjee,1 Jason Willis,1 and James
continue the previous example, although E.M., and Pennacchio, L.A. (2010). Nature 464,
Hayes1
no definitive correlation between the 409–412.
1
Program in Cancer Biology and Genetics,
rs6983267 genotype and MYC expression Memorial Sloan-Kettering Cancer Center, Wang, K., Baldassano, R., Zhang, H., Qu, H.Q.,
has been demonstrated, MYC is known to New York, NY 10065, USA Imielinski, M., Kugathasan, S., Annese, V., Du-
be tightly regulated and the right develop- *Correspondence: kleinr@mskcc.org binsky, M., Rotter, J.I., Russell, R.K., et al.
mental time point may need to be exam- DOI 10.1016/j.cell.2010.07.026 (2010). Hum. Mol. Genet. 19, 2059–2067.

Strategies for Genetic tions of the Essay may lead to misin-


terpretation of published studies by us
and others. For the broad readership of
Studies of Complex Diseases Cell and for the scientific community in
general, we highlight our concerns in
this Correspondence.
In a recent Essay published in Cell, sequencing techniques to discover The authors refer to the fact that most
McClellan and King discussed genetic disease genes, are highly relevant to single-nucleotide polymorphisms (SNPs)
heterogeneity and the potential role of genetics researchers. However, the detected in GWAS reside in intergenic
rare genetic variants in complex human authors allocated a substantial propor- regions and consequently challenge the
diseases (McClellan and King, 2010). tion of their efforts to being critical of utility and reliability of GWAS with the
These important issues, in particu- the utility of genome-wide association question: “How did genome-wide asso-
lar the application of high-throughput studies (GWAS). These particular sec- ciation studies come to be populated by

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  351


risk variants with no known function?” the beauty of whole-genome SNP data samples from Orkney (allele frequency =
When addressed in the proper context, is that inflation of test statistics due to 0.21). When dealing with whole-genome
however, it is well established that GWAS population substructure can be read- data, small sample sizes often lead to
do not attempt to identify functional ily identified and adjusted. Populations biased estimates of allele frequencies,
SNPs but rather “tag” the approximate do not differ by just one or two SNPs; which we have now proven to be the
location of disease variants, typically instead, they differ at many loci such that case in this instance. We queried the
down to 100 kb or less. This is made whole-genome data aid in identifying 101 Tuscan samples included in the
possible due to the linkage disequilib- stratification, including extremely fine­ HapMap 3 project (http://www.sanger.
rium (LD) patterns characterized by the scale subpopulations among Europeans ac.uk/humgen/hapmap3) and found
International HapMap project, that is, the (Novembre et al., 2008). The GWAS com- that the allele frequency is 0.41, which
correlation of genotypes between the munity has established methods to deal is very similar to what we reported for
yet-to-be-determined underlying disease with population stratification, and these our European American cohort (0.39
variant and neighboring SNPs. Indeed, methods effectively adjust for common in Wang et al., 2009) and substantially
the vast majority of SNPs used in GWAS variants without controversy. There are different from 0.77 quoted by McClel-
are of unknown biological function, due certainly challenges with the analyses of lan and King. Furthermore, even if the
to the fact that most SNPs reside outside rare variants, hypervariable variants, or allele frequency measures are accurate,
of coding regions and that the manufac- interrogation of recently admixed popu- McClellan and King should not claim this
turers of the SNP arrays selected SNPs lations, which are all active topics of cur- SNP as “hypervariable” without appro-
from the HapMap to facilitate efficient rent research. There are now standard priate control experiments to compare
tagging across the genome, that is, the practices to handle population stratifica- it with other SNPs. They may not appre-
priority was information capture rather tion in GWAS, such as genomic control, ciate that the vast majority of SNPs do
than putative function. Furthermore, EigenStrat, and multidimensional scal- have variable allele frequencies between
noncoding SNPs identified by GWAS ing. Furthermore, family-based study populations; in fact, 44% of SNPs on the
may reveal intergenic regulatory ele- designs have the advantage of pro- Illumina array (http://hgdp.uchicago.edu/
ments that are critical to understand, tecting against stratification. Thus, the Browser_tracks/FST) have more extreme
and it is now up to the genetics com- GWAS community has developed ways population divergence (measured by
munity to develop approaches to interro- to address population stratification, and Fst values) than rs4307059. Similarly,
gate the function of regulatory variants. odds ratios of 1.5, 1.2, or even 1.1 have another SNP, which has been replicated
The authors did refer to LD as a potential proven to be bona fide signals rather than for its association with type 1 diabetes
explanation for noncoding variants yield- artifacts of population stratification. (Hakonarson et al., 2007) in over 20 inde-
ing association in GWAS, but they failed The Essay’s authors go on to claim pendent studies, is ranked in the 29th
to recognize that the design of GWAS is that the autism locus reported by our percentile for Fst value. As such, it is our
not to directly interrogate causal variants group (Wang et al., 2009) is a “false posi- interpretation that the authors took a ran-
in the first instance. For example, the two tive” due to population stratification, but dom SNP from the middle of a ranked list
GWAS that we have conducted for sickle the study was driven by family-based and inappropriately claimed it as a “par-
cell anemia and hearing loss (Dickson et cohorts both at the discovery and repli- ticularly dramatic” example of “popula-
al., 2010) yielded top hits in intergenic cation stages, with case-control cohorts tion stratification.” Extending this further,
regions, but these are in close proximity further supporting the finding. Here, we also wish to point out that replication
to the causal genes (HBB and GJB2) that McClellan and King ignore the fact that of GWAS signals depends on many fac-
were already well-established before the family-based analysis is robust against tors, including the power of the study,
GWAS era. These studies demonstrate stratification and have mistakenly con- the sample ascertainment scheme or
the reliability of GWAS for identifying the sidered GWAS hits as “false positives” diagnostic criteria, and the heterogene-
approximate locations of disease genes if allele frequencies varied across Euro- ity of the study population (for example,
by noncoding SNPs. In addition, they pean or HapMap populations. For exam- sporadic versus familial cases). For neu-
represent two vivid examples of how ple, they claim that our reported autism- ropsychiatric diseases, it is well known
GWAS can work by leveraging LD. associated SNP, rs4307059 (Wang et that replication can be difficult with small
McClellan and King have also attrib- al., 2009), was a “particularly dramatic sample sizes, as is the case for the study
uted many published GWAS hits to example of the perils of cryptic popula- specifically referred to by the authors
population stratification. In the absence tion stratification.” Their reasoning is that (Weiss et al., 2009), which interrogated
of scientific support or statistical deriva- the frequency of the proposed risk variant 258 families with autism for replica-
tion, they claim that “an odds ratio of 3.0, varies from 0.21 to 0.77 across European tion that were not part of AGRE (Autism
or even of 2.0 depending on population populations and that it is monomorphic in Genetic Resource Exchange). Never-
allele frequencies” would be robustly African populations. However, they used theless, the autism locus on 5p14.1 has
interrogated by GWAS. The vast major- data from the Human Genome Diver- been replicated in a family-based GWAS
ity of published GWAS loci therefore sity Project that examined 938 samples without case-control comparisons (Ma et
fall below the threshold for “popula- from 51 worldwide populations (Coop al., 2009) and has been associated with
tion stratification.” However, compared et al., 2009), including 7 samples from communication-spectrum phenotypes
to candidate gene association studies, Tuscany (allele frequency = 0.77) and 15 through quantitative trait associations

352  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


in 7313 children from a UK birth cohort, Kai Wang,1 Maja Bucan,2 Struan F.A. (2007). Nature 448, 591–594.
eliminating concerns of population strati- Grant,1,3 Gerard Schellenberg,4 and Ma, D., Salyakina, D., Jaworski, J.M., Konidari, I.,
fication (St Pourcain et al., 2010). Hakon Hakonarson1,3,* Whitehead, P.L., Andersen, A.N., Hoffman, J.D.,
Like McClellan and King, we expect
1
Center for Applied Genomics, Children’s Slifer, S.H., Hedges, D.J., Cukier, H.N., et al. (2009).
Hospital of Philadelphia Ann. Hum. Genet. 73, 263–273.
that with the development of high- 2
Department of Genetics
throughput sequencing technologies, McClellan, J., and King, M.C. (2010). Cell 141,
3
Department of Pediatrics 210–217.
whole-genome sequencing will be an 4
Department of Pathology and Laboratory
invaluable tool for the study of rare Medicine Novembre, J., Johnson, T., Bryc, K., Kutalik, Z.,
genetic variants contributing to com- University of Pennsylvania, Philadelphia, PA Boyko, A.R., Auton, A., Indap, A., King, K.S., Berg-
mann, S., Nelson, M.R., et al. (2008). Nature 456,
plex diseases. However, the apparent 19104, USA
98–101.
importance of rare variants does not *Correspondence: hakonarson@chop.edu
discount the contribution of common DOI 10.1016/j.cell.2010.07.025 St Pourcain, B., Wang, K., Glessner, J.T., Golding, J.,
Steer, C., Ring, S.M., Skuse, D.H., Grant, S.F., Hako-
variants; the concerns about population References narson, H., and Smith, G.D. (2010). Am. J. Psychia-
stratification should not be overstated try. Published online July 15, 2010. 10.1176/appi.
and certainly should not be presented Coop, G., Pickrell, J.K., Novembre, J., Kudaravalli, ajp.2010.09121789.

as an argument to discredit many pub- S., Li, J., Absher, D., Myers, R.M., Cavalli-Sforza,
L.L., Feldman, M.W., and Pritchard, J.K. (2009). Wang, K., Zhang, H., Ma, D., Bucan, M., Glessner,
lished GWAS signals with an odds ratio PLoS Genet. 5, e1000500. J.T., Abrahams, B.S., Salyakina, D., Imielinski, M.,
Bradfield, J.P., Sleiman, P.M., et al. (2009). Nature
of less than 2. These sections within 459, 528–533.
Dickson, S.P., Wang, K., Krantz, I., Hakonarson, H.,
an otherwise excellent Essay must be and Goldstein, D.B. (2010). PLoS Biol. 8, e1000294.
countered so that Cell readers have a Weiss, L.A., Arking, D.E., Gene Discovery Proj-
Hakonarson, H., Grant, S.F., Bradfield, J.P., March- ect of Johns Hopkins and the Autism Consortium,
more balanced view of the current state and, L., Kim, C.E., Glessner, J.T., Grabs, R., Casa- Daly, M.J., and Chakravarti, A. (2009). Nature 461,
of the field. lunovo, T., Taback, S.P., Frackelton, E.C., et al. 802–808.

Response variant influencing the trait in question


must truly exist in the population being

Why It Is Time to Sequence


studied; and (2) the genotyped markers
used in association analyses must either
include the causal variant or be in link-
In response to our Essay “Genetic Het- Thus, the best documented common age disequilibrium (LD) with the causal
erogeneity of Human Disease” (McClellan variants with a substantial impact on variant in the population being stud-
and King, 2010), Wang et al. and Klein et al. disease risk typically are associated with ied. Common “risk SNPs” detected by
challenge our critique of GWAS findings. illnesses presenting later in life, includ- GWAS could in theory be in LD with rare
Both Correspondence suggest that our ing Alzheimer’s disease (APOE), exfoliat- disease-causing alleles, if by chance
conclusions lack an understanding of the ing glaucoma (LOXL1), and age-related rare causal alleles are disproportionately
principles of the common variant-common macular degeneration (CFH). These linked with a common variant (Dickson et
disease model and its application in GWAS alleles persist in the population because al., 2010).
methodology. We respectfully disagree, the associated illnesses do not nega- In addressing our critique, both
and in fact our Essay directly addressed tively influence reproductive fitness. Wang et al. and Klein et al. acknowl-
many of their points. We are happy to clar- However, the existence of common edge that GWAS methodologies are
ify further. The issue is not ignorance of alleles that truly influence disease does designed to detect SNPs primarily
GWAS methodology; the issue is reconcil- not imply that all GWAS findings are found in intergenic or intronic regions,
ing GWAS findings with population genet- valid. To date, published GWAS have given the construction of standard SNP
ics, evolution, and biology. reported more than 500 single-nucle- arrays. They argue that the risk SNPs
There is no dispute that common otide polymorphisms (SNPs) associated implicated by GWAS are not expected
genetic variants influence human traits. with various diseases and traits, 88% of to be causal but instead are in LD with
Alleles with the strongest influences on which lie in introns or intergenic regions, true causal variants. Wang et al. main-
human traits are associated with benign and for which the median odds ratio for tain that standard GWAS methodolo-
phenotypes, such as hair color and effect size is 1.33 (Hindorff et al., 2009). gies adequately control for population
eye color, that vary across individuals. Few of these associations have biologi- stratification. Further, they note that
Alleles for these traits lie in coding and cal support. a large proportion of HapMap SNPs
known regulatory regions (Hindorff et In order to identify alleles that influence vary widely across populations, and
al., 2009). Such variants have been influ- disease using GWAS, two fundamen- therefore it is not surprising that highly
enced by selection in human evolution. tal criteria must be met: (1) a common variable SNPs emerge as risk vari-

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  353


ants. These issues are at the crux of characterize many “risk alleles” identi- such mutations or elements have been
the debate and will be the focus of our fied by GWAS, including, for example, found by following up on GWAS findings.
response. rs4307059 reported to be associated Wide variations in allele frequency across
Genetic heterogeneity across human with autism (Wang et al., 2009), which is populations argue against shared hap-
populations limits GWAS methodology a major focus of the Correspondence by lotype structure, which is necessary to
and may result in the identification of “risk Wang et al. We suggest that associations detect causal variants in LD.
alleles” that are neither likely to be func- based on such highly variable SNPs are Common variants influencing traits or
tional nor likely to be in LD with alleles that often artifacts of cryptic population strat- disease must withstand selection in every
are. The genetic architecture of human ification. Wang et al. argue that standard generation in order to be maintained at
disease is shaped by the same evolu- GWAS strategies have been adopted to polymorphic frequencies worldwide.
tionary forces that impact the human control for population stratification. How- Regulatory elements are impacted by
genome. Genetic architecture is dic- ever, these methods control by person, the same evolutionary forces as coding
tated by four factors: mutation, selection, not by SNP. Because populations from regions. Klein et al. discuss alternative
migration, and drift. Negative selection large geographic areas (e.g., Europe) are methods whereby otherwise deleterious
globally reduces variation at missense genetically heterogeneous, outlier SNPs alleles are maintained in the population,
mutations, particularly in genes associ- that vary widely among subgroups of citing both well-known and speculative
ated with disease (Barreiro et al., 2008). such populations are not excluded by examples of balancing selection and
Frequencies of missense alleles (i.e., these methods and often drive positive gene-environment interactions. How-
nonsynonymous SNPs) are less diverse associations. ever, as is the case for much of this dis-
across populations than are frequencies Wang et al. also assert that the asso- cussion, a confirmed biological mecha-
of synonymous SNPs, SNPs in untrans- ciation of rs4307059 and autism was nism for one illness does not mean that
lated regions (UTRs), and intronic SNPs driven by family-based cohorts, which all GWAS findings can be attributed to
because selection reduces interpopula- are robust to stratification. However, similar mechanisms.
tion variation for alleles influencing phe- no SNPs reached genome-wide signifi- Klein et al. suggest that a common
notypic expression (Garte, 2010). Popula- cance in their study using a family-based risk allele may have been under neu-
tion differences in genic regions, primarily design (Wang et al., 2009). Significance tral or positive selection in early human
at nonsynonymous SNPs and 5`UTR vari- was obtained by pooling subjects from history, thus explaining its worldwide
ants, may persist as the result of selec- both family-based and case-control prevalence and maintenance in the
tion for adaptive responses or by chance samples, including thousands of unre- population despite the association with
(Barreiro et al., 2008). For alleles influ- lated cases and controls. Analyses disease. Yet evolution is an ongoing
encing phenotypes, selection constrains only comparing affected to unaffected process. If a common variant decreases
overall variation in allele frequency while persons within families will be robust reproductive fitness during modern his-
also influencing specific patterns of varia- to stratification; but comparing a mixed tory, coinciding with the vast expansion
tion defined by geographic ancestry. series of related and unrelated cases to of the human population, the frequency
Many SNPs, inversions/deletions controls can exacerbate stratification of the variant will decline. Klein et al.
(indels), and short tandem repeats due to the clustering of shared ances- also suggest that many common risk
vary widely in allele frequency among tries in the affected group. variants, either the identified SNP or
populations. This is especially true for In arguing for rs4037059 as a risk vari- a causal variant in LD, operate by vir-
variants that are not in coding or regu- ant for autism, Wang et al. dismiss the tue of some yet-to-be-determined bal-
latory regions. Such alleles vary with negative results of a replication study ancing selection or gene-environment
population clusters in patterns more (Weiss et al., 2009) due to small sample interaction. This is obviously possible
consistent with neutral drift and migra- size. Yet, they cite another study (Ma et al., but needs to be demonstrated for any
tion rather than with selection (Coop et 2009) as supporting an association with given variant. In the presence of a true
al., 2009). The colonization of the world this genomic region (5p14.1), even though balancing selection or gene-environ-
by modern humans was carried out no SNPs in this study met genome-wide ment interaction, variation in allele fre-
by a series of founder populations with significance and rs4037059 was not even quencies will correlate with geographic
subsequent rapid expansion of popula- nominally significant in the discovery variation in disease prevalence and is
tion size. Neutral alleles emerging at the sample. Variable weak associations of likely to offer clues about specific envi-
forefront of these expansions “surfed” different SNPs across the same genomic ronmental exposures or risk factors.
waves of population growth. Variations region in different cohorts do not consti- The best characterized examples of
in allele frequencies across populations tute replication. balancing selection are found in spe-
stem from differences in the timing of the Both Klein et al. and Wang et al. suggest cific geographic populations sharing
variant’s emergence in the expansion. that the vast majority of GWAS risk alleles an environmental exposure, such as an
Intergenic or intronic SNPs that vary are in LD with causal mutations, and that infectious disease. In these examples,
widely in frequency among populations intergenic and intronic risk variants rep- e.g., sickle cell disease, risk alleles are
are most likely neutral alleles reflect- resent regulatory elements. In principle, maintained at a much higher rate in the
ing the history of human migration. either or both of these hypotheses could exposed population than in populations
Wide variations in allele frequency also be true. However, thus far, virtually no from other parts of the world.

354  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


It is a leap of theoretical faith to infer that highly significant p values and very small References
SNPs with no known function and variable effects that diminish with further study.
weak associations across different disease Currently, GWAS results fail to explain Barreiro, L.B., Laval, G., Quach, H., Patin, E.,
and Quintana-Murci, L. (2008). Nat. Genet. 40,
cohorts represent prima facie evidence of the vast majority of genetic influence 340–345.
regulatory function, gene-environment on any human illness. Further, most risk
Coop, G., Pickrell, J.K., Novembre, J., Kudaravalli,
interaction, or balanced selection. We are variants implicated by GWAS have no S., Li, J., Absher, D., Myers, R.M., Cavalli-Sforza,
not suggesting that such phenomena do demonstrated biological, functional, or L.L., Feldman, M.W., and Pritchard, J.K. (2009).
not occur. Deciphering such mechanisms clinical relevance for disease. PLoS Genet. 5, e1000500.
will be a major scientific focus over the next We suggest that biological relevance Dickson, S.P., Wang, K., Krantz, I., Hakonar-
decade. However, for the vast majority of needs to be established before assert- son, H., and Goldstein, D.B. (2010). PLoS Biol. 8,
SNPs identified by GWAS, these mecha- ing that positive correlations from GWAS e1000294.
nisms are speculative in the absence of are equivalent to disease risk. Such evi- Garte, S. (2010). Am. J. Hum. Biol. 22, 297–300.
biological evidence. dence must address the functional sig-
Hindorff, L.A., Sethupathy, P., Junkins, H.A., Ra-
Many of the examples cited by Klein et nificance of the SNP, or a variant in LD mos, E.M., Mehta, J.P., Collins, F.S., and Mano-
al. and Wang et al. in support of GWAS with the SNP, rather than arguing for the lio, T.A. (2009). Proc. Natl. Acad. Sci. USA 106,
suffer from the same limitations that we appeal of a nearby gene. In their argu- 9362–9367.
highlighted in our Essay (McClellan and ments, Wang et al. and Klein et al. simply Ma, D., Salyakina, D., Jaworski, J.M., Konidari, I.,
King, 2010). For example, as Klein et al. restate GWAS principles, which serves Whitehead, P.L., Andersen, A.N., Hoffman, J.D.,
Slifer, S.H., Hedges, D.J., Cukier, H.N., et al. (2009).
describe, there is robust epidemiologi- to reify, not prove, the model. We make a
Ann. Hum. Genet. 73, 263–273.
cal data in support of individuals with plea for more rigorous analysis. Science
diabetes having a lower risk for prostate ultimately advances by evidence. McClellan, J., and King, M.C. (2010). Cell 141,
210–217.
cancer. However, associations between
putative risk/protective SNPs in HNF1B Jon McClellan1 and Stevens, V.L., Ahn, J., Sun, J., Jacobs, E.J., Moore,
and JAZF1 and the two illnesses vary S.C., Patel, A.V., Berndt, S.I., Albanes, D., and
Mary Claire King2,*
Hayes, R.B. (2010). Prostate 70, 601–607.
across cohorts, and these variants do 1
Department of Psychiatry
not mediate the relationship between the
2
Department of Medicine and Department Wang, K., Zhang, H., Ma, D., Bucan, M., Glessner,
two diseases (Stevens et al., 2010). of Genome Sciences J.T., Abrahams, B.S., Salyakina, D., Imielinski, M.,
University of Washington, Seattle, WA Bradfield, J.P., Sleiman, P.M., et al. (2009). Nature
We understand that many believe that 459, 528–533.
98195, USA
most GWAS findings are valid. Ultimately, *Correspondence: Weiss, L.A., Arking, D.E., Gene Discovery Project of
the debate hinges on how definitive one mcking@u.washington.edu Johns Hopkins and the Autism Consortium, Daly,
views variable results with statistically DOI 10.1016/j.cell.2010.07.027 M.J., Chakravarti, A. (2009). Nature 461, 802–808.

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  355


Leading Edge

Previews

Fyn-Tau-Amyloid: A Toxic Triad


Christian Haass1,2,* and Eckhard Mandelkow3,4,*
1
German Center for Neurodegenerative Diseases (DZNE), 80336 Munich, Germany
2
Adolf Butenandt Institute, Ludwig Maximilian University, 80336 Munich, Germany
3
Max Planck Unit for Structural Molecular Biology, 22607 Hamburg, Germany
4
German Center for Neurodegenerative Diseases (DZNE), 23175 Bonn, Germany
*Correspondence: chaass@med.uni-muenchen.de (C.H.), mand@mpasmb.desy.de (E.M.)
DOI 10.1016/j.cell.2010.07.032

The axonal protein tau and amyloid β-peptide (Aβ) are key players in the pathogenesis of Alzheim-
er's disease, and tau mediates Aβ toxicity, but it is not clear how. Ittner et al. (2010) now report an
unexpected physiological function for tau in neuronal dendrites that may explain how tau mediates
Aβ toxicity.

Alzheimer’s disease (AD) is a devastat- et al., 2007). In this issue of Cell, Ittner the NMDA receptor, resulting in stabili-
ing public health problem for our aging et al. (2010) now shed light on an unex- zation of this receptor’s interaction with
societies. Although it is well established pected dendritic function for normal tau PSD95, a scaffolding protein in the den-
that amyloid β-peptide (Aβ) forms toxic that suggests how tau may mediate Aβ dritic spines of neurons. This stabiliza-
oligomers in the brain (Haass and Sel- toxicity. tion, in turn, strengthens signaling by the
koe, 2007), it is not clear how Aβ initiates Most research on tau function has excitotoxic neurotransmitter glutamate,
the amyloid cascade and causes the focused on whether it is important for which enhances Aβ toxicity (Figure 1). A
death of neurons. Tau, an axonal protein, microtubule stabilization, neurite out- tau-dependent increase in Fyn in den-
seems to be an executor of Aβ toxicity growth, or the formation of tracks for dritic spines could boost excitotoxic sig-
even though it is localized to axons, and cargo transport along axons. The cata- naling, and, conversely, sequestration
Aβ toxicity is primarily triggered through strophic redistribution of tau to the soma of Fyn by tau or downregulation of tau
interactions of Aβ oligomers with den- and dendrites of neurons in AD sug- expression could mitigate excitotoxicity.
dritic spines (Haass and Selkoe, 2007). gests that an efficient sorting mechanism To address whether this is the case, Itt-
Tau binds to microtubules, a process that normally keeps tau localized to axons. ner and coworkers first generated trans-
is prevented by its abnormal phosphory- However, such “polarized” sorting like genic mice that overexpressed a variant of
lation during AD pathogenesis. Loss of other cellular sorting pathways is never tau (∆tau) that lacked the C terminus and
microtubule binding by tau is thought to completely efficient, thus enabling small thus could bind to Fyn but not to micro-
cause the disassembly of microtubules amounts of tau to become localized to tubules. Interestingly, ∆tau was completely
followed by the aggregation and depo- the somatodendritic compartment even excluded from dendrites and accumulated
sition of tau in pathogenic neurofibril- under physiological conditions. This within the soma (Figure 1). Moreover, ∆tau
lary tangles. Although amyloid plaques led Ittner and colleagues to propose a efficiently competed with endogenous tau
and tau tangles are prominent markers microtubule-independent physiological for binding to Fyn, resulting in sequestra-
of AD, one of the first and most obvious function for tau that regulates signaling in tion of Fyn in the soma. Similarly, loss of
pathological abnormalities observed dendritic spines. Their research was ini- tau also prevented postsynaptic targeting
in brain tissue from AD patients is the tially triggered by accumulating evidence of Fyn, and loss of Fyn in the dendrites
relocalization of tau from axons to the that subacute seizures occur not only in was enhanced when ∆tau was expressed
somatodendritic compartment of neu- transgenic mouse models of AD, but also in mice deficient in normal tau. This sug-
rons (Figure 1) (Ballatore et al., 2007). in AD patients (Palop and Mucke, 2009). gests that the targeting of Fyn to dendrites
During brain development, tau and other Interestingly, tau deficiency decreases depends on normal tau, a surprising find-
microtubule-associated proteins are ini- seizures induced by the Aβ-mediated ing for a protein believed to act only in
tially distributed ubiquitously throughout overstimulation of excitatory N-methyl-D- axons. But how tau-based sorting of Fyn
neurons, but then, as differentiation pro- aspartate (NMDA) receptors and improves occurs and whether other proteins are
gresses, tau becomes sorted into axons survival in a transgenic AD mouse model required remain unclear.
(Figure 1). In AD and other neurodegen- (Roberson et al., 2007). Fyn phosphorylates NMDA receptor
erative diseases involving tau (termed It is well established that tau binds not subunit 2 at tyrosine 1472 and stabilizes
“tauopathies”), this neat sorting pattern only to microtubules but also to several the interaction of this subunit with the
breaks down (Ballatore et al., 2007), nonreceptor tyrosine kinases including postsynaptic protein PSD95 (Nakazawa et
perhaps because abnormal phospho- Fyn through its N-terminal domain (Lee al., 2001). However, when Fyn is trapped
rylation of tau enables it to detach from et al., 1998). This enables tau to seques- within the soma by expression of ∆tau or
microtubules and diffuse rapidly into ter Fyn and to alter its localization in the reduced expression of endogenous tau,
other neuronal compartments (Konzack neuron. Fyn phosphorylates subunit 2 of there is decreased phosphorylation of

356  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


the NMDA receptor, the interaction of the
receptor with PSD95 is destabilized, and
excitotoxic signaling decreases. Excito-
toxicity has been implicated in Aß-medi-
ated toxicity, and a reduction in tau ame-
liorates the pathological action of Aβ in
the AD transgenic mouse model (Rober-
son et al., 2007). Similarly, overexpression
of ∆tau in the AD mice prevented seizures
and improved survival; indeed, these ben-
efits were enhanced when expression of
endogenous tau was reduced. Moreover,
the AD mice overexpressing ∆tau showed
improvements in memory, suggesting
that Aβ toxicity was reduced when Fyn
levels decreased in the dendritic spines.
This was independent of Aβ production or
the overall amyloid plaque load.
Notably, there is an interesting twist
to the Fyn-tau-microtubule story. In oli-
godendrocytes, tau is necessary for Figure 1. A Toxic Triad in Alzheimer’s Disease.
the outgrowth of cell processes and for Shown is the neuronal localization of tau protein (red) and Fyn kinase (blue) under physiological and
transport of Fyn, which is important for pathological conditions.
(Left) Normal tau is primarily located in axons but also interacts with Fyn kinase and targets it to den-
myelinating neurons. Expression of the drites. Fyn kinase phosphorylates subunit 2 of the NMDA receptor in dendritic spines, which results
N-terminal domain of tau alone causes in stabilization of the receptor’s interaction with the postsynaptic density protein PSD95, leading to
abnormal sorting of Fyn, resulting in poor enhanced excitotoxicity. Excitotoxicity is known to increase the toxic effects of oligomers of Aβ (dark
purple) on neurons.
myelination of neurons and seizures in (Middle) Overexpression of a ∆tau variant lacking the C terminus, which binds to Fyn kinase but not to
rodents (Klein et al., 2002). Thus, there are microtubules, results in the sequestration of Fyn in the soma, preventing Fyn from reaching the dendrites.
two completely distinct settings for this Consequently, Fyn-mediated phosphorylation of NMDA receptors is decreased and Aβ-mediated toxicity
is reduced (pale purple).
potentially toxic triad, which implicates (Right) Enhanced redistribution of abnormally hyperphosphorylated tau from axons to the somatoden-
abnormal tau in seizure disorders as well dritic compartment during AD pathogenesis may increase tau-dependent sorting of Fyn to the dendrites,
as in neurodegenerative diseases. boosting excitotoxic signaling and increasing the toxic effects of Aβ (black) on neurons.
But can the Fyn-tau connection in den-
dritic spines be exploited to develop new earliest stages of AD? This may be the ates the transport of Fyn to dendrites. If
therapeutic strategies for treating AD? A case, as redistribution of full-length tau that transport mechanism is disturbed,
peptide that blocks phosphorylation of (and possibly tau fragments) to the som- what other neuronal functions would be
NMDA receptors by Fyn protects neu- atodendritic compartment occurs well affected? Clearly, not only Fyn, but also
rons from excitotoxic damage (Aarts et before the formation of neurofibrillary other signaling molecules containing
al., 2002). Strikingly, when Ittner and col- tangles. The abnormal sorting of tau to SH3 domains bind to the PXXP motifs
leagues treated their transgenic AD mice the somatodendritic compartment may in the N-terminal domain of tau and of
with this peptide, memory deficits were be due to changes in signaling cascades related microtubule-associated proteins;
ameliorated and there was improved that alter kinase and phosphatase activi- but we do not know whether inhibition
survival, similar to the results when Fyn ties in those dendritic spines affected by of this binding would have deleterious
was sequestered by ∆tau. Aβ, resulting in local changes in tau sort- effects. Also, is ∆tau just a scavenger
The Ittner et al. study may provide ing and cytoskeletal rearrangements. that when overexpressed sequesters
a missing link between extracellular But do tau and Fyn still interact after tau Fyn? Could proteins other than tau per-
deposits of Aβ and intracellular tau and redistribution, given that the Fyn bind- form this activity in vivo? The enhanced
pinpoints tau and Fyn as possible medi- ing site on tau can be phosphorylated phenotypes upon reduction of endoge-
ators of Aβ toxicity. Although the idea is leading to disruption of this binding? If nous tau seem to speak against this pos-
tantalizing, the actual colocalization of they do, then relocalized tau may boost sibility. The provocative work of Ittner et
tau and Fyn within dendritic spines and Fyn action in dendrites and hence phos- al. raises new challenging questions, but
their modes of action remain to be shown phorylation of NMDA receptors, thus also brings us a significant step closer
under physiological conditions. A major enhancing excitotoxic signaling and to understanding AD pathophysiology.
surprise of this study is that a normal increasing the sensitivity of neurons to Hopefully, these findings will help in the
physiological function of tau, regarded Aβ (Figure 1). However, before targeting design of new therapeutic strategies for
as an axonal protein, mediates Aβ tox- the tau-Fyn interaction for therapeutic reducing the synaptic dysfunction and
icity at dendritic spines. Is this physio- purposes, we need to know more about neuronal loss of AD and other neurode-
logical function of tau affected during the the mechanism by which tau medi- generative diseases.

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  357


References A., v Eersel, J., Wölfing, H., Chieng, B.C., Christie, 3167–3177.
M.J., Napier, I.A., et al. (2010). Cell, this issue.
Nakazawa, T., Komai, S., Tezuka, T., Hisatsune, C.,
Aarts, M., Liu, Y., Liu, L., Besshoh, S., Arundine,
Klein, C., Kramer, E.M., Cardine, A.M., Schraven, Umemori, H., Semba, K., Mishina, M., Manabe,
M., Gurd, J.W., Wang, Y.T., Salter, M.W., and Tymi-
anski, M. (2002). Science 298, 846–850. B., Brandt, R., and Trotter, J. (2002). J. Neurosci. T., and Yamamoto, T. (2001). Biol. Chem. 276,
22, 698–707. 693–699.
Ballatore, C., Lee, V.M., and Trojanowski, J.Q.
(2007). Nat. Rev. Neurosci. 8, 663–672. Konzack, S., Thies, E., Marx, A., Mandelkow, Palop, J.J., and Mucke, L. (2009). Arch. Neurol. 66,
E.M., and Mandelkow, E. (2007). J. Neurosci. 27, 435–440.
Haass, C., and Selkoe, D.J. (2007). Nat. Rev. Mol. 9916–9927.
Cell Biol. 8, 101–112. Roberson, E.D., Scearce-Levie, K., Palop, J.J.,
Lee, G., Newman, S.T., Gard, D.L., Band, H., Yan, F., Cheng, I.H., Wu, T., Gerstein, H., Yu, G.Q.,
Ittner, L.M., Ke, Y.D., Delerue, F., Bi, M., Gladbach, and Panchamoorthy, G. (1998). J. Cell Sci. 111, and Mucke, L. (2007). Science 316, 750–754.

Noncoding RNAs: The Missing “Linc” in p53-


Mediated Repression
Anthony M. Barsotti1 and Carol Prives1,*
1
Department of Biological Sciences, Columbia University, New York, NY 10027, USA
*Correspondence: clp3@columbia.edu
DOI 10.1016/j.cell.2010.07.029

The tumor suppressor protein p53 coordinates the cellular response to stress through regulation
of gene expression. Now, Huarte et al. (2010) identify a long intergenic noncoding RNA as a new
player in p53-mediated repression of genes involved in apoptosis.

The tumor suppressor protein p53 is ery, subsequent studies identified more scriptional activation of microRNAs but
one of the cell’s most important barri- precise mechanisms occurring at specific also their processing into mature, active
ers against oncogenic transformation. By genes (reviewed in Laptenko and Prives, forms (Figure 1, middle) (Shi et al., 2010).
regulating the expression of thousands of 2006). These include p53 interacting Now, Huarte, Rinn, and their colleagues
genes, either directly or indirectly, p53 pro- with and inhibiting specific transcription (Huarte et al., 2010) add an exciting new
foundly influences cell fate in response to factors; displacement of specific activa- route through which p53 executes wide-
stress. Several decades of research have tors from promoters due to the presence spread gene repression, specifically by
established p53 as a transcriptional acti- of overlapping binding sites; the recruit- activating a long intergenic RNA (Figure
vator with high sequence specificity. How- ment by p53 of chromatin-modifying 1, bottom).
ever, p53 clearly also represses at least factors, such as histone deacetylases, LincRNAs are large RNA molecules
as many genes as it activates, if not more. which then block gene expression; and (primary transcript ≥5 kb) that are evo-
Despite this, the mechanism of repression the binding of p53 to unique “repression” lutionarily conserved across mammalian
is less well characterized than the trans- response elements. In addition, p53 may genomes. Although these RNAs are tran-
activation mechanism by p53. Now, the also inhibit genes indirectly by activat- scribed by RNA polymerase II, 5′capped,
informative study by Huarte et al. (2010) in ing transcription of factors that block and polyadenylated like normal mRNAs,
this issue lays the framework for a new and expression of specific genes. Most nota- they do not code for proteins (Guttman
elegant mechanism by which p53 globally bly, many labs have demonstrated that et al., 2009). Previous work by the Rinn
downregulates a large subset of its repres- the cell-cycle inhibitor p21 (especially group suggested that lincRNAs may
sion targets. These authors show that the within the context of Rb-family activa- repress transcription by targeting chro-
long intergenic RNA-p21 (lincRNA-p21), a tion) is a critical mediator of p53-depen- matin-modifying complexes to specific
bona fide downstream target of p53, is a dent transcriptional repression (Figure genomic loci (Khalil et al., 2009).
key inhibitor of gene expression through 1, top) (Barsotti and Prives, 2009, and In their new work, Huarte et al. (2010)
its interaction with heterogeneous ribonu- references therein). Recently, studies sought to identify specific lincRNAs that
cleoprotein K (hnRNP-K). indicate that p53 regulates microRNAs, operate within the p53 pathway. They con-
Although the first reports of gene which either degrade mRNA targets or structed a tiling microarray designed to
repression by p53 focused on suppres- inhibit their translation into protein. The detect the expression of ?400 lincRNAs.
sion of the basal transcriptional machin- p53 protein facilitates not only the tran- They then incubated this array with RNA

358  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


isolated from two mouse cell of this interaction. Indeed, a
lines engineered with control- large percentage of genes
lable expression of p53. These commonly inhibited by p53
experiments uncovered sev- and lincRNA-p21 also require
eral lincRNAs that are poten- hnRNP-K for their repression.
tial targets of p53, and the Further, hnRNP-K is recruited
authors focused on lincRNA- to promoters for genes that
p21, named for its proximity are downregulated by the
to the p53-target gene p21. p53/lincRNA-p21/ hnRNP-
Importantly, they confirmed K axis, and this recruitment
that the human ortholog of the depends on lincRNA-p21.
mouse lincRNA-p21 is con- Taken together, these results
served in both sequence and imply that hnRNP-K plays
regulation. a significant and direct role
To ascertain the role of Figure 1. Activation for Repression’s Sake in executing gene repres-
lincRNA-p21 within the p53 Activation of p53 in response to stress triggers expression of three distinct sion mediated by p53 and
pathway, the authors induced classes of targets that silence gene expression in different ways, resulting in lincRNA-p21.
different cellular outcomes.
the p53 response pathway (Top) The p21 protein inhibits cyclin-dependent kinases, thereby activating Supporting this model,
and then silenced either p53 the Rb family of proteins and inhibiting the E2F transcription factors. This previous studies found that
or lincRNA-p21. By compar- ultimately halts progression of the cell cycle. hnRNP-K is present in a
(Middle) The microRNA miR-34a degrades or inhibits translation of the mRNAs
ing microarrary data under of genes important for both cell-cycle progression and survival. repressive complex with the
these two conditions, the (Bottom) A new class of p53 targets, exemplified by the long intergenic non- linker histone H1.2. Moreover,
authors identified a large set coding RNA lincRNA-p21, is activated by p53 and then inhibits gene expres- this complex blocks chromatin
sion (Huarte et al., 2010). Although the exact mechanism of gene repression
of genes that are derepressed is unclear, lincRNA-p21 interacts with heterogeneous ribonucleoprotein K acetylation and subsequent
in response to disruption of (hnRNP-K) to downregulate a large group of genes that are repressed by p53, transcriptional activation by
both p53 and lincRNA-p21. including several that are important for cell survival. p53 and the transcription fac-
Moreover, these specific tor complex p300 (Kim et al.,
genes are highly represented in the by p53 is crucial for p53-facilitated cell 2008). In contrast, however, Moumen et
group of genes normally repressed by death (Ho and Benchimol, 2003). Further, al. (2005) found that hnRNP-K cooperates
p53. This is a significant finding because Huarte et al. show that the activation of with p53 as a coactivator of transcription
it implicates lincRNA-p21 as a potent certain proapoptotic genes by p53, such in response to DNA damage. In the system
downstream mediator of p53-dependent as Noxa and Perp, depends on lincRNA- used by Huarte and colleagues, hnRNP-K
transcriptional repression that acts on a p21. Therefore, lincRNA-p21 may also and lincRNA-p21 have independent roles
very large scale. play an important role in determining the as well, with hnRNP-K sharing an over-
As mentioned, p21 itself is also thought promoter selectivity of p53 for gene acti- lapping set of target genes with p53 that
to inhibit numerous target genes of p53. vation in addition to directing the repres- are not regulated by lincRNA-p21. Thus, it
Nevertheless, disruption of lincRNA-p21 sion of target genes. is not clear that lincRNA-p21 and hnRNP-
does not alter levels of p21 mRNA or Next, Huarte and colleagues delved K are more closely related than p53 and
protein. Furthermore, genes regulated into the mechanism by which lincRNA- hnRNP-K. Nevertheless, the large amount
by lincRNA-p21 do not appear to overlap p21 represses transcription. With great of data generated by the new microarray
with those identified in previous studies satisfaction, they identify hnRNP-K as analyses supports a more widespread
as targets of p21. These results suggest a key protein partner for lincRNA-p21. role for hnRNP-K in p53-dependent gene
that lincRNA-p21 acts independently of Best known for its role in chromatin repression than in p53-dependent gene
p21. remodeling and transcriptional regula- activation.
In response to stress, p53 initiates a tion (initiation, elongation, and termina- The Huarte et al. findings raise several
cellular program that often results in cell- tion), hnRNP-K binds to a wide range of key questions. Does the binding part-
cycle arrest or cell death. Despite the molecules, including DNA, RNA, protein ner of hnRNP-K determine whether it
large common set of genes regulated by kinases, and factors that remodel chro- acts as a positive or negative regulator
p53 and lincRNA-p21, Huarte, Rinn, and matin. Thus, hnRNP-K also participates of transcription? For example, does the
coworkers found that silencing of lin- in diverse processes, such as RNA interaction between p53 and hnRNP-K
cRNA-p21 blocks programmed cell death splicing, mRNA stability, and translation serve as a platform for other coactiva-
(i.e., apoptosis) but not cell-cycle arrest. (Bomsztyk et al., 2004). tors, whereas the association between
LincRNA-p21 represses the expression After validating the specificity of the lincRNA-p21 and hnRNP-K favors for-
of several important prosurvival factors, association between lincRNA-p21 and mation of a repressive complex? This
which may help to explain this interesting hnRNP-K and identifying the region of model would indeed explain the seem-
phenotype. This hypothesis is supported the RNA necessary for their binding, ingly contradictory roles of hnRNP-K
by previous studies demonstrating that the authors again turned to microar- with regard to p53 activity. Further, how
the repression of antiapoptotic genes ray analyses to assess the importance does lincRNA-p21 influence the binding

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  359


of hnRNP-K to specific genomic loci? References Khalil, A.M., Guttman, M., Huarte, M., Gar-
Is there functional significance to the ber, M., Raj, A., Rivea Morales, D., Thomas, K.,
Barsotti, A.M., and Prives, C. (2009). Oncogene Presser, A., Bernstein, B.E., van Oudenaarden,
remarkable proximity (?15 kb) of lin- A., et al. (2009). Proc. Natl. Acad. Sci. USA 106,
28, 4295–4305.
cRNA-p21 to the p21 gene? If these loci 11667–11672.
are regulated interdependently, they may Bomsztyk, K., Denisenko, O., and Ostrowski, J.
(2004). Bioessays 26, 629–638. Kim, K., Choi, J., Heo, K., Kim, H., Levens, D.,
act as a key molecular switch between
Kohno, K., Johnson, E.M., Brock, H.W., and An,
life and death. Finally, given the impor- Guttman, M., Amit, I., Garber, M., French, C., Lin, W. (2008). J. Biol. Chem. 283, 9113–9126.
tance of lincRNA-p21 to p53-dependent M.F., Feldser, D., Huarte, M., Zuk, O., Carey, B.W.,
Cassady, J.P., et al. (2009). Nature 458, 223–227. Laptenko, O., and Prives, C. (2006). Cell Death Dif-
cell death, is lincRNA-p21 mutated in
fer. 13, 951–961.
cancer? Answers to each of these ques- Ho, J., and Benchimol, S. (2003). Cell Death Differ.
tions will certainly enrich our under- 10, 404–408. Moumen, A., Masterson, P., O’Connor, M.J., and
standing of the functional relationship Jackson, S.P. (2005). Cell 123, 1065–1078.
Huarte, M., Guttman, M., Feldser, D., Garber, M.,
between p53 and this powerful class of Koziol, M., Kenzelmann-Broz, D., Khalil, A., Zuk, O., Shi, M., Liu, D., Shen, B., and Guo, N. (2010). Bio-
regulatory molecules, lincRNAs. Amit, I., Rabani, M., et al. (2010). Cell, this issue. chim. Biophys. Acta 1805, 218–225.

Stem Cells and DNA Damage:


Persist or Perish?
Andrew A. Lane1,2,* and David T. Scadden1,2,*
1
Center for Regenerative Medicine, Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
2
Harvard Stem Cell Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Boston, MA 02114, USA
*Correspondence: aalane@partners.org (A.A.L.), dscadden@mgh.harvard.edu (D.T.S.)
DOI 10.1016/j.cell.2010.07.030

Stem cells repopulate tissues after injury while also renewing themselves, but this makes them
vulnerable to genotoxic damage. Mohrin et al. (2010) and Milyavsky et al. (2010) now show that
mouse and human hematopoietic stem cells make opposing decisions about whether to die or to
persist in response to DNA damage.

Stem cells have the immense responsi- each day. Hematopoietic stem cells are than were more differentiated progenitor
bility of maintaining tissue and organism thought to be resistant to injury including cells (Figure 1). The unique DNA damage
homeostasis over the lifetime of an indi- DNA damage, which may be related to response of mouse HSPCs involves the
vidual. As such, stem cells are speculated their specific gene expression programs, tumor suppressor protein p53 and is lost
to have evolutionary characteristics that epigenetic factors, or exogenous protec- when stem cells are forced out of qui-
offer protection against acute insults, tion by the stem cell “niche.” Two new escence and into the cell cycle by treat-
allowing them to survive and to repopu- reports in Cell Stem Cell from the labora- ment with chemotherapy or cytokines.
late their tissues in the short term. How- tories of Emmanuelle Passegué (Mohrin Not only are quiescent HSPCs poised
ever, they must also act as self-renew- et al., 2010) and John Dick (Milyavsky et to resist apoptosis as evidenced by their
ing guardians of the genome to ensure al., 2010) further our understanding of antiapoptotic gene expression program,
maximal integrity of the genomic code how hematopoietic stem cells respond but they are also able to repair their DNA
for future stem cells and their mature tis- to radiation-induced DNA damage. by nonhomologous end joining (NHEJ).
sue progeny. The hematopoietic (blood) So how do quiescent stem cells han- Repair of DNA damage through homolo-
system is perhaps the best studied tis- dle genotoxic insults? Mohrin et al. (2010) gous recombination (which has a lower
sue in terms of its hierarchical develop- found that murine hematopoietic stem error rate than NHEJ) requires that cells
ment from a small number of long-term and progenitor cells (HSPCs)—defined as enter the cell cycle; thus, quiescent stem
stem cells that are relatively quiescent, bone marrow cells expressing the mark- cells must rely on NHEJ as an alterna-
to progenitors that proliferate and dif- ers: lineage − /c-Kit+/Sca-1+/Flk2−—were tive. The reliance of quiescent adult tis-
ferentiate, and then to mature blood cell more resistant to apoptosis induced sue stem cells on NHEJ for the repair of
lineages that are produced by the billion by a specific dose of ionizing radiation DNA damage may in fact be a general

360  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


phenomenon in mice, given umbilical cord blood HSPCs
the similar conclusions of a have a different biology than
recent study using hair follicle the bone marrow HSPCs of
stem cells as a model system mouse. However, it is tempt-
(Sotiropoulou et al., 2010). ing to see the different find-
Unfortunately, Mohrin et al. ings in the light of evolution,
(2010) also uncover a down- that is, as a reflection of the
side to short-term radioresis- different challenges faced by
tance and rapid DNA repair mammals with different life
through the error-prone NHEJ spans and ages of reproduc-
pathway. Spectral karyotyping tive maturity.
revealed gross chromosomal Recent elegant studies from
aberrations in irradiated mouse Bondar and Medzhitov (2010)
HSPCs, and some of the same and Marusyk et al. (2010)
abnormal cytogenetic find- demonstrate that competitive
ings persisted in the progeny selection takes place within
of irradiated HSPCs trans- tissue stem cell populations.
planted into mouse recipients. These authors found that irra-
Furthermore, despite their diated p53-deficient HSPCs
resistance to apoptosis imme- in the mouse have an initial
diately after injury, irradiated survival advantage but that
HSPCs were unable to con- Figure 1. Hematopoietic Cell Responses to DNA Damage long-term fitness is balanced
tribute to long-term sustained (Top) Quiescent murine hematopoietic stem and progenitor cells (HSPCs) by complex interactions with
are poised to survive DNA damage induced by low-level ionizing radiation
hematopoiesis when seri- through a DNA repair process called nonhomologous end joining (NHEJ), neighboring HSPCs and the
ally transplanted into mouse which tends to be error prone (Mohrin et al., 2010). In contrast, mouse HSPCs relative levels of p53 and DNA
recipients. Such events would and committed progenitors (CP) progressing through the cell cycle are more damage in stem cells and their
be of obvious risk to a long- likely to undergo apoptosis or repair their DNA using higher-fidelity homolo-
gous recombination. Although the short-term consequence of HSPC survival
neighbors. The studies from the
lived organism as serial expo- is maintenance of tissue integrity in the face of injury, long-term consequenc- Passegué and Dick labs indi-
sure of stem cells to genotoxic es include genomic rearrangements that persist and HSPCs with a diminished cate that hematopoietic cells
agents could readily result in functional capacity. within a tissue have adopted
(Bottom) In contrast, compared to more committed progenitors, the default
leukemia or aplasia. program for damaged human HSPCs is to undergo apoptosis. However, a de- different means of handling
Does the human hemato­ crease in p53 rescues human HSPCs from apoptosis immediately after low- DNA damage depending on
poietic system accept the level irradiation (Milyavsky et al., 2010). Despite interspecies differences in their differentiation stage. That
the short-term response to radiation, the long-term functional consequences
same tradeoff between stem of avoiding apoptosis for both mouse and human HSPCs include persistent mouse and human stem cells
cell survival in the short term DNA damage and decreased self-renewal capacity. The delicate balance be- may have acquired or under-
versus accumulation of del- tween tissue survival and the DNA damage response therefore could predis- gone selection for distinct
pose surviving HSPCs to future malignant transformation.
eterious mutations in the long responses to ionizing radiation
term? In a companion study, is a reasonable notion. How-
Milyavsky et al. (2010) addressed this ent mice. Therefore, a short-term gain ever, it remains to be seen which specific
question in human umbilical cord blood in survival could be achieved by human molecular mechanisms that differ between
cells. They observed an enhanced sen- hematopoietic stem cells as found in the stem cells and progenitors, or between
sitivity to apoptosis induced by low-dose mouse, but the default setting for irradi- stem cells of different species, lead to
ionizing radiation in these cells compared ated human HSPCs is an increase in p53 these distinctive traits. We now have a set
to more differentiated cells. In contrast expression resulting in apoptosis. of reagents with which to discover and
to the findings of Mohrin et al. in the The differences between these two understand how such important yet differ-
mouse, these authors noted that human studies may have a technical basis: mark- ent tissue stem cell traits have evolved.
hematopoietic stem and early multipotent ers for stem and progenitor populations There are other practical implications
progenitor cells were poised for apopto- are more refined in the mouse than in the of the Passegué and Dick reports. Sec-
sis in response to DNA damage. Survival human so somewhat different stem and ondary myelodysplasia and leukemia are
and the clonogenic and reconstitutive progenitor cell populations may have believed to arise from DNA damage to
capacity of the irradiated human HSPCs been analyzed. In addition, slightly differ- HSPCs from the radiation or chemother-
were rescued by blocking p53 expression ent doses of radiation were used. In the apy given to treat a primary malignancy.
or by overexpression of the antiapoptotic in vivo experiments of Milyavsky et al., Mohrin et al. show intriguing evidence that
factor Bcl-2. However, irradiated human human HSPCs from umbilical cord blood NHEJ activity and chromosomal aberra-
HSPCs lacking p53 were unable to sus- were transplanted into the mouse bone tions decrease when HSPCs are induced
tain hematopoiesis and showed evidence marrow niche, which may have provided to enter the cell cycle prior to irradiation.
of persistent DNA double-strand breaks less efficient survival signals for human Interestingly, a parallel evolving hypoth-
when serially transplanted into recipi- cells than for mouse cells. Also, human esis in the study of cancer stem cells sug-

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  361


gests that activating leukemia stem cells acute injury and long-term fitness needs Milyavsky, M., Gan, O.I., Trottier, M., Komosa, M.,
Tabach, O., Notta, F., Lechman, E., Hermans, K.G.,
from quiescence prior to chemotherapy to be more fully understood and will
Eppert, K., Konovalova, Z., et al. (2010). Cell Stem
may result in more efficient elimination of require both laboratory models and the Cell 7. Published online July 7, 2010. 10.1016/j.
these cancer-repopulating cells (Saito et thoughtful correlative study of stem cells stem.2010.05.016.
al., 2010). An unexpected benefit of such from patients receiving genotoxic chemo-
Mohrin, M., Bourke, E., Alexander, D., Warr,
a prestimulation strategy may be that nor- therapy. Understanding these events may M.R., Barry-Holson, K., Le Beau, M.M., Mor-
mal hematopoietic stem cells activated point the way to methods for preserving rison, C.G., and Passegué, E. (2010). Cell Stem
from quiescence would simultaneously short-term tissue reconstitution while Cell 7. Published online July 7, 2010. 10.1016/j.
stem.2010.06.014.
be protected from accumulating long- maintaining long-term cell and genomic
term DNA damage. However, as shown integrity. Saito, Y., Uchida, N., Tanaka, S., Suzuki, N., Tomi-
by Milyavsky and colleagues, stem cell zawa-Murasawa, M., Sone, A., Najima, Y., Takagi,
References S., Aoki, Y., Wake, A., et al. (2010). Nat. Biotech-
escape from acute damage, particularly if nol. 28, 275–280.
it involves a decrease in p53 activity, may Bondar, T., and Medzhitov, R. (2010). Cell Stem
Sotiropoulou, P.A., Candi, A., Mascré, G., De Cler-
lead to long-term deleterious effects on Cell 6, 309–322.
cq, S., Youssef, K.K., Lapouge, G., Dahl, E., Se-
stem cell fitness and repopulating ability. Marusyk, A., Porter, C.C., Zaberezhnyy, V., and meraro, C., Denecker, G., Marine, J.C., and Blan-
The interplay between the response to DeGregori, J. (2010). PLoS Biol. 8, e1000324. pain, C. (2010). Nat. Cell Biol. 12, 572–582.

Mitochondrial Matrix Reloaded with RNA


Toshiya Endo,1,* Koji Yamano,1 and Tohru Yoshihisa2,3
1
Department of Chemistry, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya 464-8602, Japan
2
Research Center for Materials Science, Nagoya University, Chikusa-ku, Nagoya 464-8602, Japan
3
Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
*Correspondence: endo@biochem.chem.nagoya-u.ac.jp
DOI 10.1016/j.cell.2010.07.024

Although mitochondrial biogenesis requires the import of specific RNAs, the pathways and cellular
machineries involved are only poorly understood. Wang et al. (2010) now find that polynucleotide
phosphorylase in the intermembrane space of mammalian mitochondria facilitates import of
several RNAs into the mitochondrial matrix.

Mitochondria, the power plants of the 2008; Lithgow and Schneider, 2010). In cursor engages with the translocase of
eukaryotic cell, are bound by two mem- this issue of Cell, Wang et al. (2010) shed the inner membrane 23 (TIM23) complex
branes and contain 1000–1500 different light on this question, revealing that poly- (Figure 1) (Chen et al., 2006; Rainey et al.,
proteins and tens of RNAs. Most of the nucleotide phosphorylase (PNPase) is a 2006). After the PNPase presequence is
genes that encode mitochondrial pro- much sought after component of the RNA removed by matrix processing pepti-
teins are found in the nuclear genome import apparatus in mammalian cells. dase (MPP), an AAA protease Yme1 in
and thus are translated in the cytosol PNPases comprise an evolutionally the inner membrane pulls PNPase into
and then imported into mitochondria. conserved enzyme family (found in bac- the IMS, where PNPase assembles into
The pathways and machineries required teria, plants, flies, and mammals but not a trimeric complex (Figure 1).
for protein import into mitochondria in yeast) that has 3′→5′ exoribonuclease Wang et al. now assess the function of
have been extensively studied and are and RNA-polymerase activities (Chen et mammalian PNPase by tissue-specific
highly conserved among fungi, plants, al., 2007). Although bacterial PNPases disruption of the PNPase gene in mouse
and mammals (Endo and Yamano 2009; are cytosolic, eukaryotic PNPases are hepatocytes. They find that mitochondria
Chacinska et al., 2009). The mitochon- mainly localized in mitochondria or chlo- from hepatocytes deficient in PNPase
drial matrix also contains several kinds of roplasts. Prior work has established display defects in oxidative phosphory-
noncoding RNAs that are also imported how PNPases get to the intermembrane lation (OXPHOS), the major ATP-gener-
from the cytosol. However, in contrast to space (IMS). After crossing the mito- ating metabolic pathway of respiration.
protein translocation, the mechanisms chondrial outer membrane via the trans- This defect is shown to arise from the
that mediate import of RNAs into mito- locase of outer mitochondrial membrane failure in the processing of polycistronic
chondria remain enigmatic (Salinas et al., 40 (TOM40) complex, the PNPase pre- mitochondrial mRNAs encoding the

362  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


subunits of the OXPHOS complexes. But
how does PNPase in the IMS affect the
processing of RNAs in the matrix?
A hint to the answer came from the
observation that the RNA component
of mammalian mitochondrial RNase
P is markedly decreased by PNPase
depletion. Given that RNase P medi-
ates processing of mitochondrial trans-
fer RNAs (tRNAs), which are encoded
between open reading frames for
OXPHOS components, failure in tRNA
excision also impairs maturation of the
OXPHOS subunit transcripts. In addi-
tion, expression of human PNPase in
yeast mitochondria, which do not pos-
sess PNPase, enhances the import effi-
ciency of heterologous human RNase P
RNA, 5S ribosomal RNA, and the RNA
component of MRP, a ribonucleopro-
tein that mediates mitochondrial DNA Figure 1. PNPase Facilitates the Import of RNAs into the Mitochondrial Matrix
replication. Similar PNPase-dependent The polynucleotide phosphorylase (PNPase) precursor is imported into the intermembrane space with
import in vitro is confirmed with mito- the aid of the translocase of outer mitochondrial membrane 40 (TOM40) complex, the translocase of
the inner mitochondrial membrane (TIM23) complex, matrix processing peptidase (MPP), and AAA
chondria isolated from PNPase-defi- protease Yme1. The trimeric PNPase promotes the import of RNAs from the cytosol into the matrix.
cient mouse liver or embryonic fibro- Mitochondrial mRNAs are processed in the matrix and their translation products are assembled into
blasts. These findings suggest that the oxidative phosphorylation (OXPHOS) complexes I–IV. The OXA complex integrates proteins from
the matrix into the inner membrane. OM, outer membrane; IM, inner membrane. Heat shock protein
PNPase in the IMS is a component of 70 (Hsp70) is shown as an import motor for presequence-containing proteins. AAC, ADP-ATP carrier;
the RNA import system in human mito- DΨ, membrane potential.
chondria.
What is the mechanism of RNA import to substrate RNAs may also contribute to place center stage. Will the unfolding
that is facilitated by PNPase? In yeast and the unidirectional translocation of RNAs drama of RNA translocation in mitochon-
plant mitochondria, substrate tRNAs are across the outer membrane via a trap- dria reveal common principles or expose
recognized by the mitochondrial surface ping mechanism or by preventing their a diversity of organism specific mecha-
components including the import recep- backward movement to the cytosol. nisms?
tor Tom20, and then move through the The identity of the import chan-
Tom40 channel or voltage-dependent nel in the inner membrane is unknown, References
anion channel (VDAC). This movement although Wang et al. find that translo-
Chacinska, A., Koehler, C.M., Milenkovic, D.,
probably involves several mechanisms, cation of RNase P RNA into yeast mito-
Lithgow, T., and Pfanner, N. (2009). Cell 138,
including a piggyback mechanism in chondria with human PNPase requires 628–644.
which the RNA substrate is associated the membrane potential. This suggests
with an appropriate escort protein (Sali- that the membrane potential may help Chen, H.-W., Rainey, R.N., Balatoni, C.E., Dawson,
D.W., Troke, J.J., Wasiak, S., Hong, J.S., McBride,
nas et al., 2008; Lithgow and Schneider, remove bound RNAs from PNPase or H.M., Koehler, C.M., Teitell, M.A., and French, S.W.
2010). Although no receptor or translo- facilitate unidirectional translocation of (2006). Mol. Cell. Biol. 26, 8475–8487.
cation channel for RNA import is known RNAs across the inner membrane (Fig-
Chen, H.-W., Koehler, C.M., and Teitell, M.A.
for mammalian mitochondria, the pres- ure 1). Translocation of RNAs through (2007). Trends Cell Biol. 17, 600–608.
ent study reveals that PNPase binds to the inner membrane import channel may
substrate RNAs with a specific stem- also require an additional protein, such as Endo, T., and Yamano, K. (2009). Biol. Chem. 390,
723–730.
loop structure. When grafted onto other mitochondrial heat shock protein 70, as
nonsubstrate RNAs, this stem-loop can an import motor for presequence-con- Lithgow, T., and Schneider, A. (2010). Philos. Trans.
direct the RNA into yeast mitochondria taining mitochondrial proteins. Although R. Soc. Lond. B Biol. Sci. 365, 799–817.
containing human PNPase. Therefore, many of the components of the yeast and Rainey, R.N., Glavin, J.D., Chen, H.-W., French,
after crossing the outer membrane by an human RNA import pathway remain to S.W., Teitell, M.A., and Koehler, C.M. (2006). Mol.
unknown mechanism, substrate RNAs be identified, the efficient in vitro system Cell. Biol. 26, 8488–8497.
with a specific stem-loop structure are developed by Wang et al. should acceler- Salinas, T., Duchêne, A.-M., and Maréchal-Drouard,
recognized by PNPase, which subse- ate the pace of discovery. L. (2008). Trends Biochem. Sci. 33, 320–329.
quently allows the transfer of a subset of The study by Wang et al. casts a spot-
Wang, G., Chen, H.-W., Oktay, Y., Zhang, J., Allen,
RNAs to downstream components of the light on mitochondrial RNA import, with E.L., Smith, G.M., Fan, K.C., Hong, J.S., French,
import pathway. The binding of PNPase mammalian PNPase taking its rightful S.W., McCaffery, J.M., et al. (2010). Cell, this issue.

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  363


Leading Edge

Minireview

A MAP for Bundling Microtubules


Claire E. Walczak1,* and Sidney L. Shaw2,*
1
Medical Sciences
2
Department of Biology
Indiana University, Bloomington, IN 47405, USA
*Correspondence: cwalczak@indiana.edu (C.E.W.), sishaw@indiana.edu (S.L.S.)
DOI 10.1016/j.cell.2010.07.023

Microtubules assemble into arrays of bundled filaments that are critical for multiple steps in cell
division, including anaphase and cytokinesis. Recent structural and functional studies, including
two papers in this issue of Cell (Bieling et al., 2010; Subramanian et al., 2010), demonstrate how
the MAP65 protein PRC1 crosslinks microtubules and cooperates with kinesin motors to control
the dynamics and size of bundled regions.

The microtubule cytoskeleton is a remarkable structure that The polarity of microtubules within a bundle of overlapping
can adopt diverse architectures uniquely suited to the indi- filaments is critical to the action of motor proteins that slide
vidual needs of a particular cell type or process. For example, filaments past each other and drive the movement of cargoes,
in vertebrate cells, the mitotic spindle, which separates chro- such as chromosomes, on these microtubules. For example,
mosomes during anaphase, contains two antiparallel arrays during late anaphase, the overlapping regions of microtubules
of microtubules with their minus ends anchored at opposing at the center of the mitotic spindle elongate as the microtubules
centrosomes and their plus ends overlapping to form a bundle push the spindle poles to opposite sides of the cell. However,
of crosslinked filaments in the middle of the spindle (Figure 1, to separate daughter cells during cytokinesis, this overlapping
bottom inset). In contrast, plant (angiosperm) cells do not pos- region shortens and forms a dense, compact array of anti-
sess discrete microtubule organizing centers (i.e., centrioles) parallel microtubules, called the midzone. How does the cell
but instead rely primarily on specific interactions between specify the size of the overlapping region in a bundle and man-
microtubules to organize the filaments into crosslinked arrays age the timing and position of its remodeling? To answer these
(Ehrhardt, 2008). questions requires a better understanding of the key molecular
The variety of microtubule structures observed across differ- players that govern the formation of microtubules.
ent cell types requires a diverse group of proteins to assemble,
stabilize, and dynamically control these microtubule arrays. MAP65 Family of Microtubule Crosslinking Proteins
Microtubule-associated proteins (MAPs), which include both One major class of proteins that crosslink microtubules into
molecular motors and nonmotor proteins, regulate the global arrays is the MAP65/Ase1/PRC1 family. Biochemical studies
properties of microtubule structures by moving and crosslinking with plant extracts identified the first members of this family
filaments. Although much is known about these individual pro- as 65 kD proteins capable of bundling microtubules (Chang-
teins, key questions remain about how they interact to control Jie and Sonobe, 1993). Subsequent studies demonstrated that
the size, shape, and dynamics of microtubule arrays. Now, two MAP65 proteins crosslink microtubules in vitro with a spacing
studies in this issue of Cell (Bieling et al., 2010; Subramanian et between microtubule filaments of 25 nm (Chan et al., 1999),
al., 2010) demonstrate how the MAP65 protein PRC1 (protein consistent with in situ observations of microtubule bundles in
regulator of cytokinesis 1) independently bundles microtubules plants. A genetic screen identified the yeast ortholog of MAP65
into antiparallel arrays and works with two motors, kinesin-4 as Ase1, which is required for properly elongating the spindle
and kinesin-5, to control the global properties of these overlap- during anaphase (Pellman et al., 1995). Ase1 was later shown
ping regions. Together, these papers suggest a model for how to be a homodimer that also bundles microtubules in vitro
microtubule bundles can persist despite the action of numer- (Schuyler et al., 2003), an activity that is critical for its role in
ous motor proteins acting along them. sliding microtubule filaments past each other during anaphase.
The vertebrate ortholog of MAP65/Ase1 is PRC1, and in mam-
Microtubule Structure and Dynamics malian cells, PRC1 regulates the organization of the central
Microtubules are linear polymers inside the cell composed of α/β- spindle during cytokinesis (Jiang et al., 1998; Mollinari et al.,
tubulin heterodimers arranged head to tail into protofilaments. The 2002). Phosphorylation of PRC1 by cyclin dependent kinase 1
protofilaments, typically 13, associate laterally to form a hollow (Cdk1) negatively regulates the crosslinking activity of PRC1,
tube with substantial flexural rigidity and inherent structural polar- which limits the bundling of microtubules by PRC1 until late
ity, described as having plus and minus ends (Figure 1). Microtu- stages of mitosis when they are needed for cytokinesis (Zhu
bules exhibit dynamic instability (Mitchison and Kirschner, 1984) et al., 2006).
wherein individual microtubules within a population interconvert Like other MAP65 proteins, PRC1 and Ase1 are not molec-
between states of growth and shortening. In general, microtubule ular motors themselves but instead work in concert with
plus ends are more dynamic than minus ends. motor proteins to organize arrays of microtubules. In fis-

364  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


Figure 1. PRC1 Controls Microtubule
­Assembly
(A) Protein regulator of cytokinesis 1 (PRC1) can
initiate crosslinking of dynamic microtubules (MT)
that interact in an antiparallel fashion (top). Kine-
sin-4 is a molecular motor directed at the plus
ends of microtubules; it accumulates in the re-
gion where microtubules crosslink as their plus
ends grow (middle). The interaction of kinesin-4
with PRC1 increases the dwell time of kinesin-4
on microtubules, which in turn limits the length
of the overlap region by blocking microtubule
growth at the plus ends (bottom) (Bieling et al.,
2010).
(B) Kinesin-5 is also a molecular motor directed at
the plus ends of microtubules, but kinesin-5 can
slide microtubules past each other (top). When
kinesin-5 is added to crosslinked microtubules
(middle), PRC1 maintains the crosslinks despite
the sliding action (bottom) (Subramanian et al.,
2010).
(C) PRC1 forms a homodimer that interacts
through its central spectrin domains with two mi-
crotubules to crosslink the antiparallel filaments.
Although PRC1 is shown to associate predomi-
nantly with α-tubulin, the current resolution of
the structures presented by Subramanian et al.
(2010) is not sufficient to distinguish between
binding to α- or β-tubulin.

tubulin dimers to polymerize dynamic


microtubules. They then added PRC1
proteins labeled with fluorescent tags
to crosslink the microtubules (Figure
1A). The PRC1 proteins bound prefer-
entially to microtubules that overlap at
antiparallel regions, showing decisively
that PRC1 alone is sufficient to cross-
sion yeast, Ase1 dynamically controls the overlap of bundles link antiparallel microtubules.
by coordinating with the kinesin-14 motor klp2 (Janson et Knowing that PRC1 interacts directly with Kif4 (Xklp1 in Xeno-
al., 2007). In mammalian cells, PRC1 is transported to the pus), Bieling et al. next determined how Xklp1 alters the forma-
midzone of the spindle by Kif4 (kinesin family member 4), a tion of microtubule bundles by PRC1. Previous studies showed
kinesin-4 motor protein that is critical for positioning chro- that a truncated version of Xklp1 could inhibit both growth and
mosomes and for cytokinesis in multiple organisms (Glotzer, shrinkage of microtubules at particular ends (Bringmann et al.,
2009; Hornick et al., 2010). Disruption of either PRC1 or Kif4 2004), suggesting that Xklp1 regulates the dynamics of micro-
perturbs the localization of the other protein, making it diffi- tubules within bundles. Indeed, bundles of antiparallel microtu-
cult to elucidate whether the Kif4 motor recruits PRC1 to the bules still formed when Xklp1 and PRC1 were added together
microtubules or whether loss of PRC1 disrupts the localiza- to the polymerizing microtubules, but, remarkably, the bundles
tion of the central spindle and thus Kif4. Nevertheless, PRC1 grew to a fixed size when Xklp1 was present. The authors show
is required to set up the central spindle before a number of that this limit in bundle length is due to cessation of growth
other kinesins locate to the spindle. These kinesins include at the plus end of the microtubules and that the steady-state
motors involved in finishing the assembly of the central spin- length of the overlapping region depends on the concentration
dle and in cytokinesis, including mitotic kinesin-like protein of Xklp1 (Figure 1A).
1 (MKLP-1) and 2 and Kif14 (Glotzer, 2009; Hornick et al., These findings uncover an elegant and simple system capable
2010). Nevertheless, it is still unknown how PRC1 interacts of self-organizing into a bundle of microtubules with a defined
with these motor proteins to direct the size, shape, and sta- length. Moreover, they show that only two additional proteins,
bility of the central spindle. PRC1 and Xklp1, are required for making stable microtubule
bundles from highly dynamic polymers, with PRC1 generating
Kinesin-4 Limits Microtubule Overlap Length the bundles and Xklp1 controlling their length.
To understand how PRC1 organizes microtubule arrays, Biel- To understand how Xklp1 “monitors” and regulates the
ing and colleagues (2010) developed a total internal reflection size of the overlap region between microtubules, Bieling et al.
fluorescence (TIRF) microscopy assay in which microtubule next measured how Xklp1 changes the dynamics of individual
seeds are attached to a microscope slide and mixed with microtubules that are not crosslinked. They found that Xklp1

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  365


alone blocks the growth of microtubules but only at much motion of the microtubules by the kinesin-5 motors. This is
higher concentrations of Xklp1 than were required to limit the important because during cytokinesis these crosslinks must
length of microtubule bundles in the presence of PRC1. Biel- remain in place to preserve the organization of the microtu-
ing et al. then demonstrate that the ability of Xklp1 to limit the bules in the midzone. At the same time, the crosslinks must
bundle length of crosslinked microtubules requires the proces- allow the microtubules to slide past each other to achieve and
sive motility of Xklp1. This suggests that Xklp1 motility plays an maintain complete segregation of the chromosomes.
active role in bundle architecture rather than simply acting to
regulate microtubule growth. PRC1 Has a Spectrin-like Microtubule-Binding Domain
The key connection, however, came when the authors found How does PRC1 maintain the structure of midzone bundles
that the presence of PRC1 actually increases the time that Xklp1 while simultaneously allowing the overlap region to adjust to
stays within the overlapping region of two microtubules (i.e., the changing architecture of the central spindle? To answer
the dwell time). Together with the previous results, this sug- this question requires a better understanding of the structural
gests a model in which PRC1 forms antiparallel crosslinks of and biophysical aspects of how PRC1 bundles microtubules.
microtubules and recruits Xklp1 within the crosslinked regions, PRC1 has three prominent domains: the N-terminal domain,
where it walks toward the plus ends of microtubules (Figure which mediates homodimerization; the central domain, which
1B). The effective concentration of Xklp1, which depends on the contains the major site for binding to microtubules; and the
concentration of PRC1, determines the extent to which Xklp1 C-terminal domain, which regulates the interaction with micro-
blocks microtubule growth and thus the steady-state length of tubules (Figure 1C). Using time-lapse TIRF microscopy, Subra-
the microtubule bundle. Thus, together Xklp1 and PRC1 deter- manian et al. found that PRC1 diffuses one-dimensionally along
mine the size and stability of the overlapping zone between the microtubule lattice for an average of 7 s, and the C-terminal
microtubules. Control of bundle length is vital because the regulatory domain enhances this association. These findings
central spindle must elongate during late anaphase but then support the idea that binding of PRC1 to microtubules is medi-
shrink to a shorter size to form the midzone during cytokinesis. ated by both the central microtubule-binding domain and the
Changes in PRC1 binding or the activity of Xklp1 during these unstructured C-terminal region. This latter domain contains a
morphological transitions may provide a mechanism to control large number of positively charged residues (i.e., lysines and
the morphology and function of the central spindle. arginines), which is a common feature of regions that interact
with microtubules.
PRC1 Forms Compliant Crosslinks between Microtubules To gain further insight into how PRC1 interacts with micro-
The above study illustrates how a stable bundle of microtu- tubules, Subramanian and colleagues determined the X-ray
bules of a fixed size can form in the presence of two proteins crystal structure of the central microtubule-binding domain
known to function at the midzone in vertebrate cells. However, of PRC1 and cryo-electron microscopy (cryo-EM) reconstruc-
the work does not explain how PRC1 behaves under conditions tions of PRC1 fragments bound to microtubules. They found
where other forces may be acting on the bundles. For example, that the central domain of PRC1 consists of a three-helix bun-
molecular motors actively slide crosslinked microtubules past dle (?70 Å long) with connecting loops between the helices.
each other during the late stages in mitosis. A cluster of highly conserved and positively charged residues
To characterize how PRC1 and its crosslinking activity exists at the interface between α helix 1 and α helix 2 within
modulates or affects the sliding of bundled microtubules, Sub- this helical bundle. Mutation of these residues diminished but
ramanian et al. (2010) take advantage of elegant microscopy did not abolish microtubule binding by PRC1, further support-
assays they developed in an earlier study to visualize microtu- ing the hypothesis that PRC1 possesses two major surfaces
bule filaments sliding past each other in vitro by the kinesin-5 that contact microtubules.
motors (Kapitein et al., 2005) (Figure 1B). Kinesin-5 proteins are Interestingly, this domain shares structural homology with
important for establishing the bipolarity of spindles during the the spectrin domains found in actin-binding proteins (Djinovic-
early stages of mitosis by actively sliding apart microtubules Carugo et al., 2002). Spectrin domains are not required for
of opposite polarity. Furthermore there is evidence that these interaction with actin filaments; instead, they typically link
molecular motors slide antiparallel microtubules apart during together different functional domains of actin-binding proteins.
late anaphase (i.e., anaphase B). The present findings by Subramanian and colleagues identify
Using this assay, Subramanian et al. now find that PRC1 a new role for spectrin domains in regulating the microtubule
displays two distinct behaviors in the presence of kinesin-5. cytoskeleton.
In certain cases, the concentration of PRC1 in the overlap Cryo-EM reconstructions of microtubules interacting with a
region and, thus the bundling length, stay constant while one truncated fragment of PRC1, which includes the homodimeriza-
microtubule slides relative to another filament in the cross- tion domain and the spectrin domain but not the C-terminal
linked region. In the second case, the length of the crosslinked domain, revealed that PRC1 crosslinks nearly all microtubules
region reduces at a rate similar to that at which the filaments in an antiparallel manner with a spacing of ?35 nm between
slide past each other. Although in this case PRC1 still tracked filaments (Figure 1C). Notably, the PRC1 molecules bound to
the microtubule overlap zone, the crosslinking protein did not crosslinked microtubules were more structured than those
reduce the velocity at which filaments slide even when excess bound to a single microtubule. This suggests that crosslink-
PRC1 was present. Together, these results demonstrate that ing itself converts PRC1 from an inherently flexible molecule to
the crosslinks by PRC1 do not significantly resist the sliding a rigid one, which may enhance PRC1’s crosslinking activity.

366  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


In both cases, the cryo-EM data indicate that PRC1 interacts ingly, another study recently found that phosphorylation of
with one α/β-tubulin heterodimer and extends as a single rod an unstructured region of the kinetochore attachment protein
shape almost perpendicular to the microtubule lattice (Figure Hec1 also controls its affinity for microtubules (Guimaraes et
1C). Remarkably, the crystal structure of the spectrin domain al., 2008). Thus, phosphorylation of unstructured domains may
of PRC1 fits nicely into the cryo-EM density with the conserved play a general role in regulating the affinities of microtubule-
basic residues between α helix 1 and α helix 2 residing at the binding proteins. Finally, understanding how distinct structural
microtubule surface. The binding site of PRC1 on the microtu- modifications of the PRC1 protein affect the morphology of
bule surface is slightly displaced relative to where many motor microtubule cytoskeletal arrays in vivo will be an important
proteins interact, providing a possible clue for how motors and avenue of future research endeavors.
crosslinking proteins can bind simultaneously to the same sur-
face of the microtubule. Acknowledgments
The above studies clearly define the key microtubule-binding
The authors thank Clive Lloyd, Yixian Zheng, and Stephanie Ems-McClung
element of PRC1, but the single-molecule studies using trun-
for helpful discussions. The authors are supported by National Institutes of
cated derivatives of PRC1 suggest that PRC1 has a second Health grant GM059618 to C.E.W. and National Science Foundation grant
microtubule-binding domain at its C terminus or this C-termi- 0920555 to S.L.S.
nal domain somehow regulates the interaction of the spectrin
domain with the microtubule surface. To address this ques- References

tion, Subramanian and colleagues obtained a second cryo-EM


Bieling, P., Telley, I.A., and Surrey, T. (2010). Cell, this issue.
reconstruction of a PRC1 construct containing the spectrin
microtubule-binding domain and the C-terminal region. This Bringmann, H., Skiniotis, G., Spilker, A., Kandels-Lewis, S., Vernos, I., and
fragment bound in a similar position on the microtubule lat- Surrey, T. (2004). Science 303, 1519–1522.
tice as the PRC1 construct containing only the dimerization Chan, J., Jensen, C.G., Jensen, L.C., Bush, M., and Lloyd, C.W. (1999). Proc.
and spectrin domains (Subramanian et al., 2010). Therefore, Natl. Acad. Sci. USA 96, 14931–14936.
the most attractive hypothesis is that the C-terminal domain Chang-Jie, J., and Sonobe, S. (1993). J. Cell Sci. 105, 891–901.
enhances the affinity of PRC1 with microtubules rather than
forming a distinct second site for microtubule binding. Djinovic-Carugo, K., Gautel, M., Ylänne, J., and Young, P. (2002). FEBS Lett.
513, 119–123.

Future Perspectives Ehrhardt, D.W. (2008). Curr. Opin. Cell Biol. 20, 107–116.
The two current papers by Bieling et al. and Subramanian et Fu, C., Yan, F., Wu, F., Wu, Q., Whittaker, J., Hu, H., Hu, R., and Yao, X. (2007).
al. provide critical insight into how inherently dynamic micro- Cell Res. 17, 449–457.
tubules are organized into functional sub-assemblies, which
Glotzer, M. (2009). Nat. Rev. Mol. Cell Biol. 10, 9–20.
are fundamental to multiple biological systems. It is remark-
able that just two proteins are sufficient to reconstitute the Guimaraes, G.J., Dong, Y., McEwen, B.F., and Deluca, J.G. (2008). Curr. Biol.
morphological subassembly of the spindle midzone. However, 18, 1778–1784.

it is essential to remember that these proteins do not work in Hornick, J.E., Karanjeet, K., Collins, E.S., and Hinchcliffe, E.H. (2010). Semin.
isolation in vivo but rather function in the complex milieu of Cell Dev. Biol. 21, 290–299.
the central spindle. The finding that PRC1 induces bundles of Janson, M.E., Loughlin, R., Loïodice, I., Fu, C., Brunner, D., Nédélec, F.J., and
microtubules that remain compliant to the action of kinesin-5 Tran, P.T. (2007). Cell 128, 357–368.
is key for understanding how the microtubules in the midzone
Jiang, W., Jimenez, G., Wells, N.J., Hope, T.J., Wahl, G.M., Hunter, T., and
slide apart while still maintaining an organized structure. It Fukunaga, R. (1998). Mol. Cell 2, 877–885.
will be interesting to add kinesin-5 to the mixture of Xklp1 and
Kapitein, L.C., Peterman, E.J., Kwok, B.H., Kim, J.H., Kapoor, T.M., and
PRC1 to see how the system responds to regulators of both
Schmidt, C.F. (2005). Nature 435, 114–118.
microtubule growth and microtubule sliding, a situation that
more closely reconstitutes the physiological one. Mitchison, T., and Kirschner, M. (1984). Nature 312, 237–242.
The work presented here also opens the doors to crucial Mollinari, C., Kleman, J.P., Jiang, W., Schoehn, G., Hunter, T., and Margolis,
structure-function and signaling studies on the PRC1 family R.L. (2002). J. Cell Biol. 157, 1175–1186.
of proteins. The identification of residues that clearly form the
Pellman, D., Bagget, M., Tu, Y.H., Fink, G.R., and Tu, H. (1995). J. Cell Biol.
attachment site to microtubules will allow for the engineering of 130, 1373–1385.
mutations in PRC1 that modulate the strength of its interaction
Schuyler, S.C., Liu, J.Y., and Pellman, D. (2003). J. Cell Biol. 160, 517–528.
with microtubules. Previous work has shown that phosphoryla-
tion regulates PRC1’s interaction with the central spindle (Fu et Subramanian, R., Wilson-Kubalek, E.M., Arthur, C.P., Bick, M.J., Campbell,
al., 2007). It is interesting that those phosphorylation sites map E.A., Darst, S.A., Milligan, R.A., and Kapoor, T.M. (2010). Cell, this issue.
to the unstructured C-terminal domain of PRC1 that is positively Zhu, C., Lau, E., Schwarzenbacher, R., Bossy-Wetzel, E., and Jiang, W. (2006).
charged and shown to enhance microtubule binding. Interest- Proc. Natl. Acad. Sci. USA 103, 6196–6201.

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  367


Leading Edge

Perspective

The Pioneer Round of Translation:


Features and Functions
Lynne E. Maquat,1,* Woan-Yuh Tarn,1,3 and Olaf Isken2,*
1
Department of Biochemistry and Biophysics and Center for RNA Biology, School of Medicine and Dentistry, University of Rochester,
Rochester, NY 14642, USA
2
Institute for Virology and Cell Biology, University of Lübeck, 23562 Lübeck, Germany
3
Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
*Correspondence: lynne_maquat@urmc.rochester.edu (L.E.M.), isken@molbio.uni-luebeck.de (O.I.)
DOI 10.1016/j.cell.2010.07.022

In mammalian cells, newly synthesized mRNAs undergo a pioneer round of translation that is
important for mRNA quality control. Following maturation of messenger ribonucleoprotein particles
during and after the pioneer round, steady-state cycles of mRNA translation generate most of the
cell’s proteins. Translation factors, RNA-binding proteins, and targets of signaling pathways that
are particular to newly synthesized mRNAs regulate critical functions of the pioneer round.

Introduction factors. These factors include not only PABPC1, which data indi-
In mammalian cells, newly synthesized transcripts are subject cate is important for activating the translation of both mRNPs,
to a series of nuclear processing steps to become mature tem- but also eIF4G, eIF3, eIF4B, eIF4A, eIF2 (Figures 1 and 2; Isken
plates for protein synthesis (Moore and Proudfoot, 2009). The and Maquat, 2008), and undoubtedly many other factors that
5′ ends of these transcripts acquire a 5′-m7GpppN cap struc- work in conjunction with ribosomes to synthesize proteins.
ture (where N is the first transcribed nucleotide) while being Although both mRNPs support protein synthesis and can be tar-
elongated by RNA polymerase II. The cap first binds to the geted for translational activation or repression, the purpose for
cap-binding protein (CBP) heterodimer CBP80-CBP20 (CBC), so doing is distinct: the translation of CBC-bound mRNAs pro-
which supports the pioneer round of mRNA translation (Isken vides a means to control the quality of gene expression; in con-
and Maquat, 2008). This round involves the loading of one or trast, the translation of eIF4E-bound mRNAs generates the bulk
more ribosomes, depending on the efficiency of translation ini- of cellular proteins (Isken and Maquat, 2008). Here, we discuss
tiation and the length of the open translational reading frame our growing understanding of how cells maintain the specialized
(Isken and Maquat, 2008; Isken et al., 2008). The cap subse- functions of each mRNP via associations with particular transla-
quently binds to the eukaryotic translation initiation factor 4E tion factors, RNA-binding proteins, and signaling targets.
(eIF4E), which directs steady-state rounds of mRNA translation
(Isken and Maquat, 2008). The Pioneer Round of Translation Supports Nonsense-
Although CBC-bound mRNAs are precursors to eIF4E- Mediated mRNA Decay
bound mRNAs, the two messenger ribonucleoprotein par- In higher eukaryotes, the vast majority of genes contain multi-
ticles (mRNPs) differ in significant ways (Figure 1). For exam- ple introns that are removed from the primary transcript by the
ple, spliced CBC-bound mRNAs differ from the eIF4E-bound process of splicing. Splicing may be accompanied by routinely
mRNAs that derive from them because they are associated made mistakes so as to result in mRNAs that contain a prema-
with one or more exon-junction complexes (EJCs) of proteins. ture termination codon (McGlincy and Smith, 2008). Premature
By the time eIF4E replaces CBC at the mRNA cap, EJCs are no termination codons can also arise during splicing as a con-
longer detectable, largely because most reside within the cod- sequence of a conditionally regulated process, as exemplified
ing region of mRNAs and therefore are displaced by translat- by those pre-mRNAs whose splice site usage is influenced by
ing ribosomes during the pioneer round (Gehring et al., 2009; the encoded RNA-binding protein (McGlincy and Smith, 2008).
Sato and Maquat, 2009). As another example, the poly(A) tails Nonsense-mediated mRNA decay is a translation-dependent
of CBC-bound mRNAs are associated with the mostly nuclear mRNA surveillance pathway that detects and eliminates tran-
but shuttling poly(A)-binding protein N1 (PABPN1) and the pri- scripts containing premature termination codons and thus
marily cytoplasmic but likewise shuttling PABPC1; in contrast, have the potential to be deleterious by virtue of the truncated
eIF4E-bound mRNAs do not detectably bind to PABPN1, the proteins they encode (see, e.g., Rebbapragada and Lykke-
replacement of which by PABPC1 is promoted by the pioneer Andersen, 2009).
round of translation (Sato and Maquat, 2009). During the pioneer round of translation, nonsense-mediated
Despite these and other differences (see below), both CBC- mRNA decay is thought to be triggered by the first ribosome
bound and eIF4E-bound mRNAs most likely engage in similar that translates newly processed CBC-bound mRNAs and
mechanisms of translation initiation, elongation, and termina- arrives at a premature termination codon (or a normal termi-
tion. Therefore, it is not surprising that both CBC-bound and nation codon) that is situated more than ?50–55 nucleotides
eIF4E-bound mRNAs use many of the same translation initiation upstream of an EJC-bearing exon-exon junction, although

368  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


Figure 1. Pioneer and Steady-State Translation Initiation Complexes
Shown is a CBP80-CBP20 (CBC)-bound mRNP from the pioneer round of translation and an eIF4E-bound mRNP from steady-state translation. CBC-bound
mRNAs direct pioneer rounds of translation, whereas eIF4E-bound mRNAs, which derive from the remodeling of CBC-bound mRNAs, support the bulk of cel-
lular protein synthesis. CBC-bound mRNAs are associated with at least one exon-junction complex (EJC) (provided they are the product of pre-mRNA splicing)
and the poly(A)-binding proteins PABPN1 and PABPC1. PYM, which interacts with EJC components and the small 40S ribosomal subunit (not shown), and
SKAR (S6 kinase 1 ALY-REF-like target), a component of EJCs, may help to activate the pioneer round of translation. eIF3e, a non-core subunit of the eukaryotic
translation initiation factor eIF3, may regulate the translation of a specific set of CBC-bound mRNAs. CTIF (CBP80-CBP20-dependent translation initiation fac-
tor) interacts directly with CBP80, as does eIF4G. It is currently unclear whether CTIF is the sole eIF4G-like molecule or if eIF4G also functions during pioneer
rounds. eIF4G has been proposed to form a complex with poly(A)-bound PABPC1 to circularize and promote the translation of CBC-bound mRNAs, similar to
how poly(A)-bound PABPC1 circularizes and promotes the translation of eIF4E-bound mRNAs. Importin (IMP)-β binds to IMP-α (which is a stable constituent of
cap-bound CBC) and augments the translation-independent replacement of CBC by eIF4E. In contrast, the pioneer round of translation promotes the removal
of EJCs and of associated RNA-binding proteins such as SF2/ASF (which also activates the pioneer round; not shown) and the replacement of PABPN1 by
PABPC1. The insets depict how the SURF complex (comprising the PIK-related protein kinase SMG1, UPF1, eRF1, and eRF3) assembles together with an
80S stalled ribosome at a premature termination codon of CBC-bound mRNA. SMG1 subsequently phosphorylates UPF1 upon UPF1 and SMG1 binding to a
downstream EJC during the process of nonsense-mediated mRNA decay. AUG, translation initiation codon; STOP, normal termination codon.

there are exceptions to this rule. The CBC plays a critical role and UPF1 join the EJC that resides downstream of the pre-
in nonsense-mediated mRNA decay not only because it com- mature termination codon. Notably, the interaction between
prises the mRNP that harbors EJCs but also because CBP80 CBP80 and UPF1 promotes nonsense-mediated mRNA decay
interacts directly with the nonsense-mediated mRNA decay at two sequential steps (Hwang et al., 2010). The first is the
factor, up-frameshift 1 (UPF1), enhancing the efficiency of this association of SMG1-UPF1 with eRF1-eRF3 at a premature
process (Isken and Maquat, 2008). In short, nonsense-medi- termination codon to form the SURF complex. The second is
ated mRNA decay is thought to involve recognition of prema- the association of SMG1-UPF1 with a downstream EJC, which
ture termination codons by the SURF complex, which consists results in SMG1-mediated UPF1 phosphorylation. Subse-
of the UPF1 phosphoinositide 3-kinase (PIK)-related protein quently, phosphorylated UPF1 elicits translational repression
kinase called SMG1, UPF1, and the two translation termina- by binding to the eIF3 constituent of the 43S preinitiation com-
tion factors referred to as eukaryotic release factor (eRF)1 and plex that is poised at the translation initiation codon (Isken et
eRF3 (Figure 1; Kashima et al., 2006; Isken and Maquat, 2008). al., 2008). Phosphorylated UPF1 also promotes the recruitment
After recognition of the premature termination codon, SMG1 of mRNA decay activities (Isken et al., 2008). The importance

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  369


Figure 2. Signaling Pathways and Translation
Shown are the signaling pathways that regulate the pioneer round of translation (top) and subsequent steady-state translation (bottom). Under condi-
tions that activate the mTOR signaling pathway, mTORC1 becomes activated and may bind to the translation initiation factor eIF3 associated with
CBP80-CBP20 (CBC)-bound mRNA resulting in the phosphorylation and dissociation of activated S6 kinase 1 (S6K1). The EJC component SKAR (S6
kinase 1 ALY-REF-like target) then recruits phosphorylated (i.e., activated) S6K1. Activated S6K1 next mediates the phosphorylation of SKAR and other
downstream effectors, including possibly eIF4B and PDCD4 (programmed cell death factor 4), that also associate with CBC-bound mRNA and thereby
promote the pioneer round of translation for spliced mRNAs. The Cdc42-dependent phosphorylation of S6K1 also may promote the binding of CBC to
cap structures, which would further stimulate the pioneer round. Another CBC-bound mRNA constituent, SF2/ASF, binds to the RNA export receptor TAP
and also triggers S6K1 signaling via mTORC1 in ways that promote CBC-bound mRNA translation. Moreover, PYM interacts with the EJC core proteins
Y14-MAGOH and with the 40S ribosomal subunit to somehow promote the translation of spliced mRNAs presumably during and certainly beyond the
pioneer round.
(Left) The boxes summarize the translational regulation of CBC-bound or eIF4E-bound mRNAs under various cell-growth conditions. Under conditions of heat
shock, hypoxia, or serum starvation, the pioneer round of translation is favored over steady-state translation; steady-state translation is largely inhibited by
phosphorylation of eIF2α (not shown) or by binding of 4E-BP1 to eIF4E.

of the CBC to nonsense-mediated mRNA decay is corrobo- Specialized Factors of the Pioneer Round of ­Translation
rated by the finding that decay is still restricted to CBC-bound The process of translation can be divided into four phases—
mRNAs in the case of mRNAs containing premature termina- initiation, elongation, termination, and ribosome recycling.
tion codons that initiate translation in a cap-independent mode The majority of regulation occurs during the initiation phase
from an internal ribosome entry site (IRES), provided that IRES (Sonenberg and Hinnebusch, 2009). There is considerably
function depends on eIF3 (Isken and Maquat, 2008). more known about the translation of eIF4E-bound mRNAs than
CBC-bound mRNAs form polysomes that are smaller than the translation of CBC-bound mRNAs, which is a relatively
the polysomes associated with their eIF4E-bound counterparts, recent discovery. Nevertheless, there are likely to be many
consistent with the replacement of CBC by eIF4E taking place on more similarities than differences between the translation of
polysomes during the pioneer round of translation. This finding, the two mRNPs, as available data have borne out. Factors
and data demonstrating that nonsense-mediated mRNA decay that specifically regulate the pioneer round of translation will
involves a step of translational repression that targets eIF3, indi- likely target, directly or via another protein, the CBC, the EJC,
cate that CBC-bound mRNAs, like eIF4E-bound mRNAs, are PABPN1, or other constituents that do not typify eIF4E-bound
generally loaded with more than one ribosome, eIF3, and other mRNA, just as factors that specifically regulate steady-state
well-characterized translation initiation factors. translation often target eIF4E.

370  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


PYM the association of eIF4E, eIF4A, and PABPC1 with mRNA. S6K1
Compared to nonspliced mRNA, it is well established that the phosphorylation results in the dissociation of activated S6K1 from
process of pre-mRNA splicing endows spliced mRNA with a mRNA-bound eIF3. Released S6K1 promotes scanning of the 43S
higher translational capacity during both pioneer and steady- preinitiation complex to the AUG translation initiation codon and,
state rounds of translation (Moore and Proudfoot, 2009). The abil- therefore, translation by phosphorylating both the eIF4A activator
ity of splicing to promote translation appears to be mediated at eIF4B and the eIF4A inhibitor and tumor suppressor programmed
least in part by EJCs, which data indicate are dynamic rather than cell death 4 (PDCD4).
either static or homogenous complexes. Different EJC constitu- In the case of CBC-bound mRNAs, activated mTORC1 may be
ents may augment different steps of translation initiation (Lee et recruited by eIF3 and, additionally, SF2/ASF (see below), promot-
al., 2009). For example, PYM may move between mRNA-bound ing the phosphorylation-induced activation of S6K1 and then the
EJCs and translationally active ribosomes: PYM can interact with dissociation of activated phosphorylated S6K1. EJC-bound SKAR
the EJC core proteins Y14-MAGOH and, via a separate domain, then recruits activated S6K1 to CBC-bound spliced mRNAs, and
with the 40S ribosomal subunit and the 48S preinitiation complex SKAR-bound S6K1 promotes the pioneer round of translation by
to somehow promote the translation of spliced mRNAs during allowing for the phosphorylation of SKAR and other downstream
and beyond the pioneer round (Diem et al., 2007). The finding that effectors that could include eIF4B and PDCD4, which also asso-
PYM detectably coimmunoprecipitates with CBP80 but not eIF4E ciate with CBC-bound mRNAs (Ma et al., 2008). The finding that
not only is consistent with the idea that PYM is present during, SKAR regulates cell growth (Richardson et al., 2004) raises the
and gone after, the pioneer round of translation but also raises interesting question of how the mTORC1-induced translation of
the possibility that PYM constitutes a fraction of ribosomes that all or a subset of CBC-bound spliced mRNAs is transduced to a
may specifically function during pioneer rounds. For example, by functionally significant increase in total-cell protein synthesis.
coupling ribosome recruitment to EJC disassembly, PYM may SF2/ASF
ensure the preferential recruitment of newly synthesized CBC- The serine-arginine-rich protein SF2/ASF plays a central role in
bound mRNAs over eIF4E-bound mRNAs to the translational recruiting the splicing machinery to pre-mRNA splice sites and
machinery. Alternatively, translation factors that are unique to can either enhance or inhibit splicing. Like PYM, SF2/ASF coim-
CBC-bound mRNAs and/or EJCs could recruit ribosomes bound munoprecipitates with CBP80 but not detectably with eIF4E
by PYM for the pioneer round. Along these lines, PYM is recruited (Sato et al., 2008). Data indicate that SF2/ASF, when bound to
to those newly exported mRNAs of Kaposi’s sarcoma-associated exonic sequences, can travel as a constituent of CBC-bound
herpesvirus that are intronless and thus devoid of EJCs, by the mRNPs from nuclei into the cytoplasm, where it has the ability
viral open reading frame 57 protein (ORF57). ORF57 interacts with to promote the pioneer round of translation (Sato et al., 2008).
PYM and enhances viral mRNA translation via the PYM-mediated Evidence indicates that SF2/ASF recruits a number of transla-
recruitment of preinitiation complexes (Boyne et al., 2010). Nota- tional activators to mRNAs, including the RNA export receptor
bly, the recent report that PYM is an EJC disassembly factor that TAP (Sato et al., 2008) and mTORC1 (Michlewski et al., 2008).
inhibits nonsense-mediated mRNA decay when overexpressed, Also, like PYM, SF2/ASF promotes mRNA translation beyond
i.e., when a fraction of PYM exists free of an association with 40S the pioneer round (Sato et al., 2008; Michlewski et al., 2008).
ribosomal subunits so as to promiscuously dissociate EJCs (Geh- CBP80
ring et al., 2009), is compatible with the idea that EJC constituents Another effector of the pioneer round could be CBP80 as the
can activate translation. binding of CBC to cap structures is stimulated by growth fac-
SKAR tors during the G1/S phase of the cell cycle, activated forms of
The EJC constituent called SKAR, for S6 kinase 1 (S6K1) ALY- the Ras proto-oncogene protein, and the Rho-GTPase Cdc42.
REF-like target, may also mark spliced CBC-bound mRNAs Cdc42 has been shown to transduce extracellular signals from
with a translational status that is distinct from the translational G-coupled protein receptors, integrins, and growth factor
status of eIF4E-bound mRNAs and provides another example receptors at least in part by binding to S6K1 in a GTP-depen-
of how EJCs positively regulate the pioneer round of transla- dent manner. Binding facilitates recognition of the Cdc42-
tion. The mammalian target of the rapamycin (mTOR) signaling S6K1 complex by upstream S6K-activating kinases (Chou and
pathway promotes cellular translation depending on the pres- Blenis, 1996; Wilson and Cerione, 2000). Moreover, signaling
ence of growth factors, the absence of stress, and the availabil- from Cdc42 to CBP80 is transmitted in part by the S6K-cata-
ity of nutrients or other energy sources (Ma and Blenis, 2009). lyzed phosphorylation of CBP80, although the biological con-
mTOR is a PIK-related protein kinase that, together with raptor sequence remains unknown (Ma and Blenis, 2009).
and LST8, forms the mTOR complex 1 (mTORC1). Interestingly, As exemplified above, it appears that an efficient pioneer
signaling through mTORC1 modulates the translation of both round of translation is followed by efficient steady-state
CBC-bound mRNAs and eIF4E-bound mRNAs by distinct but cycles of translation. Growth factors may augment cellu-
undoubtedly overlapping mechanisms (Figure 2). lar protein synthesis not only by promoting CBC binding to
In the case of eIF4E-bound mRNAs, the recruitment of acti- caps but also by expediting the replacement of CBC by eIF4E
vated mTORC1 by eIF3 into the proximity of the eIF4E-binding (see below). Despite not yet knowing mechanistic details, it is
protein 1 (4E-BP1) activates translation by mediating the phos- clear that activating or inhibiting pioneer rounds of translation
phorylation of 4E-BP1 and S6K1. 4E-BP1 phosphorylation results depending on cellular growth conditions is likely to provide
in 4E-BP1 dissociation from mRNA-bound eIF4E, which activates an important checkpoint mechanism for the bulk of cellular
translation by allowing eIF4G to interact with and thereby stabilize protein synthesis.

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  371


Importin-α bound mRNAs (Kim et al., 2009). CTIF could promote the recruit-
Regulated binding of CBC to mRNA caps is additionally influ- ment of ribosomes to newly synthesized CBC-bound mRNAs. As
enced by importin-α, which stably associates with cap-bound one possibility, CTIF, which contains a middle domain of eIF4G
CBC (Sato and Maquat, 2009). Cytoplasmic importin-β inter- (MIF4G) but lacks the eIF4E-binding domain of eIF4G, may func-
acts directly with importin-α and CBP20, thereby promoting the tionally replace eIF4G during pioneer rounds if, for example, it
replacement of CBC by eIF4E at mRNA caps (Figure 1; Dias et (possibly together with CBP80, which contains three MIF4Gs)
al., 2009; Sato and Maquat, 2009). In view of the recent structure serves as an eIF4G-like scaffold for CBC-bound mRNPs. Alter-
of an importin-α-CBC-cap complex and a structural model of natively, CTIF may function in addition to eIF4G considering that
the importin-β-importin-α-CBC complex (Dias et al., 2009), it will CTIF lacks the PABPC1- and eIF4A-binding domains that typify
be possible to study how signaling pathways, and the resulting eIF4G and that another MIF4G-containing protein, called SLIP1,
posttranslational modifications of importin-α, importin-β, and/ interacts directly with eIF4G to promote the translation of histone
or CBC, might regulate importin-α or importin-β binding to cap- mRNA (Cakmakci et al., 2008). Moreover, data indicate that eIF4G
associated CBC to either stabilize or disrupt the interaction of functions during pioneer rounds of translation. Regardless, as is
CBC with mRNA caps. When considering the critical role of CBC possible for eIF3, the use of translation initiation factors that are
in assuring the quality of gene expression through nonsense- unique to and activate pioneer rounds could be advantageous by
mediated mRNA decay, and that CBC can be replaced by eIF4E shortening the time span between transcriptional activation and
even when the pioneer round of translation is inhibited (Sato and the start of protein production.
Maquat, 2009), it is important that timing the pioneer round rela-
tive to the replacement of CBC by eIF4E be coordinated so that The Pioneer Round during Cell Stress
the pioneer round of translation largely takes place first. The bulk of cellular cap-dependent translation is rapidly downreg-
eIF3e ulated by signaling pathways in response to most physiological
Mammalian eIF3, which is a large protein complex consisting of and environmental stressors as part of a repertoire of events that
?13 subunits, has multiple functions in stimulating translation mitigate cellular damage and promote cell survival (Figure 2). This
initiation (Sonenberg and Hinnebusch, 2009). The eIF3 subunit global downregulation during, for example, viral infection, amino
eIF3e (also called p48/INT6) coimmunoprecipitates with CBP80 acid starvation, hypoxia, or heat shock is often accompanied by
as well as the EJC constituent and nonsense-mediated mRNA eIF2α phosphorylation. eIF2α phosphorylation decreases the
decay factor UPF2, but it does not detectably coimmunoprecipi- translation of the majority of CBC-bound as well as eIF4E-bound
tate with eIF4E and, like all eIF3 subunits that have been tested, mRNAs by limiting ternary complex abundance while mediating
is essential for nonsense-mediated mRNA decay (Morris et al., the selective translation of a subset of cellular mRNAs that ini-
2007; Isken et al., 2008). eIF3e, which is a core subunit of mam- tiate translation in a mechanism that depends on an upstream
malian eIF3 that is not present in all species, appears to contribute open reading frame. These mRNAs encode specific transcription
to the recruitment of ribosome-bound eIF3 to mRNA by directly factors that help the cell adapt to stress and ultimately restore
associating with eIF4G (Sonenberg and Hinnebusch, 2009). How- translation (Holcik and Sonenberg, 2005). In addition to eIF2α
ever, at least in fission yeast, eIF3e is not critical for global pro- phosphorylation, stress-induced proteolysis of translation initia-
tein synthesis, and there exist distinct eIF3 complexes that differ tion factors can also compromise cellular translation.
in their non-core subunits. Interestingly, those complexes that There are variations to this theme that result in the differen-
contain eIF3e associate with a restricted set of mRNAs, some of tial regulation of pioneer rounds of translation relative to sub-
which are implicated in the response to cell stress (Zhou et al., sequent rounds of translation. For example, the preferential
2005). Therefore, it is possible that a specialized eIF3e-containing translation of CBC-bound mRNAs is maintained during serum
eIF3 complex, if present in mammals, could specifically function starvation and heat shock, when the translation of eIF4E-bound
during pioneer rounds of translation by recruiting a defined set of mRNAs is compromised (Oh et al., 2007b; Marín-Vinader et al.,
newly synthesized mRNAs to the translational machinery. By so 2006). Serum starvation does not result in eIF2α phosphoryla-
doing, this specialized complex could route those mRNAs that tion but instead activates 4E-BP1.
are targets of nonsense-mediated mRNA decay to this pathway As another example, during early phases of hypoxia, activa-
and activate the steady-state translation of those that are not non- tion of the endoplasmic reticulum kinase PERK leads to eIF2α
sense-mediated mRNA decay targets. phosphorylation and the translational inhibition of both CBC-
CTIF bound and eIF4E-bound mRNAs (Oh et al., 2007a). However,
Another initiation factor that is central to steady-state transla- during late stages of hypoxia, eIF2α dephosphorylation allows
tion is eIF4G, which binds directly to cap-associated eIF4E and for the resumption of CBC-bound mRNA translation (Oh et al.,
serves critical roles as a scaffold for eIF3, PABPC1, and eIF4A. 2007a). Concomitantly, in an eIF2α-independent pathway, dis-
eIF4G, which also binds directly to CBC, is thought to function in ruption of eIF4E-eIF4G-eIF4A and sequestration of eIF4E by
a similar fashion during pioneer rounds of translation. Recently, dephosphorylated 4E-BP1 and the eIF4E-transporter (which
an eIF4G-like protein called CBP80-CBP20-dependent transla- moves eIF4E into the nucleus and processing bodies) maintain
tion initiation factor (CTIF) has been implicated specifically during the repression of eIF4E-bound mRNA translation (Koritzinsky
pioneer rounds. CTIF interacts directly with CBP80 as well as pre- et al., 2006; Lee et al., 2008). Conceivably, the restoration of
mRNAs, coimmunoprecipitates with eIF3 in an RNase-resistant CBC-bound mRNA translation as a first step toward transla-
manner, and functions in the translation of CBC-bound mRNAs; tional normalcy promotes cell survival after stress by allowing
however, it plays no detectable role in the translation of eIF4E- for the surveillance of newly synthesized mRNAs. Furthermore,

372  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


the renewed engagement of newly made mRNAs with the Features of the pioneer translation initiation complex also
translational machinery could speed up the cellular response appear to be important for proper localization of oskar mRNA
after stress. to the posterior pole in Drosophila oocytes. For example, splic-
ing of the first intron of oskar pre-mRNA (or another intron at
Spatial Regulation of the Pioneer Round of Translation the site of the first intron) is required for oskar mRNA localiza-
Proper embryonic patterning and development as well as neu- tion, presumably by virtue of the resulting EJC in conjunction with
ronal function often involve proteins that bind to the 3′ untrans- nearby mRNA sequences (Hatchet and Ephrussi, 2004). In fact,
lated regions (3′ UTRs) of particular mRNAs in a sequence-spe- demonstrated roles for the EJC constituents eIF4AIII, Tsunami-
cific manner to couple mRNA translational activation and mRNA Magonashi, and Barentz (the latter two of which are orthologous
localization (Sonenberg and Hinnebusch, 2009). By so doing, an to mammalian Y14-MAGOH and MLN51, respectively) in oskar
expression gradient of the encoded morphogens, in the case of mRNA localization and their colocalization with oskar mRNA at
oocytes, or of proteins that maintain synaptic function or plastic- the posterior pole (Besse and Ephrussi, 2008) indicate that the
ity, in the case of neurons, is spatially established. pioneer round of translation and removal of the first EJC occur
There have been many reports of translational repression after the regulatory steps of translational repression and local-
occurring prior to mRNA localization so as to inhibit ectopic ization. As noted above, these regulatory steps may involve the
protein production from unlocalized mRNAs. If the sole mecha- RNA-binding protein Bruno. As oskar mRNA is also translation-
nism of translational repression targets eIF4E, then an mRNA ally repressed by Bruno-mediated recruitment of the Cup protein,
should have undergone the pioneer round of translation prior which targets eIF4E (Besse and Ephrussi, 2008), there appear
to repression although, as noted above, it is possible for eIF4E to be multiple pathways to translationally silence oskar mRNA.
to replace CBC without a pioneer round. In theory, other mech- In fact, a number of mRNAs are repressed at different steps of
anisms of repression might target both CBC-bound mRNA and translation when bound by CBC or eIF4E.
eIF4E-bound mRNA.
Examples of translational repression include sequestrating MicroRNAs
mRNAs into translationally silenced particles, as Bruno does It was recently reported that the microRNA (miRNA) miR-2 medi-
for oskar mRNA in Drosophila; recruiting the CCR4 deadeny- ates translational repression, which interferes with the interac-
lase to shorten poly(A) tail length, as Bicaudal-C does for its tion between eIF4E and eIF4G in a way that is predicted to have
own mRNA and certain other germline mRNAs in Drosophila; no consequence to the pioneer round of translation (Zdanow-
and blocked joining of the 60S ribosomal subunit to the 48S icz et al., 2009). Therefore, mRNA caps bound by eIF4E could
preinitiation complex, as Zipcode-binding protein 1 (ZBP1, represent another signature target used by cells to differentially
also called IMP1) does for β-actin mRNA at the leading lamel- regulate the translation of CBC-bound mRNA relative to eIF4E-
lipodia of fibroblasts or neurite growth cones (Besse and bound mRNA. Nevertheless, other mechanisms of miRNA-
Ephrussi, 2008; Sonenberg and Hinnebusch, 2009). Notably, mediated translational repression can function simultaneously
the findings that ZBP1 associates cotranscriptionally with to repress the translation of CBC-bound mRNA. This became
β-actin mRNA (Besse and Ephrussi, 2008), and RNP granules evident through the recent demonstration that (1) Ago2, which
undergoing ZBP1-mediated transport contain CBP80 and EJC is known to inhibit the translation of its target mRNAs, is loaded
constituents but lack detectable eIF4E (Jønson et al., 2007), onto both CBC-bound mRNAs and eIF4E-bound mRNAs, (2)
suggest that ZBP1-mediated translational repression targets the loading of endogenous microRNA-induced silencing com-
CBC-bound transcripts. plexes (miRISCs) onto the 3′ UTRs of CBC-bound mRNAs that
To date, studies of arc mRNA and other specific mRNAs in contain a premature termination codon abrogates nonsense-
mammalian neurons provide the best examples of translational mediated mRNA decay, and (3) several natural substrates of
repression that targets CBC-bound mRNAs (Giorgi et al., 2007). nonsense-mediated mRNA decay are stabilized by miRISC-
arc mRNA, which harbors two 3′ UTR introns, is a natural tar- mediated translational repression (Choe et al., 2010). It will be
get of nonsense-mediated mRNA decay: When the pioneer interesting to determine if factors that are specific for CBC-
round of translation terminates normally, arc mRNA undergoes bound mRNA and the pioneer round of translation are recog-
nonsense-mediated mRNA decay. The arc gene is transcrip- nized during miRNA-mediated translational repression.
tionally induced upon intense synaptic activation, and the
resulting mRNA is translated and thus targeted for nonsense- Future Directions
mediated mRNA decay, once it localizes to activated synapses Mammalian cells have evolved two mRNP templates for protein
in a mechanism that depends on glutamatergic receptors. By synthesis that manifest a precursor-product relationship and
essentially limiting ARC protein synthesis to the pioneer round serve distinct functions. Although these two templates share
of translation at activated synapses, improper protein synthe- many constituent proteins that homogenize their regulation, at
sis at inopportune times and cellular locations is prevented. least some unique features impart a means for their differential
These findings are consistent with data indicating that CBP80- control. Examples also exist of distinct regulatory mechanisms
bound mRNAs migrate to spines when glutamatergic recep- that coordinate CBC-bound mRNA translation and eIF4E-bound
tors of rat dendrites are stimulated (di Penta et al., 2009) and mRNA translation by targeting factors that are particular to each
thus have yet to undergo the pioneer round of translation. At mRNP. As eIF4E-bound mRNA supports the bulk of cellular protein
present, which signaling factors control these pioneer rounds synthesis, it is the logical target for a rapid response to changes
of translation in neurons remain unknown. in physiological conditions. However, CBC-bound mRNA holds

Cell 142, August 6, 2010 ©2010 Elsevier Inc.  373


the unique capability of enhancing subsequent rounds of transla- eRF1-eRF3 complex (SURF) to the exon junction complex triggers Upf1 phos-
phorylation and nonsense-mediated mRNA decay. Genes Dev. 20, 355–367.
tion, downregulating further translation, or completely stopping
not only CBC-supported mRNA surveillance but also eIF4E- Kim, K.M., Cho, H., Choi, K., Kim, J., Kim, B.W., Ko, Y.G., Jang, S.K., and Kim,
supported protein production. Future studies are expected to Y.K. (2009). A new MIF4G domain-containing protein, CTIF, directs nuclear cap-
binding protein CBP80/20-dependent translation. Genes Dev. 23, 2033–2045.
illuminate molecular aspects of known and unforeseen regulatory
mechanisms. Koritzinsky, M., Magagnin, M.G., van den Beucken, T., Seigneuric, R., Savelkouls,
K., Dostie, J., Pyronnet, S., Kaufman, R.J., Weppler, S.A., Voncken, J.W., et al.
(2006). Gene expression during acute and prolonged hypoxia is regulated by dis-
Acknowledgments tinct mechanisms of translational control. EMBO J. 25, 1114–1125.

We thank Chenguang Gong and Hanae Sato for helpful comments and ac- Lee, H.C., Cho, H., and Kim, Y.K. (2008). Ectopic expression of eIF4E-transporter
knowledge NIH R01 grants GM074593 and GM59614 (L.E.M.), the National triggers the movement of eIF4E into P-bodies, inhibiting steady-state translation
Science Council of Taiwan (W.-Y.T.), and start-up funds from the University of but not the pioneer round of translation. Biochem. Biophys. Res. Commun. 369,
1160–1165.
Lübeck (O.I.) for support.
Lee, H.C., Choe, J., Chi, S.G., and Kim, Y.K. (2009). Exon junction complex en-
References hances translation of spliced mRNAs at multiple steps. Biochem. Biophys. Res.
Commun. 384, 334–340.
Besse, F., and Ephrussi, A. (2008). Translational control of localized mRNAs: re- Ma, X.M., and Blenis, J. (2009). Molecular mechanisms of mTOR-mediated trans-
stricting protein sythesis in space and time. Nat. Rev. Mol. Cell Biol. 9, 971–980. lational control. Nat. Rev. Mol. Cell Biol. 10, 307–318.
Boyne, J.R., Jackson, B.R., Taylor, A., Macnab, S.A., and Whitehouse, A. (2010). Ma, X.M., Yoon, S.O., Richardson, C.J., Jülich, K., and Blenis, J. (2008). SKAR
Kaposi’s sarcoma-associated herpesvirus ORF57 protein interacts with PYM to links pre-mRNA splicing to mTOR/S6K1-mediated enhanced translation efficien-
enhance translation of viral intronless mRNAs. EMBO J. 29, 1851–1864. cy of spliced mRNAs. Cell 133, 303–313.
Cakmakci, N.G., Lerner, R.S., Wagner, E.J., Zheng, L., and Marzluff, W.F. (2008). Marín-Vinader, L., van Genesen, S.T., and Lubsen, N.H. (2006). mRNA made dur-
SLIP1, a factor required for activation of histone mRNA translation by the stem- ing heat shock enters the first round of translation. Biochim. Biophys. Acta 1759,
loop binding protein. Mol. Cell. Biol. 28, 1182–1194. 535–542.
Choe, J., Cho, H., Lee, H.C., and Kim, Y.K. (2010). microRNA/Argonaute 2 regu- McGlincy, N.J., and Smith, C.W. (2008). Alternative splicing resulting in nonsense-
lates nonsense-mediated messenger RNA decay. EMBO Rep. 11, 380–386. mediated mRNA decay: what is the meaning of nonsense? Trends Biochem. Sci.
3, 385–393.
Chou, M.M., and Blenis, J. (1996). The 70S kDa S6 kinase complexes with and is
activated by the Rho family of G proteins Cdc42 and Rac1. Cell 85, 573–583. Michlewski, G., Sanford, J.R., and Cáceres, J.F. (2008). The splicing factor SF2/
ASF regulates translation initiation by enhancing phosphorylation of 4E-BP1. Mol.
Dias, S.M., Wilson, K.F., Rojas, K.S., Ambrosio, A.L., and Cerione, R.A. (2009). Cell 30, 179–189.
The molecular basis for the regulation of the cap-binding complex by the import-
ins. Nat. Struct. Mol. Biol. 16, 930–937. Moore, M.J., and Proudfoot, N.J. (2009). Pre-mRNA processing reaches back to
transcription and ahead to translation. Cell 136, 688–700.
di Penta, A., Mercaldo, V., Florenzano, F., Munck, S., Ciotti, M.T., Zalfa, F., Mer-
canti, D., Molinari, M., Bagni, C., and Achsel, T. (2009). Dendritic LSm1/CBP80- Morris, C., Wittmann, J., Jäck, M., and Jalinot, P. (2007). Human INT6/eIF3e is
mRNPs mark the early steps of transport commitment and translational control. required for nonsense-mediated mRNA decay. EMBO Rep. 8, 596–602.
J. Cell Biol. 184, 423–435.
Oh, N., Kim, K.M., Choe, J., and Kim, Y.K. (2007a). Pioneer round of translation
Diem, M.D., Chan, C.C., Younis, I., and Dreyfuss, G. (2007). PYM binds the cyto- mediated by nuclear cap-binding proteins CBP80/20 occurs during prolonged
plasmic exon-junction complex and ribosomes to enhance translation of spliced hypoxia. FEBS Lett. 581, 5158–5164.
mRNAs. Nat. Struct. Mol. Biol. 14, 1173–1179.
Oh, N., Kim, K.M., Choe, J., and Kim, Y.K. (2007b). Pioneer round of transla-
Gehring, N.H., Lamprinaki, S., Kulozik, A.E., and Hentze, M.W. (2009). Disassem- tion occurs during serum starvation. Biochem. Biophys. Res. Commun. 362,
bly of exon junction complexes by PYM. Cell 137, 536–548. 145–151.
Giorgi, C., Yeo, G.W., Stone, M.E., Katz, D.B., Burge, C., Turrigiano, G., and Rebbapragada, I., and Lykke-Andersen, J. (2009). Execution of nonsense-medi-
Moore, M.J. (2007). The EJC factor eIF4AIII modulates synaptic strength and ated mRNA decay: what defines a substrate? Curr. Opin. Cell Biol. 21, 394–402.
neuronal protein expression. Cell 130, 179–191.
Richardson, C.J., Bröenstrup, M., Fingar, D.C., Jülich, K., Ballif, B.A., Gygi, S.,
Hatchet, O., and Ephrussi, A. (2004). Splicing of oskar RNA in the nucleus is and Blenis, J. (2004). SKAR is a specific target of S6 kinase 1 in cell growth con-
coupled to its cytoplasmic localization. Nature 428, 959–963. trol. Curr. Biol. 14, 1540–1549.

Holcik, M., and Sonenberg, N. (2005). Translational control in stress and apopto- Sato, H., and Maquat, L.E. (2009). Remodeling of the pioneer translation initia-
sis. Nat. Rev. Mol. Cell Biol. 6, 318–327. tion complex involves translation and the karyopherin importin β. Genes Dev.
23, 2537–2550.
Hwang, J., Sato, H., Tang, Y., Matsuda, D., and Maquat, L.E. (2010). UPF1 as-
sociation with the cap-binding protein, CBP80, promotes nonsense-mediated Sato, H., Hosoda, N., and Maquat, L.E. (2008). Efficiency of the pioneer round
mRNA decay at two distinct steps. Mol. Cell. Published online August 5, 2010. of translation affects the cellular site of nonsense-mediated mRNA decay. Mol.
10.1016/j.molcel.2010.07.004. Cell 29, 255–262.

Isken, O., and Maquat, L.E. (2008). The multiple lives of NMD factors: balancing Sonenberg, N., and Hinnebusch, A.G. (2009). Regulation of translation initiation in
roles in gene and genome regulation. Nat. Rev. Genet. 9, 699–712. eukaryotes: Mechanisms and biological targets. Cell 136, 731–745.

Isken, O., Kim, Y.K., Hosoda, N., Mayeur, G.L., Hershey, J.W., and Maquat, L.E. Wilson, K.F., and Cerione, R.A. (2000). Signal transduction and post-transcrip-
(2008). Upf1 phosphorylation triggers translational repression during nonsense- tional gene expression. Biol. Chem. 381, 357–365.
mediated mRNA decay. Cell 133, 314–327.
Zdanowicz, A., Thermann, R., Kowalska, J., Jemielity, J., Duncan, K., Preiss, T.,
Jønson, L., Vikesaa, J., Krogh, A., Nielsen, L.K., Hansen, T.O., Borup, R., Johns- Darzynkiewicz, E., and Hentze, M.W. (2009). Drosophila miR2 primarily targets
en, A.H., Christiansen, J., and Nielsen, F.C. (2007). Molecular composition of the m7GpppN cap structure for translational repression. Mol. Cell 35, 881–888.
IMP1 ribonucleoprotein granules. Mol. Cell. Proteomics 6, 798–811.
Zhou, C., Arslan, F., Wee, S., Krishnan, S., Ivanov, A.R., Oliva, A., Leatherwood,
Kashima, I., Yamashita, A., Izumi, N., Kataoka, N., Morishita, R., Hoshino, S., J., and Wolf, D.A. (2005). PCI proteins eIF3e and eIF3m define distinct translation
Ohno, M., Dreyfuss, G., and Ohno, S. (2006). Binding of a novel SMG-1-Upf1- initiation factor 3 complexes. BMC Biol. 3, 14.

374  Cell 142, August 6, 2010 ©2010 Elsevier Inc.


Direct Reprogramming of Fibroblasts
into Functional Cardiomyocytes
by Defined Factors
Masaki Ieda,1,2,3,6,* Ji-Dong Fu,1,2,3 Paul Delgado-Olguin,1,2,4 Vasanth Vedantham,1,5 Yohei Hayashi,1
Benoit G. Bruneau,1,2,4 and Deepak Srivastava1,2,3,*
1Gladstone Institute of Cardiovascular Disease
2Department of Pediatrics
3Department of Biochemistry and Biophysics
4Cardiovascular Research Institute
5Department of Medicine

University of California, San Francisco, San Francisco, CA 94158, USA


6Present address: Departments of Cardiology and of Clinical and Molecular Cardiovascular Research, Keio University School of Medicine,

Shinanomachi 35, Shinjuku-ku, Tokyo 160-8582, Japan


*Correspondence: ieda@cpnet.med.keio.ac.jp (M.I.), dsrivastava@gladstone.ucsf.edu (D.S.)
DOI 10.1016/j.cell.2010.07.002

SUMMARY malformations. Because postnatal cardiomyocytes have little


or no regenerative capacity, current therapeutic approaches
The reprogramming of fibroblasts to induced plurip- are limited. Embryonic stem cells possess clear cardiogenic
otent stem cells (iPSCs) raises the possibility that potential, but efficiency of cardiac differentiation, risk of tumor
a somatic cell could be reprogrammed to an alterna- formation, and issues of cellular rejection must be overcome
tive differentiated fate without first becoming a stem/ (Ivey and Srivastava, 2006; Laflamme et al., 2007; Nussbaum
progenitor cell. A large pool of fibroblasts exists in et al., 2007; van Laake et al., 2008). The ability to reprogram
fibroblasts into induced pluripotent stem cells (iPSCs) with four
the postnatal heart, yet no single ‘‘master regulator’’
defined factors might address some of these issues by providing
of direct cardiac reprogramming has been identified. an alternative source of embryonic-like stem cells (Takahashi
Here, we report that a combination of three develop- and Yamanaka, 2006). However, generating sufficient iPSC-
mental transcription factors (i.e., Gata4, Mef2c, and derived cardiomyocytes that are pure and mature and that can
Tbx5) rapidly and efficiently reprogrammed post- be delivered safely remains challenging (Zhang et al., 2009).
natal cardiac or dermal fibroblasts directly into differ- The human heart is composed of cardiomyocytes, vascular
entiated cardiomyocyte-like cells. Induced cardio- cells, and cardiac fibroblasts. In fact, cardiac fibroblasts com-
myocytes expressed cardiac-specific markers, had prise over 50% of all the cells in the heart (Baudino et al.,
a global gene expression profile similar to cardio- 2006; Camelliti et al., 2005; Snider et al., 2009). Cardiac fibro-
myocytes, and contracted spontaneously. Fibro- blasts are fully differentiated somatic cells that provide support
blasts transplanted into mouse hearts one day after structure, secrete signals, and contribute to scar formation
upon cardiac damage (Ieda et al., 2009). Fibroblasts arise from
transduction of the three factors also differentiated
an extracardiac source of cells known as the proepicardium,
into cardiomyocyte-like cells. We believe these find- and do not normally have cardiogenic potential (Snider et al.,
ings demonstrate that functional cardiomyocytes 2009). The large population of endogenous cardiac fibroblasts
can be directly reprogrammed from differentiated is a potential source of cardiomyocytes for regenerative therapy
somatic cells by defined factors. Reprogramming if it were possible to directly reprogram the resident fibroblasts
of endogenous or explanted fibroblasts might pro- into beating cardiomyocytes. Unfortunately, although embryonic
vide a source of cardiomyocytes for regenerative mesoderm can be induced to differentiate into cardiomyocytes
approaches. (Takeuchi and Bruneau, 2009), efforts to accomplish this in
somatic cells have thus far been unsuccessful, and to our knowl-
edge, no ‘‘master regulator’’ of cardiac differentiation, like MyoD
INTRODUCTION for skeletal muscle (Davis et al., 1987), has been identified
to date.
Heart disease is a leading cause of adult and childhood mortality. The generation of iPSCs suggests that a specific combination
The underlying pathology is typically loss of cardiomyocytes that of defined factors, rather than a single factor, could epigeneti-
leads to heart failure or improper development of cardiomyo- cally alter the global gene expression of a cell and allow greater
cytes during embryogenesis that leads to congenital heart plasticity of cell type than previously appreciated. Consistent

Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc. 375


A C Control αMHC-GFP explant Figure 1. Screening for Cardiomyocyte-
Off On
FACS 10 5 0% 0%10 5
68% 0% Inducing Factors
αMHC promoter GFP αMHC promoter GFP
10 4 10
4
(A) Schematic representation of the strategy to test
TFs 10 3 10
3 candidate cardiomyocyte-inducing factors.

Thy1
10 2 10 2
(B) Morphology and characterization of fibroblast-
Cardiac fibroblasts Cardiomyocytes 0
100% 0%
0
31.4% 0.6% like cells migrating from aMHC-GFP heart
2 3 4 5
0 102 103 10 4 10 5
0 10 10 10 10
explants. Phase contrast (left), GFP (middle), and
αMHC-GFP
B D Thy-1 immunostaining (right). Insets are high-
6 magnification views. See also Figure S1.

Explant
*

αMHC-GFP+
5 (C) Thy-1+/GFP cells were FACS sorted from

cells (%)
4
3 ** explant cultures for reprogramming.
2 (D) Summary of FACS analyses for a-MHC-GFP+
1 * cells. Effect on GFP+ cell induction with 14 factors
0

Control

Baf60c
Gata4
Hand2
Hopx
Hrt2
Isl1
Mef2c
Mesp1
Myocd
Nkx2.5
Pitx2c
Smyd1

Tbx5
14 factors

Srf
Phase αMHC-GFP Thy1 or the removal of individual factors from the pool of
14 factors (n = 3). Removal of Baf60c, Hand2,
14 factors - 1 Hopx, Hrt2, or Pitx2c did not decrease the percent
E of GFP+ cells and were excluded for further anal-
10 4 10
4
10 4 10 4
Control 14 factors 14 factors - Gata4 14 factors - Pitx2c yses. See also Figure S2.
10
3
10
3
10 3 10
3
(E) FACS plots for analyses of GFP+ cells. GFP+
100% 97.3% 98.9% 93.4% cells were analyzed 1 week after 14 factor trans-
10 2 10 2 10 2 10 2
duction. The number of GFP+ cells were reduced
10
1 0% 10 1
1.7% 10 1
0.5% 10
1 5.1% by removal of Gata4, but increased by removal of
PI

Pitx2c from 14 factors.


10
0
10
0
10
0
10
0
(F–H) Effect on GFP+ cell induction of the removal
100 10 1 10 2 10 3 10 4 10 0 10 1 10 2 10 3 10 4 10
0
10 1 10 2 10 3 10 4 100 10 1 10 2 10 3 10 4
of individual factors from the pool of 9 (F), 6 (G), or 5
αMHC-GFP
(H) factors (n = 3). Factors that did not decrease
F G H efficiency upon removal were excluded from
20
* 20 20 **
** further study.
αMHC-GFP+

αMHC-GFP+

αMHC-GFP+

15 ** 15 15
cells (%)

cells (%)

cells (%)

(I) GFP+ (20%) cells were induced from fibroblasts


10 **** * 10 * * * 10
*
* * by the combination of four factors, Gata4, Mef2c,
5
* 5 * * 5 Mesp1, and Tbx5. Representative data are shown
0 0 0 in each panel. PI, propidium iodine. All data are pre-
Control
9 factors
6 factors
Gata4
Mef2c
Mesp1
Myocd
Nkx2.5
Tbx5
Control

9 factors
Gata4
Isl1
Mef2c
Mesp1
Myocd
Nkx2.5
Smyd1

Tbx5
14 factors

Srf

Control
6 factors
5 factors
Gata4
Mef2c
Mesp1
Myocd
Tbx5

sented as means ± SD. *p < 0.01; **p < 0.05 versus


relevant control. Scale bars represent 100 mm.
9 factors - 1 6 factors - 1 5 factors - 1
See also Figures S1 and S2.
I 10 4 10
4

Control Gata4/Mef2c/Mesp1
10
3
10
3
/Tbx5

10 2
100% 76.5%
10 2
RESULTS
10
1 0% 10
1 20.6%
PI

10
0
10
0
Screening for Cardiomyocyte-
10 0 10 1 10 2 10 3 10 4 100 101 10 2 10 3 10 4
Inducing Factors
αMHC-GFP
We developed an assay system in
which the induction of mature cardiomyo-
with this, the bHLH transcription factor, Neurogenin 3, in combi- cytes from fibroblasts could be analyzed quantitatively by
nation with Pdx1 and Mafa, can efficiently reprogram pancreatic reporter-based fluorescence-activated cell sorting (FACS) (Fig-
exocrine cells into functional b cells in vivo, although the exocrine ure 1A). To accomplish this, we generated aMHC promoter-
cells were known to have some potential to become islet cells driven EGFP-IRES-puromycin transgenic mice (aMHC-GFP), in
in vitro and share a common parent cell with islet cells (Baeyens which only mature cardiomyocytes expressed the green fluores-
et al., 2005; Zhou et al., 2008). A combination of three factors, cent protein (GFP) (Gulick et al., 1991). We confirmed that only
Ascl1, Brn2, and Myt1l, converts dermal fibroblasts to functional cardiomyocytes, but not other cell types such as cardiac fibro-
neurons (Vierbuchen et al., 2010), although the degree of global blasts, expressed GFP in the transgenic mouse hearts and in
reprogramming of the neurons is unknown. primary cultured neonatal mouse cardiac cells (Figure S1 avail-
In this study, we examined whether key developmental able online).
cardiac regulators could reprogram cardiac fibroblasts into car- To have enough cardiac fibroblasts for FACS screening, we
diomyocytes. We found that out of a total of 14 factors, a specific obtained GFP cardiac fibroblasts from neonatal aMHC-GFP
combination of three transcription factors, Gata4, Mef2c, and hearts by explant culture. Fibroblast-like cells migrated from
Tbx5, was sufficient to generate functional beating cardiomyo- the explants after 2 days and were confluent after 1 week. The
cytes directly from mouse postnatal cardiac or dermal fibro- migrating cells did not express GFP, but expressed Thy1 and
blasts and that the induced cardiomyocytes (iCMs) were globally vimentin, markers of cardiac fibroblasts (Figure 1B and data
reprogrammed to adopt a cardiomyocyte-like gene expression not shown) (Hudon-David et al., 2007; Ieda et al., 2009). To avoid
profile. contamination of cardiomyocytes, we filtered the cells by cell

376 Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc.


strainers to remove heart tissue fragments and isolated Thy1+/ GFP+ cells, but cTnT expression was abolished, suggesting
GFP cells by FACS (Figure 1C). Using FACS, we confirmed that Gata4 was also required. Whereas the combination of two
Thy1+/GFP cells did not express cardiac troponin T (cTnT), factors, Mef2c and Tbx5, induced GFP expression but not
a specific sarcomeric marker of differentiated mature cardio- cTnT, no combination of two factors or single factor induced
myocytes (Figure S1) (Kattman et al., 2006). With these proce- both GFP and cTnT expression in cardiac fibroblasts (Figure 2C).
dures, we had no cardiomyocyte contamination in the fibroblast These data suggested that the combination of three factors,
culture and could generate greater than twice the number of Gata4, Mef2c, and Tbx5, is sufficient to induce cardiac gene
cardiac fibroblasts than by conventional fibroblast isolation tech- expression in fibroblasts.
niques (Ieda et al., 2009). We found that 30% of GFP+ cells expressed cTnT 1 week after
To select potential cardiac reprogramming factors, we used the three-factor transduction. Next, to confirm our screening
microarray analyses to identify transcription factors and epige- results, we transduced cardiac fibroblasts with three factors
netic remodeling factors with greater expression in mouse (Gata4, Mef2c, and Tbx5, hereafter referred to as GMT) plus
cardiomyocytes than in cardiac fibroblasts at embryonic day Nkx2-5, a critical factor for cardiogenesis but excluded by our
12.5 (Ieda et al., 2009). Among them, we selected 13 factors initial screening. Surprisingly, adding Nkx2-5 to GMT dramati-
that exhibited severe developmental cardiac defects and cally inhibited the expression of GFP and cTnT in cardiac
embryonic lethality when mutated (Figure S2). We also included fibroblasts. We also transduced cardiac fibroblasts with the
the cardiovascular mesoderm-specific transcription factor combination of Baf60c, Gata4, and Tbx5, which can transdiffer-
Mesp1 because of its cardiac transdifferentiation effect in entiate noncardiogenic mesoderm to cardiomyocytes in mouse
Xenopus (David et al., 2008). We generated individual retrovi- embryos (Takeuchi and Bruneau, 2009). We found that this
ruses to efficiently express each gene in cardiac fibroblasts combination did not efficiently induce cTnT or GFP expression
(Figure S2). above that of Tbx5 alone, confirming our screening results
We transduced Thy1+/GFP neonatal mouse cardiac fibro- (Figure 2D).
blasts with a mixture of retroviruses expressing all 14 factors To determine if other cardiac genes were enriched in GFP+
or with DsRed retrovirus (negative control) (Hong et al., 2009). cells, we sorted GFP+ cells and GFP cells 7 days after transduc-
We did not observe any GFP+ cells in cardiac fibroblasts 1 week tion with GMT and compared gene expression of cardiomyo-
after Ds-Red retrovirus infection or 1 week of culture without any cyte-specific genes, Myh6 (a-myosin heavy chain), Actc1
viral infection. In contrast, transduction of all 14 factors into fibro- (cardiac a-actin), Actn2 (actinin a2), and Nppa (natriuretic
blasts resulted in the generation of a small number of GFP+ cells peptide precursor type A) by quantitative RT-PCR (qPCR). We
(1.7%), indicating the successful activation of the cardiac- found that these cardiac genes were upregulated significantly
enriched aMHC gene in some cells (Figures 1D and 1E). more in GFP+ than in GFP cells (Figure 2E). Next, we used
To determine which of the 14 factors were critical for activating immunocytochemistry to determine if cardiac proteins were
cardiac gene expression, we serially removed individual factors expressed in GFP+ cells. Despite the detection of cTnT in only
from the pool of 14. Pools lacking five factors (Baf60c, Hand2, 30% of GFP+ cells, most GFP+ cells induced with the three fac-
Hopx, Hrt2, and Pitx2c) produced an increased number of tors expressed sarcomeric a-actinin (a-actinin) and had well-
GFP+ cells, suggesting they are dispensable in this setting defined sarcomeric structures, similar to neonatal cardiomyo-
(Figures 1D and 1E). Of note, removing Gata4 decreased the cytes (Figure 2F; Figure S1). In addition to a-actinin, some
percentage of GFP+ cells to 0.5%, and removing Pitx2c GFP+ cells also expressed cTnT and ANF (atrial natriuretic
increased it to 5%. Removal of the five factors listed above factor), indicating GFP+ cells expressed several cardiomyo-
resulted in an increase in the percentage of GFP+ cells to 13% cyte-specific markers (Figures 2G and 2H). We also confirmed
(Figure 1F). We conducted three further rounds of withdrawing that neither GFP+ nor GFP cells expressed smooth muscle or
single factors from nine-, six-, and five-factor pools, removing endothelial cell markers (Figure S2), suggesting specificity of
those that did not decrease efficiency upon withdrawal, and GMT effects.
found that four factors (Gata4, Mef2c, Mesp1, and Tbx5) were
sufficient for efficient GFP+ cell induction from cardiac fibro- Induced Cardiomyocytes Originate from Differentiated
blasts (Figures 1F–1H). The combination of these four factors Fibroblasts and Are Directly Reprogrammed
dramatically increased the number of fibroblasts activating the We next isolated neonatal cardiac fibroblasts by the conven-
aMHC-GFP reporter to over 20% (Figure 1I). tional fibroblast isolation method in which hearts were digested
with trypsin and plated on plastic dishes (Ieda et al., 2009).
Gata4, Mef2c, and Tbx5 Are Sufficient for More than 85% of the cells expressed Thy1, and we isolated
Cardiomyocyte Induction Thy1+/GFP cells by FACS to exclude cardiomyocyte contami-
Next, we examined the expression of cTnT by FACS. We found nation (Figure 3A). Fibroblasts transduced with GMT expressed
that 20% of GFP+ cells expressed cTnT at high enough levels GFP, cTnT, and actinin after 1 week at the same level as fibro-
to detect by FACS 1 week after the four-factor transduction. blasts isolated from explant cultures (Figures 3B and 3C). Similar
Again removing individual factors from the four-factor pool in results were obtained on introduction of GMT into adult cardiac
transduction, we found that Mesp1 was dispensable for cTnT fibroblasts, with full formation of sarcomeric structures (Fig-
expression (Figures 2A and 2B). In contrast, we did not observe ure 3D; Figure S2).
cTnT+ or GFP+ cells, when either Mef2c or Tbx5 was removed. To determine if the induced cardiomyocyte-like cells (iCMs)
Removal of Gata4 did not significantly affect the number of were arising from a subpopulation of stem-like cells, we

Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc. 377


A Control 4 factors 4 f - Gata4 4 f - Mef2c 4 f - Mesp1 4 f - Tbx5
Figure 2. Combination of Three Transcrip-
10
4

0% 0%
10
4

0.7% 4.3%
10 4
0.1% 0.5%
10 4
0.2% 0.1%
10
4

1.0% 6.5%
10
4

0.7% 0% tion Factors Induces Cardiac Gene Expres-


10
3
10
3
10
3
10
3
10
3
10 3
sion in Fibroblasts
(A) FACS analyses for a-MHC-GFP and cardiac
cTnT

10 2 10 2 10 2 10 2 10 2 10 2

10
1
10
1
10 1 10
1
10 1 10
1 Troponin T (cTnT) expression. Effects of the
10
0 100% 0% 10
0 70.3% 24.7% 10
0 70.9% 28.3% 10
0 98.6% 1.2 % 10
0 72.7% 19.7% 10
0 98.9% 0.3% removal of individual factors from the pool of four
100 10 1 10 2 10 3 10 4 10 0 101 10 2 10 3 10 4 10 0 10 1 10 2 10 3 10 4 100 101 10 2 103 10 4 100 101 10 2 103 10 4 100 10 1 10 2 10 3 10 4
factors on GFP+ and cTnT+ cell induction.
αMHC-GFP (B) Quantitative data of GFP+ cells and cTnT+ cells
B C
30 (%) 30
(%) in (A) (n = 3).
αMHC-GFP+ cells
25 25 (C) Effect of the transduction of pools of three, two,
* cTnT+ cells
20 20 and one factors on GFP+ and cTnT+ cell induction
15 15 (n = 3).
10 10 (D) FACS analyses for a-MHC-GFP and cTnT
expression. Effects of GMT plus Nkx2.5 and
* ** ** ** ** **
5 5
0
* ** ** 0 Baf60c/Gata4/Tbx5 transduction are shown.
Control

4 factors

Gata4

Mef2c

Mesp1

Tbx5

3 factors
Control

Gata4

Mef2c

Gata4

Mef2c
Tbx5

Tbx5
(E) The mRNA expression in GFP+ and GFP cells
7 days after GMT transduction was determined by
4 factors - 1 factor 3 factors - 1 1 factor qPCR (n = 3).
(F) Immunofluorescent staining for GFP, a-actinin,
D 3 factors 3 f + Nkx2.5 Baf60c/Gata4/Tbx5 F αMHC-GFP α-actinin Merged
10
4
10
4
10
4
and DAPI. The combination of the three factors,
0.8% 4.0% 0.3% 0.9% 0.3% 0.5%
10
3
10
3
10 3 GMT, induced abundant GFP, and a-actinin
expression in cardiac fibroblasts 2 weeks after
cTnT

10 2 10 2 10 2

10
1
10
1
10 1
transduction. High-magnification views in insets
0 82.2% 13.0% 0 96.4% 2.4% 0 96.5% 2.7% control show sarcomeric organization. See also Figures
10 10 10
100 10 1 10 2 10 3 10 4 10 0 10 1 10 2 10 3 10 4 10 0 10 1 10 2 10 3 10 4
S1 and S2.
αMHC-GFP (G) Induced cardiomyocytes expressed cTnT by
immunocytochemistry with clear sarcomeric orga-
E
Relative mRNA expression

250 * (αMHC-GFP+ vs. αMHC-GFP- cells) nization 4 weeks after transduction. Insets are
200 Myh6 3 factors high-magnification views.
Actc1 (H) Induced cardiomyocytes expressed ANF at
150
* Actn2 perinuclear sites 2 weeks after transduction.
100 Nppa All data are presented as means ± SD. *p < 0.01
50
* * versus relevant control. Scale bars represent
0 3 factors 100 mm.
See also Figures S1 and S2.
αMHC-GFP cTnT Merged αMHC-GFP ANF Merged
G H

(Figures 3E–3G). Like cardiac fibroblasts,


tail-tip fibroblast-derived GFP+ cells ex-
pressed a-actinin and had well-defined
sarcomeric structures (Figure 3H; Fig-
ure S3), suggesting noncardiac fibro-
blasts can also be reprogrammed into
cardiomyocytes by GMT induction.
analyzed c-kit expression (Beltrami et al., 2003) in the Thy1+/ These results excluded the possibility that the iCMs arose from
GFP cells. Most c-kit+ cells coexpressed Thy1, whereas 15% contamination of cardiomyocytes or cardiac progenitors before
of Thy1+ cells expressed c-kit, which is consistent with a previous cardiac induction in the fibroblast population.
report of cardiac explant-derived cells (Davis et al., 2009). We We also determined whether the reprogramming of fibroblasts
isolated GFP /Thy1+/c-kit+ cells and GFP /Thy1+/c-kit cells to differentiated cardiomyocytes was a direct event or if the
by FACS and transduced each population of cells with GMT. fibroblasts first passed through a cardiac progenitor cell fate
We found 2–3-fold more cardiomyocyte induction in GFP / before further differentiation. To distinguish between these two
Thy1+/c-kit cells than in GFP /Thy1+/c-kit+ cells (Figure S3). possibilities, we used mice expressing Isl1–yellow fluorescent
These results suggest that most of the iCMs originated from protein (YFP) obtained by crossing Isl1-Cre mice and R26R-
a c-kit-negative population. EYFP mice (Srinivas et al., 2001) (Figure S3). Isl1 is an early
We then sought to more definitively exclude the possibility of cardiac progenitor marker that is transiently expressed before
rare cardiac progenitors giving rise to iCMs. We tested the cardiac differentiation. If iCMs generated from fibroblasts
potential of mouse tail-tip dermal fibroblasts to generate iCMs. passed through a cardiac progenitor state, they and their
We found that sorted Thy1+/GFP tail-tip dermal fibroblasts descendants would permanently express YFP (Laugwitz et al.,
transduced with GMT expressed GFP at the same level as 2005). We isolated Isl1-YFP /Thy1+cells from Isl1-YFP heart
GMT-transduced cardiac fibroblasts, although the percentage explants by FACS and transduced the cells with GMT. The
of cTnT+ cells was less than cardiac fibroblast-derived iCMs resulting cTnT+ cells did not express YFP in significant numbers,

378 Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc.


A Control αMHC-GFP CF B Control Gata4/Mef2c/Tbx5 Figure 3. Induced Cardiomyocytes Origi-
10 4 10 4
10 5 0% 0% 10 5 86.6% 0% 0% 0% 1.9% 6.5% nate from Differentiated Fibroblasts and
10
3
10
3 Are Directly Reprogrammed
10 4 10 4
(A) Cardiac fibroblasts (CF) isolated by the conven-
10 2 10 2
10
3
10 3 tional isolation method. Most cells were positive
for Thy1, and Thy-1+/GFP cells were sorted by

cTnT
Thy1

1
2 2 10 1 10
10 10
0 0
FACS for transduction.
100% 0% 13.2% 0.1% 0 100% 0% 0
73.3% 18.2%
2 3 4 5 2 3 4 5
10
100 10 1 10 2 10 3 10 4
10
10 0 10 1 10 2 10 3 10 4
(B) FACS analyses for aMHC-GFP and cTnT
0 10 10 10 10 0 10 10 10 10
expression in cardiac fibroblasts isolated in (A)
αMHC-GFP αMHC-GFP
1 week after transduction by GMT.
C D Adult αMHC-GFP Explant (C) Immunofluorescent staining for GFP, a-actinin,
αMHC-GFP CF-derived iCMs Gata4/Mef2c/Tbx5
10 4

3.5%
and DAPI in the GMT induced cardiomyocytes
5.2%
10
3
originated from (A).
(D) Cardiac fibroblasts isolated from adult aMHC-
10 2
cTnT GFP hearts were transduced with three factors.
10 1
See also Figure S2.
αMHC-GFP α-Actinin Merged (E) Thy-1+/GFP tail-tip dermal fibroblasts (TTFs)
0
73.9% 17.4%
10
100 10 1 10 2 10 3 10 4
were sorted by FACS for transduction.
αMHC-GFP (F) FACS analyses for GFP and cTnT expres-
E F G sion in TTFs isolated in (E) 1 week after GMT
αMHC-GFP TTF Gata4/Mef2c/Tbx5
transduction.
88.4% 0%
10 4 30 (%) αMHC-GFP+ cells
10 5 1.5% 2.5%
25 cTnT+ cells (G) Quantitative data of GFP+ cells and cTnT+ cells
10 3
10
4
20 indicated in (F) (n = 3 in each group).
10 3
10 2 15 (H) Immunofluorescent staining for GFP, a-actinin,
cTnT
Thy1

10 and DAPI in TTF-derived iCMs. See also Figure S3.


2 10 1
10
0
5 * (I) Isl1-YFP /Thy1+cells were sorted from Isl1-Cre/
11.6% 0% 74.5% 21.5%
2 3 4 5
10
0
0 1 2 3 4
0 Rosa-YFP heart explants and transduced with
0 10 10 10 10 10 10 10 10 10 CF TTF
GMT. See also Figure S3.
αMHC-GFP αMHC-GFP One week
(J) The vast majority of cTnT+ cells induced from
H I Isl1-YFP /Thy1+cells was negative for YFP.
αMHC-GFP TTF-derived iCMs Control Isl1-YFP Explant
10 5 0% 0% 35.5% 27.5%
(K) Mesp1-YFP /Thy1+cells were sorted from
10 5
Mesp1-Cre/Rosa-YFP TTFs and transduced with
4 4
10 10
GMT. See also Figure S3.
10
3
10
3
(L) All cTnT+ cells induced from Mesp1-YFP /
Thy1

Thy1+cells were negative for YFP.


10 2
10 2

αMHC-GFP α-Actinin Merged 0 0 All data are presented as means ± SD. *p < 0.01
100% 0% 19.4% 17.5%
0 10
2
10
3
10
4
10
5 0 102 10 3 10 4 10 5
versus relevant control. Scale bars represent
Isl1-YFP 100 mm. See also Figure S3 for analyses of c-kit+
cells.
J Gata4/Mef2c/Tbx5 K Mesp1-YFP TTF L 10 4
Gata4/Mef2c/Tbx5
10 4
0.4% 87.7% 0.2% 5.7% 0% See also Figures S2 and S3.
8.6% 10 5

3
10 3 10
4
10

10 2 10 2
10 3
cTnT

Thy1

cTnT

10 1 2 10 1
10

89.7% 1.3%
0
11.9% 0.2% 94.3% 0%
3L). The resulting cTnT+ cells did not
0 0
10 10
100 10 1 10 2 10 3 10 4 0 102 10 3 10 4 10 5 100 101 10 2 10 3 10 4 express YFP, suggesting that the iCMs
Isl1-YFP Mesp1-YFP Mesp1-YFP were not converted into the cardiac
mesoderm cell state for reprogramming,
but rather they were directly reprog-
suggesting that the iCMs were not first reprogrammed into Isl1+ rammed into differentiated cardiomyocytes by the three factors
cardiac progenitor cells (Figures 3I and 3J). Moreover, these (Figure 3L).
results provided supportive evidence that the iCMs did not orig-
inate from a rare population of cardiac progenitor cells that might Induced Cardiomyocytes Resemble Postnatal
exist in neonatal hearts. Cardiomyocytes in Global Gene Expression
Whereas Isl1 marks most early cardiac progenitors, a subpop- We next analyzed the time course of cardiomyocyte induction
ulation of cardiac progenitors remains Isl1 negative. Mesp1 is from cardiac fibroblasts. GFP+ cells were detected 3 days after
the earliest pan-cardiovascular progenitor cell marker that is induction and gradually increased in number up to 20% at day
transiently expressed in nascent mesoderm before further 10 and were still present after 4 weeks (Figure 4A). GFP+ cells
cardiovascular differentiation (Figure S3) (Saga et al., 1999). were less proliferative than GFP cells and, over time, decreased
We therefore generated Mesp1-YFP mice by crossing Mesp1- in percentage relative to the total number of cells. Importantly, the
Cre and R26R-EYFP mice to determine if iCMs were reprog- percentage of cTnT+ cells among the a-MHC-GFP+ iCMs and the
rammed into early cardiac mesoderm before further differentia- intensity of cTnT expression increased significantly over time
tion. We isolated Mesp1-YFP /Thy1+ tail-tip dermal fibroblasts (Figures 4B and 4C). We sorted GFP+ cells at 1, 2, and 4 weeks
by FACS and transduced the cells with GMT (Figures 3K and after transduction with GMT and compared cardiac gene

Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc. 379


A B Percent of cTnT+ cells in αMHC-GFP+ iCMs Figure 4. Gene Expression of Induced Car-
diomyocytes Is Globally Reprogrammed
Percent (%) of total cells

4
Control 4
1 week 4
2 weeks 4
4 weeks
(A) The percent of aMHC-GFP+ cells after GMT
10 10 10 10
30 αMHC-GFP+ cells
25
* 10
3
100% 10
3
59.2% 10
3
55.9% 10
3
51% transduction (n = 3). The number of GFP+ cells
20
* * 10 2 10 2 10 2 10 2
was counted by FACS at each time point and
15
** divided by the number of plated cells.
10 10
1 0% 10
1 31.8% 10 1
35.5% 10
1 45.1%
5 (B) FACS analyses of percent of cells with cTnT
(vs. 3d)
0 10
0

10 0 10 1 10 2 10 3
10
10 4
0

100 101 10 2 10 3
10
10 4
0

10 0 101 10 2 10 3
10
10 4
0

10 0 10 1 10 2 103 10 4
expression among aMHC-GFP+ iCMs. Note that
0 1 3 7 10 14 28
Time (days)
cTnT
cTnT + cell number and cTnT intensity were both
increased over time (n = 4).
C Relative
D Relative mRNA expression (C) Quantitative data of cTnT intensity in (B) (n = 4).
2000
CF
cTnT intensity Actc1 Myh6 Ryr2 (D) Actc1, Myh6, Ryr2, Gja1, and Col1a2 mRNA
1600
* 1.0 1.0 1.0 iCMs (1W)
0.8 0.8 0.8 expression in cardiac fibroblasts (CF), induced
iCMs (2W)
1200 * cardiomyocytes (iCMs) (1 week (W), 2 W, 4 W after
0.6 0.6 0.6 iCMs (4W)
800 * * * transduction), and neonatal cardiomyocytes (CM),
0.4 0.4 0.4 CM
400 0.2 ** 0.2 * 0.2 determined by qPCR (n = 3).
*
(vs. 1W) ND ND ND (E) Heatmap image of microarray data illustrating
0 0 0 0
0 1 2 4 differentially expressed genes among CF, a-
Time (weeks) 1.0 Gja1 16 Col1a2
(vs. 1W iCMs) MHC-GFP , iCMs (FACS sorted 2 and 4 weeks
0.8 12 after transduction), and CM (n = 3 in each group).
0.6
* 8 The scale extends from 0.25- to 4-fold over
0.4
4
mean ( 2 to +2 in log2 scale). Red indicates
0.2
increased expression, whereas green indicates
0 0 *
decreased expression. Group 1 includes the
E genes upregulated only in CM, and group 2
includes the genes upregulated in CM and 4W-
iCMs compared to CF. Lists of genes are shown
CF 2W GFP- 4W GFP- 2W iCMs 4W iCMs CM
in Table S1 and Table S2. All data are presented
as means ± SD. *p < 0.01; **p < 0.05 versus rele-
vant control. See also Figure S4 for endogenous
and exogenous expression of reprogramming
factors and Table S1 and Table S2 for differentially
expressed genes.
See also Tables S1 and S2 and Figure S4.

1
We next compared the progressive
global gene expression pattern of iCMs,
neonatal cardiomyocytes, and cardiac
2
fibroblasts by mRNA microarray anal-
yses. We sorted GFP+ cells and GFP
FDR-adj p<0.0001 in at least one comparison cells 2 and 4 weeks after GMT transduc-
tion. The iCMs at both stages were similar
to neonatal cardiomyocytes, but were
expression with cardiac fibroblasts and neonatal cardiomyo- distinct from GFP cells and cardiac fibroblasts in global gene
cytes. The cardiomyocyte-specific genes, Actc1, Myh6, Ryr2 expression pattern (Figure 4E). We found that functionally impor-
(ryanodine receptor 2), and Gja1 (connexin43), were significantly tant cardiac genes were upregulated significantly more in 4 week
upregulated in a time-dependent manner in GFP+ cells, but iCMs than in 2 week iCMs, including Pln (phospholamban),
were not detected in cardiac fibroblasts by qPCR (Figure 4D). Slc8a1 (sodium/calcium exchanger), Myh6, Sema3a (sema-
Col1a2 (collagen 1a2), a marker of fibroblasts, was dramatically phorin 3a), Id2 (inhibitor of DNA binding 2), and Myl2 (myosin, light
downregulated in GFP+ cells from 7-day culture to the same level polypeptide 2, regulatory, cardiac, slow, also known as MLC2v)
as in cardiomyocytes. These data indicated that the three factors (Table S1). Conversely, some genes were downregulated more
induced direct conversion of cardiac fibroblasts to cardiomyo- in 4 week iCMs than in 2 week iCMs (Table S1). The array analyses
cytes rapidly and efficiently, but full maturation was a slow also identified genes that were upregulated more in neonatal car-
process that occurred over several weeks. Total gene expression diomyocytes than in 4 week iCMs or cardiac fibroblasts (group 1
of the three reprogramming factors was upregulated 6- to 8-fold in in Figure 4E), including Bmp10 (bone morphogenetic protein 10),
iCMs over neonatal cardiomyocytes. However, only endogenous Erbb4 (v-erb-a erythroblastic leukemia viral oncogene homolog
expression of Gata4 was upregulated in iCMs to the same level as 4), Irx4 (Iroquois related homeobox 4), and Atp1a2 (ATPase,
in neonatal cardiomyocytes, whereas endogenous Mef2c and Na+/K+ transporting, a 2 polypeptide) (Table S2). We also identi-
Tbx5 expression was lower in iCMs than in cardiomyocytes, fied genes that were expressed to a greater extent in both cardi-
potentially reflecting negative autoregulatory loops (Figure S4). omyocytes and 4 week iCMs than in fibroblasts (group 2 in

380 Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc.


A B Figure 5. Fibroblasts Are Stably Reprog-
CF iCMs (CF) TTF iCMs (TTF) Cardiac cells
Nppa Myh6 rammed into iCMs by Gata4, Mef2c, and
H3K27me3 H3K4me3
20 5 Tbx5
4
* ** (A) The promoters of Actn2, Ryr2, and Tnnt2 were
15
Actn2

3 CF analyzed by ChIP for trimethylation status of


10
2 histone H3 of lysine 27 or 4 in cardiac fibroblasts
5 * 1
** (CF), CF-derived iCMs, tail-tip fibroblasts (TTF),
Fold enrichment/IgG

0 0
20 5 αMHC- TTF-derived iCMs, and neonatal cardiac cells.
GFP- Data were quantified by qPCR.
15 4
(B) The promoters of Nppa and Myh6 were
Ryr2

3
10 ** analyzed with bisulfite genomic sequencing for
2
5
** * 1
iCMs
DNA methylation status in CF, a-MHC-GFP cells,
0 0 a-MHC-GFP+ iCMs (FACS sorted 4 weeks after
15 5 transduction), and neonatal CM. Open circles indi-
4 cate unmethylated CpG dinucleotides; closed
10
Tnnt2

3 circles indicate methylated CpGs.


2 CM
5 * 1
* * (C) Schematic representation of the strategy to
0
* 0
test expression kinetics of the doxycycline (Dox)-
inducible lentiviral system.
C WT TTF
+Dox -Dox (D) Wild-type TTFs were infected with pLVX-tetO-
1 day 6 days GFP and pLVX-rtTA and imaged before (off Dox),
1 day after Dox addition, and at time points after
infection
Dox withdrawal (–Dox). All images were taken
5´-LTR tetO GFP 3´-LTR
using constant exposure times and identical
5´-LTR CMV rtTA 3´-LTR
camera settings.
(E) Schematic representation of the strategy to
D Off Dox 1d, +Dox 1d, -Dox 3d, -Dox 6d, -Dox determine temporal requirement of Gata4/Mef2c/
Tbx5 in reprogramming. Thy1+/GFP TTF were
GFP

infected with the pLVX-tetO-GMT and pLVX-rtTA


lentiviruses, and Dox was added for 2 weeks and
thereafter withdrawn for 1 week.
(F) Immunofluorescent staining for GFP, a-actinin,
E Off On and DAPI in iCMs 2 weeks after lentiviral infection
αMHC promoter GFP αMHC promoter GFP and Dox induction.
αMHC-GFP TTF +Dox iCMs -Dox iCMs (G) Immunofluorescent staining for GFP, a-actinin,
2 weeks 1 week and DAPI 1 week after Dox withdrawal. iCMs
infection maintained a-MHC GFP expression and had
5´-LTR tetO Gata4/Mef2c/Tbx5 3´-LTR
a-actinin positive sarcomeric structures. High-
5´-LTR CMV rtTA 3´-LTR
magnification views in insets show sarcomeric
organization. Representative data are shown in
F +Dox G +Dox -Dox each panel. All data are presented as means ±
SD. *p < 0.01; **p < 0.05 versus relevant control.
Scale bars represent 100 mm.

αMHC-GFP α-actinin Merged αMHC-GFP α-actinin Merged

Figure 4E), including Actc1, Myl7 (myosin, light polypeptide 7, iCMs, and neonatal cardiac cells by chromatin immunoprecipita-
regulatory, also known as MLC2a), Tnnt2 (troponin T2, cardiac), tion, followed by qPCR (Figure 5A). After reprogramming,
Tbx3 (T-box 3), and Srf (serum response factor) (Table S2). H3K27me3 was significantly depleted at the promoters of all
Thus, iCMs were similar, but not identical, to neonatal cardiomyo- the genes analyzed in iCMs, reaching levels comparable to those
cytes, and the reprogramming event was broadly reflected in in cardiac cells, whereas H3K4me3 increased on the promoter
global gene expression changes. regions of Actn2 and Tnnt2 in iCMs, as compared with cardiac
fibroblasts. Ryr2 had similar levels of H3K4me3 in iCMs as in
Fibroblasts Are Epigenetically Reprogrammed fibroblasts, suggesting that its activation reflects the resolution
to a Cardiomyocyte-like State by Gata4/Mef2c/Tbx5 of a ‘‘bivalent’’ chromatin mark (Bernstein et al., 2006). These
To determine if iCMs have gained a cardiomyocyte-like chro- results suggested that cardiac fibroblast-derived iCMs gained
matin state, we analyzed the enrichment of histone modifications a chromatin status similar to cardiomyocytes at least in some
in the promoter regions of the cardiac-specific genes Actn2, cardiac specific genes. Intriguingly, H3K27me3 levels were
Ryr2I, and Tnnt2. We analyzed the enrichment of trimethylated higher in tail-tip fibroblasts than cardiac fibroblasts on all three
histone H3 of lysine 27 (H3K27me3) and lysine 4 (H3K4me3), genes analyzed and, despite a significant reduction upon
which mark transcriptionally inactive or active chromatin, reprogramming to iCMs, remained somewhat higher than in
respectively (Li et al., 2007), in cardiac fibroblasts, 4 week cardiac cells and cardiac fibroblast-derived iCMs.

Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc. 381


The DNA methylation status of specific loci also reflects the iCMs displayed action potentials that resembled those of adult
stability of the reprogramming event and we therefore investi- mouse ventricular cardiomyocytes (Figure 6G). Thus, the reprog-
gated such changes during reprogramming from cardiac fibro- ramming of fibroblasts to iCMs was associated with global
blasts to iCMs. We performed bisulfite genomic sequencing in changes in gene expression, epigenetic reprogramming, and
the promoter regions of Nppa and Myh6 in cardiac fibroblasts, the functional properties characteristic of cardiomyocytes.
4 week GFP cells, iCMs, and neonatal cardiomyocytes. Both
promoter regions were hypermethylated in cardiac fibroblasts Transplanted Cardiac Fibroblasts Transduced
and GFP cells, as expected from the cardiomyocyte-specific with Gata4/Mef2c/Tbx5 Reprogram In Vivo
expression of these genes, but were comparatively demethy- To investigate whether GMT-transduced cardiac fibroblasts can
lated in iCMs, similar to neonatal cardiomyocytes (Figure 5B). be reprogrammed to express cardiomyocyte-specific genes in
These results indicated that reprogramming by Gata4, Mef2c, their native environment in vivo, we harvested GFP /Thy1+
and Tbx5 induced epigenetic resetting of the fibroblast genome cardiac fibroblasts 1 day after viral transduction and injected
to a cardiomyocyte-like state. them into immunosuppressed NOD-SCID mouse hearts. GMT-
To further assess the stability of the reprogramming event, we infected cells did not express GFP at the time of transplantation
generated a doxycycline-inducible lentiviral system in which (Figure 4A). Cardiac fibroblasts were infected with either the
transgene expression of the reprogramming factors was con- mixture of GMT and DsRed retroviruses or DsRed retrovirus
trolled by doxycycline administration. We first transduced wild- (negative control) to be readily identified by fluorescence.
type tail-tip fibroblasts with a mixture of lentiviruses containing Cardiac fibroblasts infected with DsRed did not express a-acti-
pLVX-tetO-GFP and pLVX-rtTA to determine the expression nin or GFP, confirming cardiomyocyte conversion did not
kinetics of this system (Figure 5C). We confirmed that the happen in the negative control (Figures 7A and 7B). Despite
majority of fibroblasts infected with both viruses expressed being injected into the heart only 1 day after viral infection,
GFP within 1 day after doxycycline induction, and the GFP a subset of cardiac fibroblasts transduced with GMT and DsRed
expression was instantly diminished by withdrawal of doxycy- expressed GFP in the mouse heart within 2 weeks (Figure 7B).
cline and disappeared within 6 days (Figure 5D). Thy1+/GFP Importantly, the GFP+ cells expressed a-actinin and had sarco-
tail-tip fibroblasts were harvested from aMHC-GFP neonatal meric structures (Figure 7C). These results suggested that
mice, transduced with a pool of lentiviruses containing inducible cardiac fibroblasts transduced with Gata4, Mef2c, and Tbx5
Gata4, Mef2c, and Tbx5, along with pLVX-rtTA, and subse- can reprogram to cardiomyocytes within 2 weeks upon trans-
quently treated with doxycycline (Figure 5E). We found that plantation in vivo.
aMHC-GFP expression was induced from tail-tip fibroblasts
after doxycycline administration and that the iCMs had well- DISCUSSION
defined sarcomeric structures marked with an anti-a-actinin
antibody after 2 weeks of culture (Figure 5F). Doxycycline was Here we demonstrated that the combination of three transcrip-
withdrawn after 2 weeks of culture, and cells were subsequently tion factors, Gata4, Mef2c, and Tbx5, can rapidly and efficiently
cultured without doxycycline for 1 week to fully remove exoge- induce cardiomyocyte-like cells from postnatal cardiac and
nous expression of the reprogramming factors (Figure 5E). The dermal fibroblasts. iCMs were similar to neonatal cardiomyo-
iCMs maintained aMHC-GFP expression and had sarcomeric cytes in global gene expression profile, electrophysiologically,
structures after doxycycline withdrawal, suggesting that the and could contract spontaneously, demonstrating that func-
fibroblasts were stably reprogrammed into iCMs after 2 weeks tional cardiomyocytes can be generated from differentiated
exposure to Gata4, Mef2c, and Tbx5 (Figure 5G). somatic cells by defined factors. Although much refinement
and characterization of the reprogramming process will be
Induced Cardiomyocytes Exhibit necessary, the findings reported here raise the possibility of
Spontaneous Contraction reprogramming the vast pool of endogenous fibroblasts that
To determine if iCMs possessed the functional properties char- normally exists in the heart into functional cardiomyocytes for
acteristic of cardiomyocytes, we analyzed intracellular Ca2+ regenerative purposes.
flux in iCMs after 2–4 weeks of culture. Around 30% of cardiac The three reprogramming factors, Gata4, Mef2c, and Tbx5,
fibroblast-derived iCMs showed spontaneous Ca2+ oscillations are core transcription factors during early heart development
and their frequency was variable, resembling what was observed (Olson, 2006; Srivastava, 2006; Zhao et al., 2008). They interact
in neonatal cardiomyocytes (Figures 6A, 6B, and 6D; Movie S1). with one another, coactivate cardiac gene expression (e.g.,
We observed that tail-tip dermal fibroblast-derived iCMs also Nppa, Gja5 [Cx40], and Myh6), and promote cardiomyocyte
exhibited spontaneous Ca2+ oscillations, but the oscillation differentiation (Bruneau et al., 2001; Garg et al., 2003; Ghosh
frequency was lower than that of cardiomyocytes and cardiac et al., 2009; Lin et al., 1997). Gata4 is considered a ‘‘pioneer’’
fibroblast-derived iCMs (Figures 6C and 6E; Movie S2). factor and might open chromatin structure in cardiac loci (Cirillo
In addition to the characteristic Ca2+ flux, cardiac fibroblast- et al., 2002), thus allowing binding of Mef2c and Tbx5 to their
derived iCMs showed spontaneous contractile activity after specific target sites and leading to full activation of the cardiac
4–5 weeks in culture (Movies S3 and S4; Figure S5). Single-cell program. Although the reprogramming event appears stable at
extracellular recording of electrical activity in beating cells the epigenetic level, as marked by histone methylation and
revealed tracings similar to the potential observed in neonatal DNA methylation, the global gene expression of iCMs is similar
cardiomyocytes (Figure 6F). Intracellular electrical recording of but not identical to neonatal cardiomyocytes. Whether they are

382 Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc.


A C Rhod-3 intensity Figure 6. Induced Cardiomyocytes Exhibit
Rhod-3 intensity (Cardiac fibroblast-derived iCMs) (Tail tip fibroblast-derived iCMs) Spontaneous Ca2+ Flux, Electrical Activity,
and Beating
(A and B) Cardiac fibroblast (CF)-derived iCMs
showed spontaneous Ca2+ oscillation with varying
1s 1s 1s
frequency (A), similar to neonatal cardiomyocytes
(B). Rhod-3 intensity traces are shown.
B Rhod-3 intensity (Neonatal cardiomyocytes) (C) Tail-tip dermal fibroblast (TTF)-derived iCMs
showed spontaneous Ca2+ oscillation with lower
frequency. The Rhod-3 intensity trace is shown.
(D) Spontaneous Ca2+ waves observed in CF-
1s 1s derived a-MHC-GFP+ iCMs (white dots) or
neonatal cardiomyocytes (arrows) with Rhod-3 at
Ca2+ max and min is shown. Fluorescent images
D Cardiac fibroblast-derived iCMs Neonatal cardiomyocytes correspond to the Movie S1.
(E) Spontaneous Ca2+ oscillation observed in the
TTF-derived a-MHC-GFP+ iCMs with Rhod-3 at
Ca2+ max and min is shown. Fluorescent images
correspond to the Movie S2.
αMHC-GFP Ca2+ max Ca2+ min Ca2+ max Ca2+ min (F) Spontaneously contracting iCMs had electrical
activity measured by single cell extracellular elec-
trodes. Neonatal cardiomyocytes showed similar
E Tail tip fibroblast-derived iCMs
electrical activity.
(G) Intracellular electrical recording of CF-derived
iCMs cultured for 10 weeks displayed action
potentials that resembled those of adult mouse
ventricular cardiomyocytes. Representative data
αMHC-GFP Ca2+ max Ca2+ min are shown in each panel (n = 10 in A–F, n = 4 in
G). See also Figure S5 and Movies S1, S2, S3
F Cardiac fibroblast-derived iCMs G and S4.
Cardiac fibroblast-derived iCMs See also Movies S1, S2, S3, and S4 and Figure S5.
extracellular electrical recording
0 mV
2 mV

20 mV

1s
50 ms
ming factors. Remarkably, reprogram-
Neonatal cardiomyocytes ming of cardiac fibroblasts to myocytes
Adult mouse ventricular cardiomyocytes
extracellular electrical recording occurred in a relatively short period,
with the first GFP+ cells appearing at
day 3, in contrast to iPSC reprogram-
2 mV

0 mV
ming, which typically takes 10–20 days
1s
and occurs with much lower efficiency
20 mV

(<0.1%) (Takahashi and Yamanaka,


50 ms 2006). Despite the early initiation of
reprogramming, the process appears to
continue for several weeks, with progres-
more similar to adult ventricular cardiomyocytes or other sive changes in gene expression, contractile ability, and electro-
subpopulations remains to be determined. Additional epigenetic physiologic maturation.
regulators, microRNAs, or signaling proteins may be leveraged Although many questions remain regarding the mechanisms
to increase the efficiency and robustness of the reprogramming of reprogramming, we were able to genetically test the ‘‘route’’
event. Furthermore, other combinations of factors likely also of cell fate alteration. Our findings suggest that cardiomyocytes
induce cardiac reprogramming, much like the experience in the were directly induced from cardiac fibroblasts without reverting
iPSC field. to a cardiac progenitor cell state, which may explain the rapid
Several lines of evidence suggest that the iCMs we describe early reprogramming process. This conclusion was supported
here originated from differentiated fibroblasts. We found that by the absence of Isl1-Cre-YFP or Mesp1-Cre-YFP activation
any potential rare cardiac ‘‘progenitor-like’’ cells, marked by during the process of reprogramming, which would have marked
c-kit or Isl1, were dispensable for cardiomyocyte induction any cells that transiently expressed Isl1 or Mesp1 (Laugwitz
(Beltrami et al., 2003). Furthermore, the high efficiency of et al., 2005; Saga et al., 1999).
cardiac induction (up to 20%) does not favor the interpretation The ability to reprogram endogenous cardiac fibroblasts into
that rare stem or progenitor cells were the origin of induced cardiomyocytes has many therapeutic implications. First, the
cardiomyocytes. Most importantly, the ability to reprogram avoidance of reprogramming to pluripotent cells before cardiac
dermal fibroblasts into iCMs supports the conclusion that differentiation would greatly lower the risk of tumor formation
cardiac progenitors are not the target cells for the reprogram- in the setting of future cell-based therapies. Second, large

Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc. 383


A DsRed-cell injected DsRed-cell injected DsRed-cell injected Figure 7. Transplanted Cardiac Fibroblasts Trans-
duced with Gata4/Mef2c/Tbx5 Can Be Reprog-
rammed to Cardiomyocytes In Vivo
(A) DsRed infected cardiac fibroblasts (DsRed-cell) were
transplanted into NOD-SCID mouse hearts 1 day after
infection and cardiac sections were analyzed by immuno-
cytochemistry after 2 weeks. Transplanted fibroblasts
marked with DsRed did not express a-actinin (green).
DsRed α-actinin Merged (B) Cardiac fibroblasts infected with DsRed or Gata4/
Mef2c/Tbx5 with DsRed (GMT/DsRed-cell) were trans-
B
planted into NOD-SCID mouse hearts 1 day after infection
DsRed-cell injected DsRed-cell injected DsRed-cell injected and visualized by histologic section. Note that a subset of
GMT/DsRed cells expressed a-MHC-GFP. Data were
analyzed 2 weeks after transplantation.
(C) Gata4/Mef2c/Tbx5-transduced cardiac fibroblasts
(GMT-cell) were transplanted into mouse hearts and histo-
logic sections analyzed. A subset of induced GFP+ cells
DsRed αMHC-GFP Merged expressed a-actinin (red) and had sarcomeric structures.
Insets are high-magnification views of cells indicated by
GMT/DsRed-cell injected GMT/DsRed-cell injected GMT/DsRed-cell injected
arrows. Data were analyzed 2 weeks after transplantation.
Representative data are shown in each panel (n = 4 in each
group). Scale bars represent 100 mm. Note that GMT/
DsRed or GMT-infected cells did not express GFP at the
time of transplantation (Figure 4A).

DsRed αMHC-GFP Merged


C GMT-cell injected GMT-cell injected GMT-cell injected
Cell Culture
For explant culture, isolated neonatal or adult mouse
hearts were minced into small pieces less than 1 mm3 in
size. The explants were plated on gelatin-coated dishes
and cultured for 7 days in explant medium (IMDM/20%
FBS) (Andersen et al., 2009). Migrated cells were har-
vested and filtered with 40 mm cell strainers (BD) to avoid
α-actinin αMHC-GFP Merged contamination of heart tissue fragments. aMHC-GFP /
Thy1+, Isl1-YFP /Thy1+, aMHC-GFP /Thy1+/c-kit , or
aMHC-GFP /Thy1+/c-kit+ live cells (as defined by the
amounts of an individual’s own fibroblasts can be grown from lack of propidium iodine staining) were isolated using FACS Aria 2 (BD Biosci-
a cardiac biopsy or skin biopsy in vitro for transduction with ences). For conventional isolation of neonatal cardiac fibroblasts, hearts were
the defined factors, followed by delivery of cells to damaged digested with 0.1% trypsin and plated on plastic dishes (Ieda et al., 2009).
For isolation of tail-tip fibroblasts, tails were digested with 0.1% trypsin and
hearts. Third, and most promising, is the potential to introduce
plated on plastic dishes. Attached fibroblasts were cultured for 7 days and
the defined factors, or factors that mimic their effects, directly aMHC-GFP /Thy1+ or Mesp1-YFP /Thy1+ cells were sorted and cultured in
into the heart to reprogram the endogenous fibroblast popula- DMEM/M199 medium containing 10% FBS at a density of 104/cm2. Cells
tion, which represents more than 50% of the cells, into new car- were transduced by retroviruses or lentiviruses after 24 hr.
diomyocytes that can contribute to the overall contractility of the
heart. Our observation that injection of fibroblasts into the heart Isolation of Cardiomyocytes
To isolate cardiomyocytes, neonatal aMHC-GFP+ ventricles were cut into
only 1 day after induction of Gata4/Mef2c/Tbx5 resulted in
small pieces and digested with collagenase type II solution (Ieda et al.,
reprogramming of the transplanted cells suggests that this 2009). A single-cell suspension was obtained by gentle triturating and passing
may be possible. Future studies in human cells and advances through a 40 mm cell strainer. aMHC-GFP+ live cells were isolated by FACS
in safe delivery of defined factors will be necessary to advance Aria 2. To obtain cardiac cells, cells were plated on gelatin-coated plastic
this technology for potential regenerative therapies. dishes and treated with Ara C (Sigma) to inhibit nonmyocyte proliferation.

Molecular Cloning and Retroviral/Lentiviral Infection


EXPERIMENTAL PROCEDURES Retroviruses or inducible lentiviruses containing the cardiac developmental
factors were generated as described and as detailed in the Extended Experi-
Generation of aMHC-GFP, Isl1-YFP, and Mesp1-YFP Mice mental Procedures (Kitamura et al., 2003; Takahashi and Yamanaka, 2006).
To generate aMHC-GFP mice, EGFP-IRES-Puromycin cDNA was subcloned The pMXs-DsRed Express retrovirus infection in cardiac fibroblasts resulted
into the expression vector containing a-myosin heavy chain promoter (Gulick in >95% transduction efficiency (Hong et al., 2009).
et al., 1991). Pronuclear microinjection and other procedures were performed
according to the standard protocols (Ieda et al., 2007). PCR primers are listed FACS Analyses and Sorting
in the Extended Experimental Procedures. Isl1-YFP mice were obtained by For GFP expression analyses, cells were harvested from cultured dishes and
crossing Isl1-Cre mice and R26R-EYFP mice (Srinivas et al., 2001). Mesp1- analyzed on a FACS Calibur (BD Biosciences) with FlowJo software. For
YFP mice were obtained by crossing Mesp1-Cre mice and R26R-EYFP mice aMHC-GFP/cTnT expression, cells were fixed with 4% PFA for 15 min, per-
(Saga et al., 1999). meabilized with Saponin, and stained with anti-cTnT and anti-GFP antibodies,

384 Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc.


followed by secondary antibodies conjugated with Alexa 488 and 647 (Katt- Electrophysiology
man et al., 2006). After 4 week transduction with GMT, the electrophysiological activities of iCMs
For aMHC-GFP /Thy1+, Isl1-YFP /Thy1+, and Mesp1-YFP /Thy1+ cell were analyzed using extracellular electrode recording with an Axopatch 700B
sorting, cells were incubated with PECy7 or APC-conjugated anti-Thy1 amplifier and the pClamp9.2 software (Axon Instruments). iCMs were visually
antibody (eBioscience) and sorted by FACS Aria 2 (Ieda et al., 2009). identified by GFP expression and spontaneous contraction. Glass patch
For aMHC-GFP /Thy1+/c-kit and aMHC-GFP /Thy1+/c-kit+ cell sorting, pipettes, with typical resistances of 2–4 MU, were directly attached on single
PECy7-conjugated anti-Thy1 and APC-conjugated anti-c-kit antibodies GFP+ cells for extracellular recording in Tyrode’s bath solution. For recording
(BD) were used. We used bone marrow cells as a positive control for c-kit intracellular action potentials, single GFP+ cells were held at 70 mV
staining. membrane potential with a stimulation of 0.1–0.5 nA for 5 ms to elicit
a response after 10-week transduction with GMT.
Cell Transplantation
Fibroblasts were harvested 1 day after retroviral infection. A left thoracotomy Statistical Analyses
was carried out in NOD-SCID mice, and 106 cultured cells were injected Differences between groups were examined for statistical significance using
into the left ventricle. After 1–2 weeks, the hearts were excised for Student’s t test or ANOVA. p values of <0.05 were regarded as significant.
immunohistochemistry.
ACCESSION NUMBERS
Histology and Immunocytochemistry
Cells or tissues were fixed, processed and stained with antibodies against Microarray data have been submitted and can be accessed by the Gene
numerous proteins in standard fashion as detailed in the Extended Experi- Expression Omnibus (GEO) accession number GSE22292.
mental Procedures.
SUPPLEMENTAL INFORMATION
Quantitative RT-PCR
Supplemental Information includes Extended Experimental Procedures, five
Total RNA was isolated from cells, and qRT-PCR was performed on an ABI
figures, two tables, and four movies and can be found with this article online
7900HT (Applied Biosystems) with TaqMan probes (Applied Biosystems),
at doi:10.1016/j.cell.2010.07.002.
which are listed in the Extended Experimental Procedures. To quantify endog-
enous-specific transcripts and both endogenous and transgene common tran-
ACKNOWLEDGMENTS
scripts, primers were designed using Vector NTI, and SYBR green technology
was used. Primer information is available on request. mRNA levels were
We are grateful to members of the Srivastava lab, to K. Tomoda for critical
normalized by comparison to Gapdh mRNA.
discussions and comments on the manuscript, to J. Olgin and C. Ding for elec-
trophysiology assistance, to Z. Yang and K. Worringer for help with lentivirus
Microarray Analyses experiments, to Y. Huang for cell transplantation experiments, to B. Taylor,
Mouse genome-wide gene expression analyses were performed using G. Howard, and S. Ordway for editorial assistance and manuscript prepara-
Affymetrix Mouse Gene 1.0 ST Array. aMHC-GFP+ cardiomyocytes were tion, to C. Barker and L. Ta in the Gladstone Genomics core, to C. Miller and
collected by FACS. Three-factor transduced GFP+ cells and GFP cells J. Fish in the Gladstone Histology core, to A. Holloway in the Gladstone Bioin-
were collected by FACS after 2 and 4 weeks of culture. Cardiac fibroblasts formatics core, and to S. Elmes in the Laboratory for Cell Analysis in UCSF. We
were also collected after 4 weeks of culture. RNA was extracted using Pico- also thank S. Yamanaka for helpful discussions and providing pMXs-DsRed
Pure RNA Isolation (Arcturus). Microarray analyses were performed in triplicate Express plasmid, T. Kitamura for Plat-E cells, J. Robbins for a-MHC promoter
from independent biologic samples, according to the standard Affymetrix plasmid, T.M. Jessell for Isl1-Cre mice, and F. Costantini for R26R-EYFP mice.
Genechip protocol. Data were analyzed using the Affymetrix Power Tool M.I. and Y.H. are supported by a grant from the Uehara Memorial Foundation.
(APT, version 1.8.5). See the Extended Experimental Procedures for additional V.V. is supported by grants from the GlaxoSmithKline Cardiovascular
statistical methods. Research and Education Foundation and the NIH/NHLBI. P.D.-O. was a post-
doctoral scholar of the California Institute for Regenerative Medicine. D.S. and
Chromatin Immunoprecipitation Assay B.G.B are supported by grants from NHLBI/NIH and the California Institute for
Chromatin immunoprecipitations were performed on cardiac fibroblasts, tail- Regenerative Medicine. The J. David Gladstone Institutes received support
tip dermal fibroblasts, iCMs, and neonatal cardiac cells. Immunoprecipitations from a National Center for Research Resources Grant RR18928-01. D.S. is
were done using the Imprint Chromatin Immunoprecipitation Kit (Sigma) a member of the Scientific Advisory Board of iPierian, Inc., and RegeneRx.
following the manufacturer instructions. Antibodies against H3K27me3 and B.G.B. is a member of the Scientific Advisory Board of iPierian Inc.
H3K4me3 were from Active motif, and normal rabbit IgG was from Cell
Signaling Technology. Primer sequences for qPCR custom TaqMan gene Received: February 18, 2010
expression assays (Applied Biosystems) are listed in the Extended Experi- Revised: May 18, 2010
mental Procedures. Accepted: June 25, 2010
Published: August 5, 2010
Bisulfite Genomic Sequencing
Bisulfite treatment was performed using the Epitect Bisulfite Kit (QIAGEN) REFERENCES
according to the manufacturer’s recommendations. PCR primers are listed
in the Extended Experimental Procedures. Amplified products were cloned Andersen, D.C., Andersen, P., Schneider, M., Jensen, H.B., and Sheikh, S.P.
into pCR2.1-TOPO (Invitrogen). Ten randomly selected clones were (2009). Murine ‘‘cardiospheres’’ are not a source of stem cells with cardiomyo-
sequenced with the M13 forward and M13 reverse primers for each gene. genic potential. Stem Cells 27, 1571–1581.
Baeyens, L., De Breuck, S., Lardon, J., Mfopou, J.K., Rooman, I., and Bou-
2+ wens, L. (2005). In vitro generation of insulin-producing beta cells from adult
Ca Imaging
exocrine pancreatic cells. Diabetologia 48, 49–57.
Ca2+ imaging was performed according to the standard protocol. Briefly, cells
were labeled with Rhod-3 (Invitrogen) for 1 hr at room temperature, washed, Baudino, T.A., Carver, W., Giles, W., and Borg, T.K. (2006). Cardiac fibroblasts:
and incubated for an additional 1 hr to allow de-esterification of the dye. friend or foe? Am. J. Physiol. Heart Circ. Physiol. 291, H1015–H1026.
Rhod-3-labeled cells were analyzed by Axio Observer (Zeiss) with MiCAM02 Beltrami, A.P., Barlucchi, L., Torella, D., Baker, M., Limana, F., Chimenti, S.,
(SciMedia). Kasahara, H., Rota, M., Musso, E., Urbanek, K., et al. (2003). Adult cardiac

Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc. 385


stem cells are multipotent and support myocardial regeneration. Cell 114, Kitamura, T., Koshino, Y., Shibata, F., Oki, T., Nakajima, H., Nosaka, T., and
763–776. Kumagai, H. (2003). Retrovirus-mediated gene transfer and expression
Bernstein, B.E., Mikkelsen, T.S., Xie, X., Kamal, M., Huebert, D.J., Cuff, J., Fry, cloning: powerful tools in functional genomics. Exp. Hematol. 31, 1007–1014.
B., Meissner, A., Wernig, M., Plath, K., et al. (2006). A bivalent chromatin struc- Laflamme, M.A., Chen, K.Y., Naumova, A.V., Muskheli, V., Fugate, J.A., Dup-
ture marks key developmental genes in embryonic stem cells. Cell 125, 315– ras, S.K., Reinecke, H., Xu, C., Hassanipour, M., Police, S., et al. (2007).
326. Cardiomyocytes derived from human embryonic stem cells in pro-survival
Bruneau, B.G., Nemer, G., Schmitt, J.P., Charron, F., Robitaille, L., Caron, S., factors enhance function of infarcted rat hearts. Nat. Biotechnol. 25, 1015–
Conner, D.A., Gessler, M., Nemer, M., Seidman, C.E., et al. (2001). A murine 1024.
model of Holt-Oram syndrome defines roles of the T-box transcription factor Laugwitz, K.L., Moretti, A., Lam, J., Gruber, P., Chen, Y., Woodard, S., Lin,
Tbx5 in cardiogenesis and disease. Cell 106, 709–721. L.Z., Cai, C.L., Lu, M.M., Reth, M., et al. (2005). Postnatal isl1+ cardioblasts
Camelliti, P., Borg, T.K., and Kohl, P. (2005). Structural and functional charac- enter fully differentiated cardiomyocyte lineages. Nature 433, 647–653.
terisation of cardiac fibroblasts. Cardiovasc. Res. 65, 40–51. Li, B., Carey, M., and Workman, J.L. (2007). The role of chromatin during tran-
Cirillo, L.A., Lin, F.R., Cuesta, I., Friedman, D., Jarnik, M., and Zaret, K.S. scription. Cell 128, 707–719.
(2002). Opening of compacted chromatin by early developmental transcription Lin, Q., Schwarz, J., Bucana, C., and Olson, E.N. (1997). Control of mouse
factors HNF3 (FoxA) and GATA-4. Mol. Cell 9, 279–289. cardiac morphogenesis and myogenesis by transcription factor MEF2C.
David, R., Brenner, C., Stieber, J., Schwarz, F., Brunner, S., Vollmer, M., Men- Science 276, 1404–1407.
tele, E., Muller-Hocker, J., Kitajima, S., Lickert, H., et al. (2008). MesP1 drives Nussbaum, J., Minami, E., Laflamme, M.A., Virag, J.A., Ware, C.B., Masino, A.,
vertebrate cardiovascular differentiation through Dkk-1-mediated blockade of Muskheli, V., Pabon, L., Reinecke, H., and Murry, C.E. (2007). Transplantation
Wnt-signalling. Nat. Cell Biol. 10, 338–345. of undifferentiated murine embryonic stem cells in the heart: teratoma forma-
Davis, D.R., Zhang, Y., Smith, R.R., Cheng, K., Terrovitis, J., Malliaras, K., Li, tion and immune response. FASEB J. 21, 1345–1357.
T.S., White, A., Makkar, R., and Marban, E. (2009). Validation of the cardio- Olson, E.N. (2006). Gene regulatory networks in the evolution and develop-
sphere method to culture cardiac progenitor cells from myocardial tissue. ment of the heart. Science 313, 1922–1927.
PLoS ONE 4, e7195.
Saga, Y., Miyagawa-Tomita, S., Takagi, A., Kitajima, S., Miyazaki, J., and In-
Davis, R.L., Weintraub, H., and Lassar, A.B. (1987). Expression of a single oue, T. (1999). MesP1 is expressed in the heart precursor cells and required
transfected cDNA converts fibroblasts to myoblasts. Cell 51, 987–1000. for the formation of a single heart tube. Development 126, 3437–3447.
Garg, V., Kathiriya, I.S., Barnes, R., Schluterman, M.K., King, I.N., Butler, C.A., Snider, P., Standley, K.N., Wang, J., Azhar, M., Doetschman, T., and Conway,
Rothrock, C.R., Eapen, R.S., Hirayama-Yamada, K., Joo, K., et al. (2003). S.J. (2009). Origin of cardiac fibroblasts and the role of periostin. Circ. Res.
GATA4 mutations cause human congenital heart defects and reveal an inter- 105, 934–947.
action with TBX5. Nature 424, 443–447.
Srinivas, S., Watanabe, T., Lin, C.S., William, C.M., Tanabe, Y., Jessell, T.M.,
Ghosh, T.K., Song, F.F., Packham, E.A., Buxton, S., Robinson, T.E., Ronksley,
and Costantini, F. (2001). Cre reporter strains produced by targeted insertion
J., Self, T., Bonser, A.J., and Brook, J.D. (2009). Physical interaction between
of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4.
TBX5 and MEF2C is required for early heart development. Mol. Cell. Biol. 29,
2205–2218. Srivastava, D. (2006). Making or breaking the heart: from lineage determination
to morphogenesis. Cell 126, 1037–1048.
Gulick, J., Subramaniam, A., Neumann, J., and Robbins, J. (1991). Isolation
and characterization of the mouse cardiac myosin heavy chain genes. Takahashi, K., and Yamanaka, S. (2006). Induction of pluripotent stem cells
J. Biol. Chem. 266, 9180–9185. from mouse embryonic and adult fibroblast cultures by defined factors. Cell
126, 663–676.
Hong, H., Takahashi, K., Ichisaka, T., Aoi, T., Kanagawa, O., Nakagawa, M.,
Okita, K., and Yamanaka, S. (2009). Suppression of induced pluripotent Takeuchi, J.K., and Bruneau, B.G. (2009). Directed transdifferentiation of
stem cell generation by the p53-p21 pathway. Nature 460, 1132–1135. mouse mesoderm to heart tissue by defined factors. Nature 459, 708–711.
Hudon-David, F., Bouzeghrane, F., Couture, P., and Thibault, G. (2007). Thy-1 van Laake, L.W., Passier, R., Doevendans, P.A., and Mummery, C.L. (2008).
expression by cardiac fibroblasts: lack of association with myofibroblast Human embryonic stem cell-derived cardiomyocytes and cardiac repair in
contractile markers. J. Mol. Cell. Cardiol. 42, 991–1000. rodents. Circ. Res. 102, 1008–1010.
Ieda, M., Kanazawa, H., Kimura, K., Hattori, F., Ieda, Y., Taniguchi, M., Lee, Vierbuchen, T., Ostermeier, A., Pang, Z.P., Kokubu, Y., Sudhof, T.C., and
J.K., Matsumura, K., Tomita, Y., Miyoshi, S., et al. (2007). Sema3a maintains Wernig, M. (2010). Direct conversion of fibroblasts to functional neurons by
normal heart rhythm through sympathetic innervation patterning. Nat. Med. defined factors. Nature 463, 1035–1041.
13, 604–612. Zhang, J., Wilson, G.F., Soerens, A.G., Koonce, C.H., Yu, J., Palecek, S.P.,
Ieda, M., Tsuchihashi, T., Ivey, K.N., Ross, R.S., Hong, T.T., Shaw, R.M., and Thomson, J.A., and Kamp, T.J. (2009). Functional cardiomyocytes derived
Srivastava, D. (2009). Cardiac fibroblasts regulate myocardial proliferation from human induced pluripotent stem cells. Circ. Res. 104, e30–e41.
through beta1 integrin signaling. Dev. Cell 16, 233–244. Zhao, R., Watt, A.J., Battle, M.A., Li, J., Bondow, B.J., and Duncan, S.A.
Ivey, K.N., and Srivastava, D. (2006). The paradoxical patent ductus arteriosus. (2008). Loss of both GATA4 and GATA6 blocks cardiac myocyte differentiation
J. Clin. Invest. 116, 2863–2865. and results in acardia in mice. Dev. Biol. 317, 614–619.
Kattman, S.J., Huber, T.L., and Keller, G.M. (2006). Multipotent flk-1+ cardio- Zhou, Q., Brown, J., Kanarek, A., Rajagopal, J., and Melton, D.A. (2008). In vivo
vascular progenitor cells give rise to the cardiomyocyte, endothelial, and reprogramming of adult pancreatic exocrine cells to beta-cells. Nature 455,
vascular smooth muscle lineages. Dev. Cell 11, 723–732. 627–632.

386 Cell 142, 375–386, August 6, 2010 ª2010 Elsevier Inc.


Dendritic Function of Tau Mediates
Amyloid-b Toxicity in
Alzheimer’s Disease Mouse Models
Lars M. Ittner,1,6,* Yazi D. Ke,1,6 Fabien Delerue,1 Mian Bi,1 Amadeus Gladbach,1 Janet van Eersel,1 Heidrun Wölfing,1
Billy C. Chieng,2 MacDonald J. Christie,2 Ian A. Napier,2 Anne Eckert,3 Matthias Staufenbiel,4 Edna Hardeman,5
and Jürgen Götz1,*
1Alzheimer’s and Parkinson’s Disease Laboratory
2Neuropharmacology Laboratory
Brain and Mind Research Institute, University of Sydney, Sydney NSW 2050, Australia
3Neurobiology Research Laboratory, Psychiatric University Clinic, University of Basel, Basel CH-4025, Switzerland
4Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
5Neuromuscular and Regenerative Medicine Unit, University of New South Wales, Sydney NSW 2052, Australia
6These authors contributed equally to this work

*Correspondence: littner@med.usyd.edu.au (L.M.I.), jgoetz@med.usyd.edu.au (J.G.)


DOI 10.1016/j.cell.2010.06.036

SUMMARY where familial mutations have been identified in the tau-encod-


ing MAPT gene (Ballatore et al., 2007). Evidence that tau
Alzheimer’s disease (AD) is characterized by pathology in AD is induced by Ab comes from our previous
amyloid-b (Ab) and tau deposition in brain. It has observation that intracerebral Ab injections exacerbate hyper-
emerged that Ab toxicity is tau dependent, although phosphorylation of tau and NFT formation in transgenic mice
mechanistically this link remains unclear. Here, we that express FTD mutant P301L tau (Götz et al., 2001b). A similar
show that tau, known as axonal protein, has finding was obtained by crossing transgenic mice with NFT and
plaque pathologies (Lewis et al., 2001).
a dendritic function in postsynaptic targeting of the
Ab-plaque formation along with memory impairment and tau
Src kinase Fyn, a substrate of which is the NMDA pathology with increased phosphorylation, in the absence of
receptor (NR). Missorting of tau in transgenic mice deposition and NFT formation, has been reproduced in several
expressing truncated tau (Dtau) and absence of tau transgenic mouse lines that express human APP together with
in tau/ mice both disrupt postsynaptic targeting pathogenic mutations identified in familial AD (Götz and Ittner,
of Fyn. This uncouples NR-mediated excitotoxicity 2008; Hsiao et al., 1996; Mucke et al., 2000; Sturchler-Pierrat
and hence mitigates Ab toxicity. Dtau expression et al., 1997). In one of these, PDAPP, tau deficiency (tau/)
and tau deficiency prevent memory deficits and was shown to rescue lethality and memory deficits by an uniden-
improve survival in Ab-forming APP23 mice, a model tified mechanism (Roberson et al., 2007).
of AD. These deficits are also fully rescued with Tau is known as axonal protein that regulates MT stability
a peptide that uncouples the Fyn-mediated interac- and MT-dependent processes (Dixit et al., 2008; Drechsel
et al., 1992; Lee et al., 1988), while Ab likely exerts toxicity
tion of NR and PSD-95 in vivo. Our findings suggest
at the postsynapse (Selkoe, 2002; Shankar et al., 2008;
that this dendritic role of tau confers Ab toxicity at Zhao et al., 2006). Although in AD, hyperphosphorylated tau
the postsynapse with direct implications for patho- accumulates in the somatodendritic compartment of neurons
genesis and treatment of AD. (Ballatore et al., 2007), given the spatial separation it remains
unknown how tau is involved in mediating Ab toxicity when AD
INTRODUCTION is initiated.
Seizures characterize several APP transgenic strains (Minkevi-
Alzheimer’s disease (AD) is characterized by two hallmark ciene et al., 2009; Palop et al., 2007; Palop and Mucke, 2009) and
lesions, amyloid-b (Ab) plaques and neurofibrillary tangles have been associated with AD; the extent of their contribution to
(NFTs) (Ballatore et al., 2007). Ab is derived from the amyloid-b pathology, however, remains to be established (Minkeviciene
precursor protein (APP) by proteolytic cleavage (Haass et al., et al., 2009; Palop et al., 2007; Palop and Mucke, 2009). Excito-
1992; Selkoe, 1997). The major constituent of NFTs is tau, toxicity results from overactivation of N-methyl-D-aspartate
a microtubule (MT)-associated protein (Goedert et al., 1988). In (NMDA) receptors (NRs). Interestingly, tau reduction decreases
the course of AD, tau becomes phosphorylated, forming aggre- susceptibility to excitotoxic seizures in vivo, which may explain
gates that deposit as NFTs and neuropil threads (Geschwind, the concomitant improvement of the PDAPP phenotype (Rober-
2003). Tau can also form aggregates in the absence of an overt son et al., 2007). How tau prevents excitotoxic damage at
Ab pathology, for example in frontotemporal dementia (FTD), a molecular level is not understood.

Cell 142, 387–397, August 6, 2010 ª2010 Elsevier Inc. 387


A B wt ∆tau74 Figure 1. Truncated Tau Is Excluded from
Fyn binding site Dendrites in Dtau74 Mice
htau40 (A) The longest human tau isoform (htau40; 441 aa)
1 441
is composed of an amino-terminal projection
mThy1.2 ∆tau cx hp
domain (PD), the microtubule-binding (MTB)
1 255
domain with four repeats (gray boxes), and the car-
PD MTB C’ boxy-terminal tail (C0 ). Dtau transgenic mice
express only the PD of tau under control of the
wt ∆tau74 am neuronal mThy1.2 promoter. Dtau lacks the MTB
C kD domain and therefore the MT-binding and aggre-
50
Tau-5 37 gation properties of full-length tau, but contains
50
a Fyn binding site.
HT7 37 D ∆tau74 pR5 (B) Expression pattern of Dtau in Dtau74 brains.
Immunohistochemistry (IHC) with a human tau-
Gapdh specific antibody (HT7; brown) reveals Dtau
S expression within several brain regions, including
HT7 DAPI

4
(fold of wt)

wt
tau levels

3 ∆tau74 hippocampus (hp), cortex (cx), and amygdala (am).


2 (C) Western blotting of wild-type and Dtau74
1
D hippocampal extracts reveals endogenous murine
tau (50 kD) in all and Dtau (37 kD) only in transgenic
0
total endogenous samples. Quantification shows comparable levels
tau tau of endogenous tau, while endogenous tau and
Dtau levels add up to 2.4-fold increased total levels
in Dtau74 compared to WT mice.
(D) IHC of the hippocampal CA1 region reveals that in Dtau74 mice, Dtau localizes to the soma (S) but is excluded from dendrites (D), whereas expression of P301L
mutant full-length tau in pR5 mice results in a somato-dendritic localization of transgenic tau (HT7; reactive with Dtau and pR5 tau, but not endogenous tau,
in red). The scale bar represents 50 mm.
Error bars represent the standard error. See also Figure S1.

Tau interacts via its amino-terminal projection domain (PD) expressed Dtau throughout the brain (Figure 1B) at comparable
with the kinase Fyn (Figure 1A) (Lee et al., 1998). Fyn phosphor- levels, with line Dtau74 expressing the transgene at 1.4-fold
ylates the NR subunit 2 (NR2) to facilitate interaction of the NR higher levels than endogenous tau (Figure 1C). Expression of
complex with the postsynaptic density protein 95 (PSD-95) Dtau neither affected levels nor distribution of endogenous tau
(Nakazawa et al., 2001; Rong et al., 2001; Tezuka et al., 1999), (Figure 1C and Figures S1A–S1C available online). Consistent
linking NRs to synaptic excitotoxic downstream signaling (Salter with previous in vitro findings (Maas et al., 2000), Dtau localized
and Kalia, 2004). Disruption of the NR/PSD-95 interaction to the cell membrane, as indicated by coimmunostaining with
prevents excitotoxic damage in cultured neurons and a rat cadherin and subcellular fractionation of membranes (Figures
model of stroke, without affecting synaptic NMDA currents S1D and S1E). In AD and also full-length P301L mutant tau trans-
(Aarts et al., 2002). Reduction of Fyn in APP transgenic mice genic pR5 mice, tau is hyperphosphorylated and redistributed
prevents Ab toxicity, while overexpression enhances it (Chin into the somatodendritic compartment (Figure 1D) (Götz et al.,
et al., 2005; Chin et al., 2004). 2001a). In contrast to full-length tau, Dtau, while in the soma,
To address how tau confers Ab toxicity, we generated trans- was virtually excluded from dendrites (Figure 1D). In pR5
genic mice (Dtau74) that express only the amino-terminal projec- mice, tau becomes progressively hyperphosphorylated and
tion domain (PD) of tau and crossed them with Ab-forming insoluble, and eventually the mice develop NFTs. Surprisingly,
APP23 and tau/ mice. We found that tau has an important Dtau in Dtau74 mice is hardly phosphorylated at all (Figure S1F).
dendritic function, as in Dtau74 and tau/ mice, postsynaptic
Fyn localization is reduced, resulting in reduced NR phosphory- Postsynaptic Targeting of Fyn Is Tau Dependent
lation, destabilized NR/PSD-95 interaction, and protection from Different from full-length human tau in pR5 mice, in the absence
excitotoxicity. of an MTB domain, Dtau fails to interact with MTs, as determined
by MT precipitation from hippocampi (Figure 2A). However, Dtau
RESULTS contains motifs that mediate interaction with the Src kinase Fyn,
as shown in vitro (Lee et al., 1998). Accordingly, Fyn can be coim-
Truncated Tau Is Excluded from Dendrites munoprecipitated with Dtau from Dtau74 hippocampi in vivo,
Tau comprises an amino-terminal projection domain, an MT using a human tau-specific antibody (HT7) (Figure 2B). Immuno-
binding (MTB) domain that mediates interaction with MTs precipitation (IP) with tau-specific antibodies to epitopes not
(Butner and Kirschner, 1991; Lee et al., 1988) and is essential present on the Dtau construct reveals a significantly reduced
for tau aggregation (Crowther et al., 1989; Ksiezak-Reding and interaction of Fyn with endogenous tau (Figure 2B). Likewise,
Yen, 1991) and a carboxy-terminal tail region (Figure 1A). We IP with Fyn antibodies shows a reduced interaction with endog-
generated truncated (Dtau) transgenic mice that express the enous tau in Dtau74 mice (Figure 2B). Together, this suggests
projection domain of tau in neurons, intended to compete with a dominant negative effect of Dtau on the normal interaction of
functions of endogenous tau. Four phenotypically normal lines Fyn and endogenous tau. A similar effect on the Fyn/Tau

388 Cell 142, 387–397, August 6, 2010 ª2010 Elsevier Inc.


A C Figure 2. Dtau Impairs Tau-Dependent
p R 74

p R 74

74
Dendritic Targeting of the Src Kinase Fyn
au

au

au
5

5
pR

wt ∆tau74
∆t

∆t

∆t
human tau (A) Dtau from Dtau74 mice does not interact with
Tau-5 murine tau
S
microtubules. Endogenous murine tau, but not
∆tau Dtau, precipitates with microtubules in extracts
tubulin from Dtau74 mice. In contrast, both full-length
actin
D human and endogenous murine tau precipitate
extract no MT MT prec with microtubules in extracts from pR5 mice.

Fyn Drebrin
(B) Expression of Dtau results in a 74% ± 6%
B
74
u -/-
au

(n = 8; *p < 0.01) reduced interaction of Fyn with


5
pR
∆t
t

-/- -/-
ta
w

human tau
tau ∆tau74.tau endogenous murine tau (mtau) compared to the
mTau S wild-type (wt), as revealed by coimmunoprecipita-
murine tau
input

HT7 ∆tau tion (coIP) with antibodies to endogenous murine


Fyn tau (mTau). In Dtau74 mice, Fyn instead coimmu-
Gapdh noprecipitates with Dtau, as revealed by antibody
D HT7. Similarly, in pR5 mice, Fyn precipitates with
IP: mTau

mTau
full-length human tau (htau). CoIP with Fyn anti-
Fyn bodies predominantly pulled down endogenous
tau in WT, Dtau in Dtau74, and full-length human
mTau D
IP: HT7

40 tau in pR5 mice. No precipitation was observed


HT7 * * from tau/ tissue.
intensity (AFU)

wt
Fluorescence

30
Fyn * ∆tau74 (C) Fyn accumulates in cell bodies in Dtau74,
20 tau-/-
Fyn
* * tau/, and Dtau74.tau/ mice. While Fyn staining
IP: Fyn

∆tau74.tau-/-
mTau 10
* (red) colocalizes with dendritic drebrin (green) in
HT7 0 WT CA1 neurons, Fyn staining is evident in the
soma dendrites
soma (S) and is reduced in the dendrites (D) of
Dtau74 and tau/ neurons. The insets show
E wt ∆tau74 tau-/- F wt ∆tau74 tau-/- G wt ∆tau74 tau-/- higher magnification of dendritic staining. The
IP:Fyn Fyn
scale bar represents 50 mm.
Fyn
420
(D) Quantification of fluorescence intensity of Fyn
Gapdh p -Fyn PSD-95
staining in cell bodies and dendrites shows accu-
p531-Fyn
1.0 1.0
mulation of Fyn in cell bodies of Dtau74, tau/,
(fold of wt)
(fold of wt)

and Dtau74.tau/ mice (n = 15, *p < 0.0001).


Fyn
Fyn

0.5 0.5
* * (E) Total Fyn levels are not reduced in Dtau74 and
0 0 tau/ mice. Western blots of hippocampal
u -/-

u -/-
∆t w t

∆t wt
74

74

extracts from WT, Dtau74, and tau/ brains


au

au
ta

ta

show comparable levels of Fyn, normalized to


Gapdh (n = 6).
(F) Phosphorylation of activating (Y420) and inactivating (Y531) sites of immunopurified Fyn from WT, Dtau74, and tau/ brains is similar.
(G) Hippocampal synaptosomal preparations reveal reduced levels of Fyn in Dtau74 and tau/ postsynapses compared to the WT (n = 6, *p < 0.005).
Error bars represent the standard error. See also Figure S2.

interaction was obtained by overexpression of full-length tau in as determined by total and phosphorylation site-specific anti-
pR5 mice (Figure 2B). Given the dendritic exclusion of Dtau in Fyn antibodies (Figures 2E and 2F). To quantify changes in the
Dtau74 in contrast to full-length tau in pR5 mice (Figure 1D), subcellular localization of Fyn, we prepared synaptosomes
we speculated that the aberrant Dtau/Fyn interaction might from WT, Dtau74, and tau/ hippocampi. Consistent with the
affect the normal intracellular distribution of Fyn. Immunohisto- immunohistochemical findings of reduced postsynaptic target-
chemistry showed that Fyn colocalized with drebrin in wild- ing, levels of synaptic Fyn were reduced by 73% and 62% in
type (WT) brain, consistent with postsynaptic targeting, while in Dtau74 and tau/ mice, respectively, compared to WT controls
Dtau74 brains it accumulated in the soma, an effect enhanced (Figure 2G). Taken together, both the presence of Dtau and
by crossing of Dtau and tau/ (Figures 2C and 2D). Together absence of endogenous tau impair synaptic localization of Fyn.
with reduced dendritic Fyn staining, this suggests impaired
postsynaptic targeting of Fyn. To determine the role of tau in Uncoupled NMDA Receptors and PSD-95 in Dtau
dendritic localization of Fyn, we also analyzed tau/ mice. Fyn and tau/ Synapses
also accumulated in the soma (Figures 2C and 2D), suggesting The postsynaptic NR subunit NR2b is a known substrate of
that postsynaptic targeting of Fyn is, at least in part, tau depen- Fyn (Nakazawa et al., 2001). NR2b phosphorylation at Y1472
dent. This is consistent with reduced localization of Fyn-DsRED strengthens the NR/PSD-95 interaction (Rong et al., 2001). In
in primary hippocampal neurons either from tau/ mice or mice both Dtau74 and tau/ mice, Y1472 phosphorylation is signifi-
coexpressing Dtau (Figure S2). Interestingly, further truncation of cantly reduced compared to the WT, while total levels of NR1,
Dtau shows that the Fyn-interactive motif, PXXP (Lee et al., NR2a, and NR2b are unaffected (Figure 3A). To determine
1998), is critical for Fyn localization. whether this affects the stability of NR/PSD-95 complexes,
Despite changes in the localization of Fyn, its total levels we performed coimmunoprecipitations (coIPs). Markedly less
and activity were comparable in Dtau74, tau/, and WT mice, NR1, NR2a, and NR2b coimmunoprecipitated with PSD-95

Cell 142, 387–397, August 6, 2010 ª2010 Elsevier Inc. 389


tau-/- total pH6 pH8 SDS Figure 3. Destabilized NMDA Receptors in
A wt ∆tau74 C
the Postsynaptic Density of Dtau74 and
NR1 Gap43
tau/ Mice

Y1472 (fold of wt)


NR2a 1.0 SNAP25
(A) Levels of NR subunits NR1, NR2a and NR2b,
NR2b * PSD-95
0.5 * and PSD-95 are comparable in extracts from WT,
Y1472 mTau Dtau74, and tau/ brains, whereas phosphoryla-
0
PSD-95 wt Fyn tion of NR2b at the Fyn site, Y1472, that is known
∆tau74
tau-/- NR1 to stabilize NR/PSD-95 complexes (Roche et al.,
Gapdh
NR2a 2001), is significantly reduced in Dtau74 and
tau/ than in the WT (n = 6, *p < 0.005).
NR2b
(B) PSD-95 antibodies coimmunoprecipitate much
B wt ∆tau74 tau-/- D wt ∆tau74 tau-/- less NR subunits NR1, NR2a, and NR2b from
IP:PSD-95 1.0 NR1 Dtau74 and tau/ than from WT hippocampi.
NR1

0.5 * * NR2a
Similarly, coimmunoprecipitation (coIP) of Fyn
NR1 0
1.0 with PSD-95 is reduced in Dtau74 and tau/
pH8
NR2a

NR2a NR2b
0.5 * * compared to WT hippocampi, while that of nNOS
NR2b 0 Fyn
1.0 and Homer was unaffected. Endogenous murine
NR2b

mTau 0.5
* * SNAP25 Tau (mTau) coprecipitates with PSD-95 from WT
WB 0
HT7 1.0
NR1 hippocampi, while much less mTau, but no Dtau
mTau

0.5 * (HT7), is recovered from Dtau74 hippocampi.


Fyn NR2a
mTau is absent in tau/ coIPs, consistent with
0
1.0
nNOS * * SDS
Fyn

0.5 NR2b tau deficiency. (n = 3, *p < 0.005.)


0
Homer Fyn (C) Sequential extraction of synaptosomes. Puri-
PSD-95 fied WT synaptosomes were further fractionated
with buffers of increasing stringency (pH6 <
E wt ∆tau74 tau-/- ∆tau74.tau-/- G pH8 < SDS), to purify proteins that are stably asso-
6 80
ciated with the PSD (Phillips et al., 2001). Brain
EPSC (NMDA/AMPA)

extracts (total) are loaded for comparison. NR


Mini EPSC (pA)

5
60
4 subunits NR1, NR2a and NR2b, PSD-95, tau,
3 40
and Fyn are purified in the SDS fraction, consistent
2
1
20 with strong anchoring in the PSD. Soluble proteins,
50 pA
0 0 such as GAP43 and proteins that are not (such as
u -/-

u -/-

u -/-

u -/-

50 ms
t

74

74
w

SNAP25) or less stably associated with the PSD,


ta

.ta

ta

.ta
au

au

H
74

74
∆t

∆t

are extracted with less stringent pH 6 and pH 8


au

au

100 5
∆t

∆t

F wt ∆tau74 tau-/- ∆tau74.tau-/- buffers, respectively.


EPSC (% NR2b)

Frequency (s-1)

75 4

50 pA
3 (D) SDS fractions from synaptosomes show that
50
2 stable anchoring of NRs in the PSD is reduced in
50 ms
25 1 Dtau74 and tau/ mice. While NRs are recovered
0 0 in the SDS fraction of WT synaptosomes, they are
u -/-

u -/-

u -/-

u -/-
t

74

74
w

primarily found in the pH 8 and hardly at all in SDS


ta

.ta

ta

.ta
au

au
74

74
∆t

∆t

fractions from Dtau74 and tau/ mice.


au

au
∆t

∆t

(E) Representative traces of AMPAR- (gray) and


NR- (black) mediated components of electrically
evoked (e) EPSCs in CA1 hippocampal neurons from WT, Dtau74, tau/, and Dtau74.tau/ mice (average of 12 sweeps per neuron) normalized with AMPAR-
mediated component. Neurons were voltage clamped and held at +40 mV. AMPAR-mediated eEPSCs are inverted for clarity. There is no significant difference of
NMDA/AMPA ratios between genotypes (n = 19–20).
(F) Representative traces of eEPSCs (average of 12 sweeps per neuron) separating total NMDAR-mediated and NR2b subunit-mediated components
(black traces, total NR eEPSC minus component in CP-101,606 [5 mM]) normalized to the amplitude of the total NR eEPSC. Neurons were voltage clamped
at +20 mV. NR2b EPSCs were obtained by subtraction of EPSCs generated in CP101 606 (5 mM) from total NR-mediated EPSCs, i.e., before CP applications.
There is no significant difference in the percentage of NR2b component between genotypes (n = 18–20).
(G and H) Mean amplitude (G) and rate (s1) (H) of AMPAR-mediated mEPSCs (recorded in 1 mM TTX) were unaffected in Dtau74, tau/, and Dtau74.tau/ mice
(n = 10–12).
Error bars represent the standard error. See also Figure S3.

from Dtau74 and tau/ compared to WT extracts, consistent buffers of increasing stringency (Phillips et al., 2001). In line with
with a decreased interaction of NR and PSD-95 in both strains a strong PSD association in WT synaptosomes, NR subunits
(Figure 3B). The PSD-95-interacting proteins Homer and were mostly found in the SDS fraction (Figure 3C and 3D). In
nNOS, however, were coimmunoprecipitated to a similar extent contrast, they were markedly reduced in Dtau74 and tau/,
from WT, Dtau74, and tau/ brains, suggesting intact interac- appearing instead in earlier fractions, suggestive of a weakened
tions. The NR/PSD-95 interaction facilitates stable anchoring anchoring in the PSD (Figure 3D). Interestingly, endogenous
of NRs in the postsynaptic density (PSD) (Roche et al., 2001). tau that was enriched by synaptosome preparation, recovered
Therefore, we next extracted purified synaptosomes from WT, in WT SDS fractions and coimmunoprecipitated with PSD-95
Dtau74, and tau/ mice that show similar levels of NR subunits, from WT, and to a lesser degree from Dtau74, brains (Figures
but reduced NR2b phosphorylation at Y1472 (Figure S3A), using 3B and 3C and Figure S1C).

390 Cell 142, 387–397, August 6, 2010 ª2010 Elsevier Inc.


A B Figure 4. Dtau Expression Improves
4
100 APP23.∆tau74.tau-/- Memory and Ameliorates Premature
APP23.∆tau74.tau+/-
Survival (% alive mice)

APP23.tau-/- Mortality of APP23 Mice

Errors (T-maze)
3
APP23.tau+/-
APP23.∆tau74 (A) APPswe transgenic APP23 mice (n = 76) present
Acquisition
75 2 with a pronounced premature mortality that is * 2h
24h
**
** ** ameliorated by reducing tau levels in APP23.tau+/
**
1
**** ** **
** ** ** (n = 41, p < 0.001) and even more in APP23.tau/
APP23
mice (n = 108, p < 0.0001). Expression of Dtau
50 0
improves the survival of APP23.Dtau74 mice

-/-

-/-

u -/-
t

74

74
0 2 4 6 8 10

P2
u

u
au

au
ta

ta

.ta
Age (months) (orange, n = 43, p < 0.01) similar to APP23.tau+/.

AP

3.
∆t

∆t

74
P2
3.

au
P2

AP

∆t
AP
-/-

Interestingly, combination of Dtau expression with

3.
P2
C D 1.5
u
.ta

AP
(fold of APP23)
74

74

hAPP mRNA

tau reduction completely rescues APP23.Dtau74.-


-
u -/
au

au

1.0
∆t

∆t
ta

tau+/ (purple, n = 38, p < 0.0001) and


3.

3.

3.
3
P2

P2

P2

P2

0.5
G APP23
AP

AP

AP

AP

APP23.Dtau74.tau/ (red, n = 52, p < 0.0001)


t
w

0.0
APP23.∆tau74
Fyn
mice from lethality.
NR2b E 300 90
(B) Improved memory acquisition of APP23.D
Aβ1-40 (ng/mg)

Aβ 1-42 (ng/mg)

Y1472 200 60
tau74, APP23.tau/, and APP23.Dtau74.tau/
PSD-95 100 30
APP23.tau APP23.∆tau74.tau compared to APP23 mice in the T maze, 2 and 24 -/- +/-
0 0
2.0 3.0 hr after a five-trial acquisition, at 8 months of age.
Y1472 (fold of wt)
Fyn (fold of wt)

*
1.5
2.0 While WT, Dtau74, and tau/ mice only make few
1.0
*
* 1.0
* * F 100
errors during the trials, memory deficits of APP23
per section

0.5 **
Plaques

** 75
0
0
25
mice are obvious from the continuously high
-/-
APP23 APP23.tau
APP23.∆tau74 APP23.∆tau74.tau 0 +/- numbers of errors made during the entire test. In
contrast, both APP23.Dtau74, APP23.tau/, and
APP23.Dtau74.tau/ mice presented with WT-
like numbers of errors (n = 8, *p < 0.05, **p < 0.01).
(C) In synaptosomal preparations obtained from 4-month-old APP23, both Fyn levels and NR2b phosphorylation at Y1472 are increased as compared to wild-
type (wt) mice (n = 6, *p < 0.05). However, in synaptosomes from APP23.Dtau74 and APP23.tau/, and even more in APP23.Dtau74.tau/, both levels of Fyn
and NR2b phosphorylation are significantly lower than in APP23 mice (n = 6, *p < 0.05, **p < 0.01). Representative western blots from three independent exper-
iments are shown.
(D–G) Dtau expression and tau deficiency do not affect APP mRNA expression, Ab levels, or plaque burden.
(D) Levels of APP mRNA are not altered in APP23 mice in the presence of Dtau or when tau is absent (tau/).
(E) Ab1–40 and Ab1–42 levels are comparable in APP23, APP23.Dtau74, and APP23.tau/ mice.
(F and G) Thioflavine S staining (green) reveals Ab plaques (arrows; insets) at similar numbers (F) and with similar morphology (G) in APP23 mice, independent of
coexpression of Dtau or tau reduction.
Error bars represent the standard error. See also Figure S4.

The organization of NRs within the PSD is important for coor- contribute to toxicity in mice; however, primary disease-related
dinated signal transduction (Kim and Sheng, 2004). Hence, alter- effects are attributed to Ab, as suggested by reverted deficits
ations of NRs in Dtau74 and tau/ mice may affect synaptic in Ab-immunized APP models (Röskam et al., 2010) and absence
currents. Therefore, we determined excitatory postsynaptic of seizure-induced hippocampal remodeling in APP transgenic
currents (ESPCs) in acute hippocampal slices from WT, mice with low Ab levels (Palop et al., 2007). Excitotoxicity has
Dtau74, tau/, and Dtau74.tau/ mice. In Dtau74, tau/, been linked to premature lethality in APP transgenic mice (Chis-
and Dtau74.tau/ mice, we found np significant changes in hti et al., 2001; El Khoury et al., 2007; Leissring et al., 2003;
synaptic currents (Figure 3E). Similarly, no significant reduction Roberson et al., 2007). Hence, we speculated that in Dtau74
emerged in the contribution of NR2b-containing NRs to ESPCs alterations in NR/PSD-95 interaction might similarly rescue the
in Dtau74, tau/, and Dtau74.tau/ mice (Figure 3F). Baseline early lethality that characterizes APPswe mutant APP23 mice
miniature amplitudes and frequency were also comparable (Figures S4A and S4B) (Sturchler-Pierrat et al., 1997). APP23
(Figures 3G and 3H and Figures S3B–S3D). Taken together, mice have high Ab levels already at a very young age (Kuo
these data indicate that both expression of Dtau or tau deficiency et al., 2001; Van Dam et al., 2003), eventually forming plaques
reduces the interaction of NRs with PSD-95 without affecting and presenting with neuronal loss and memory deficits (Calhoun
synaptic NR levels and currents. et al., 1998; Kelly et al., 2003; Sturchler-Pierrat et al., 1997).
When we crossed APP23 either with Dtau74 or tau/ mice
Dtau Expression Prevents Premature Lethality (Figure S4C), this caused both a significantly delayed onset of
and Memory Deficits in APP23 Mice mortality and an improved overall survival (Figure 4A). Whereas
It has been shown previously that perturbing the interaction of any rescue (either on a tau/ background or by expressing
NRs with PSD-95 had no effect on NR-mediated currents but Dtau) was partial, expression of Dtau on a heterozygous or
reduced the resilience of neurons to NMDA-mediated excitotox- homozygous tau-deficient background rescued lethality
icity (Aarts et al., 2002). Interestingly, excitotoxicity has been completely, suggesting complementary beneficial effects of
proposed to contribute to Ab toxicity in PDAPP mice, which tau deficiency and Dtau expression on survival (Figure 4A).
was reduced when the mice were crossed onto a tau/ back- In contrast, crossing of APP23 mice with pR5 mice with an
ground (Roberson et al., 2007). APP expression per se may increased dendritic accumulation of tau (Figure 1D) (Götz et al.,

Cell 142, 387–397, August 6, 2010 ª2010 Elsevier Inc. 391


2001a), resulted in increased premature lethality, with no survival
beyond 4 months of age (Figure S4D). Interestingly, both Fyn
A 7 B 8 *
levels and Y1472 phosphorylation of NR2b are increased in 6
*

Seizure Severity
pR5 synaptosomes (Figure S4E). Because of possible confound- 6

Latency (min)
5
ing effects of APP overexpression and Ab formation in APP **
4
mutant mouse strains, we used also primary neurons treated ** *** 4
with Ab, in the absence of APP overexpression, as a model. In 3
tau/ and Dtau74-expressing neurons, acute Ab toxicity was 2
wt
2 ∆tau74
markedly reduced (Figure S2E). Interestingly, deletion of the tau-/-
Fyn-interacting motif, PXXP (Lee et al., 1998), from Dtau abro- 1 ∆tau74.tau-/-
gated the protective effect. 0 0

-/-

u- /-
∆t wt
74
We next determined whether Dtau expression or tau reduction 1 2 3 4 5 6 7

au tau
.ta
au
Seizure Severity

74
also improves memory functions in APP23 mice. Memory defi-

∆t
cits were both improved to WT levels in APP23.Dtau74
and APP23.tau/ mice using the water T maze (Figure 4B).
Consistent with the findings in Dtau and tau/ mice, both
C 7 D 8 APP23 * *
synaptic Fyn levels and NR2b phosphorylation were reduced 6 *
APP23.∆tau74 *

Seizure Severity
in APP23.Dtau74 and APP23.tau/ and even more so in ** 6 *

Latency (min)
5
APP23.Dtau74.tau/ synaptosomes, while they were increased
in APP23 compared to WT brains (Figure 4C). Interestingly, in 4 ***
4
APP23 mice, neither Dtau expression nor tau reduction affected 3
human APP messenger RNA (mRNA) levels (Figure 4D), Ab levels
2 2
(Figure 4E), or plaque burden (Figures 4F and 4G). Similarly,
1 APP23.tau-/-
phosphorylation of endogenous tau was comparable in APP23 APP23.∆tau74.tau-/-
and APP23.Dtau74 mice (data not shown). Taken together, 0 0
expression of Dtau in APP23 or crossing of APP23 with tau/
74 u -/-
u- /-
AP .∆t 23
3. 2 74

1 2 3 4 5 6 7
au ta
.ta
3 P
P2 P au
P2 AP

Seizure Severity
∆t 3.

mice reduces Fyn-mediated NR2b phosphorylation, attenuates


premature mortality, and improves memory deficits without
AP

changing Ab levels or plaque load.


AP

Dtau Reduces Susceptibility to Excitotoxic Seizures E 7 F 8 wt + vehicle


wt + MK801
Ab-induced aberrant excitatory neuronal activity may contribute 6 *
Seizure Severity

to the deficits that characterize AD mouse models (Busche et al., 6


Latency (min)

5
2008; Palop and Mucke, 2009). APP23 mice show spontaneous *
seizures (Lalonde et al., 2005), similar to other APP transgenic 4
4
strains (Minkeviciene et al., 2009; Palop et al., 2007; Palop and 3
Mucke, 2009). Hence, reduced mortality of APP23.Dtau74 and
2
APP.tau/ mice may be related to a reduced susceptibility to 2
APP23 + vehicle
excitotoxic seizures. We therefore first induced convulsions in 1
APP23 + MK801
Dtau74, tau/, Dtau74.tau/, and WT mice using the g-amino- 0 0
butyrate (GABA) antagonist pentylenetetrazole (PTZ). Seizure wt APP23 1 2 3 4 5 6 7
severity was significantly reduced in Dtau74, tau/, and vehicle MK801
Seizure Severity
Dtau74.tau/ compared to the WT (Figure 5A), while the latency
to develop severe convulsion increased (Figure 5B). Next, we Figure 5. Dtau Expression Reduces Susceptibility to Excitotoxic
induced seizures in APP23, APP23.Dtau74, APP23.tau/, and Seizures
APP23.Dtau74.tau/ mice. APP23 mice presented with a (A) When excitotoxic seizures were induced by i.p. injection of PTZ (50 mg/kg),
reduced convulsion latency and showed the most severe seizure mean seizure severity was significantly reduced in both Dtau74, tau/, and
Dtau74.tau/ compared to WT mice (n = 10, **p < 0.01, ***p < 0.001).
response, with the lowest survival rate (1/11) and all mice reach-
(B) Similarly, the latency to more severe seizure stages is increased in Dtau74,
ing status epilepticus (n = 11) (Figure 5C). However, when APP tau/, and Dtau74.tau/ mice.
expression was combined with Dtau expression or tau defi- (C) In APP23 mice, PTZ-induced seizures are mostly lethal (10 of 11), whereas
ciency, this significantly decreased seizure severity, reduced in APP23.Dtau74, APP23.tau/, and APP23.Dtau74.tau/ seizure severity is
fatality, and increased convulsion latency (Figures 5C and 5D). markedly reduced (n = 10, *p < 0.05, **p < 0.01, ***p < 0.001).
The double mutant Dtau74.tau/ prevented severe seizures (D) APP23.Dtau74, APP23.tau/, and APP23.Dtau74.tau/ mice show an
better than Dtau74 or tau/ alone, on both WT and APP23 back- increased latency to more severe seizures compared to APP23 mice.
(E and F) Pretreatment of WT or APP23 mice with MK801 (0.1 mg/kg) reduced
grounds, in agreement with the survival data (Figure 4A). Inter-
seizure severity (n = 8, p < 0.05) (E) and increased latency to more severe
estingly, we found a similar degree of protection from PTZ- seizures (F).
induced seizures as in Dtau74, tau/, APP23.Dtau74, or Error bars represent the standard error.
APP23.tau/ mice when we pretreated WT and APP23 mice,

392 Cell 142, 387–397, August 6, 2010 ª2010 Elsevier Inc.


Figure 6. Peptide-Driven Uncoupling of the

AA

9c
t

2B
2B
en

A D E NR/PSD-95 Interaction Reduces Ab Toxicity

74
m

R
R
t

u -/-
en

-N
-N
at

au
is
tre

ys
and Improves Survival and Memory of
m

t
t

∆t
Ta
Ta

ta
h

w
at

al
e-

as
7
tre

an
PI
pr

PSD-95 APP23 Mice


1h 1h 5’ 6
24h NR1 (A) Twenty-day-old primary cortical neurons were

Seizure Severity
5

input
control NMDA Aβ
B PI/Hoechst Tau-54 *
pretreated with 100 nM Tat-NR2B9c peptide,
which disrupts the NR/PSD-95 interaction (Aarts
vehicle

3
Gapdh
2 et al., 2002), prior to treatment with the toxins
PSD-95 NMDA, Ab, H2O2, and staurosporine. Twenty-
1

IP
Tat-NR2B9c Tat-NR2BAA

NR1 four hours after treatment, cell death was deter-


0

t-N AA

9c
mined by propidium iodide (PI) uptake. Control

2B
Ta 2B
R
R
cells were pretreated with vehicle or 100 nM Tat-

-N
F

t
p

Ta
m
NR2BAA (inactive peptide).

ed
d
pu

ge

ov
ch mp

re mp
an
B

.v.

m
O
(B and C) Tat-NR2B9c (bottom row) significantly

pu

pu
i.c
D

6wks 28d 28d H reduces toxicity of NMDA and Ab to cortical


C 4mo 8mo neurons, as indicated by lower numbers of PI-
100 ## ## ## control G 4
positive cells (red; arrows) compared to pretreated
## ## ## H2 O 2 *
##
## ##
## Staurosporine 100 vehicle (top row) or Tat-NR2BAA (middle row)

Errors (T-maze)
## 3
Percentage survival

80 ## NMDA
Aβ (1μM) Tat-NR2B9c controls. Nuclei were stained with Hoechst
% of dying cells

A β (0.1μM) 75
#
Aβ (1μM) + NMDA (n=17)
60 Aβ (0.1μM) + NMDA 2 (blue). Treatment with H2O2, staurosporine,
# Tat-NR2BAA
50 (n=9) NMDA, Ab, or NMDA/Ab causes significant cell
40 * **
pump
1st pump

* 25 aCSF (n=11) 1 death (#p < 0.005, ##p < 0.0001), which for
* * NMDA- and Ab-treated neurons is reduced by
nd

20 *
2

* 0 0 pretreatment with Tat-NR2B9c, but not for H2O2-


0 30 60 240 ed

9c

SF
0 days after implantation and staurosporine-treated neurons (*p < 0.05,
2B
at

aC
vehicle Tat-NR2BAA Tat-NR2B9c
tre

R
-N
un

**p < 0.01). One hundred cells each were counted


t
Ta

in three independent experiments. The scale bar


represents 25 mm.
(D) Immunoprecipitation (IP) with a PSD-95 antibody from hippocampus of WT mice that were i.c.v. infused with Tat-NR2B9c and Tat-NR2BAA for 1 week, and
from untreated WT, Dtau74, and tau/ mice. Less NRs were coimmunoprecipitated upon Tat-NR2B9c, but not Tat-NR2BAA treatment. The reduction was
comparable to Dtau74 and tau/ mice.
(E) One week of i.c.v. infusion of Tat-NR2B9c reduced PTZ-induced seizure severity significantly, compared to inactive Tat-NR2BAA (n = 10, *p < 0.01).
(F) APP23 mice treated with vehicle (artificial cerebrospinal fluid [aCSF]) alone or together with Tat-NR2B9c or Tat-NR2BAA, using osmotic mini pumps. DOB,
date of birth.
(G) Survival of APP23 mice upon i.c.v. delivery of Tat-NR2B9c (n = 17) is markedly improved compared to vehicle (aCSF)-treated (n = 11, p < 0.01) and Tat-
NR2BAA-treated (n = 9, p < 0.05) controls. Gray boxes indicate time of drug delivery from two consecutively implanted pumps.
(H) Tat-NR2B9c-treated APP23 mice show markedly improved memory functions at 4 and 8 months after initiating treatment, compared to aCSF-treated or age-
matched untreated APP23 mice (n = 8, n = 4 for aCSF, *p < 0.05, **p < 0.01).
Error bars represent the standard error.

respectively, with the NR-antagonist MK801 (Figures 5E and 5F). death, consistent with shared signaling pathways mediating their
Hence, reduced susceptibility to excitotoxicity is consistent with toxicity (Figures 6B and 6C). Cell death induced by NMDA and
a reduced NR contribution and may contribute to reduced Ab, both separate and in combination, was significantly reduced
mortality in APP23 mice in the presence of Dtau or absence of by preincubation with Tat-NR2B9c, but not when induced by
endogenous tau. hydrogen peroxide or staurosporine (Figures 6B and 6C). Tat-
NR2BAA had no protective effects. Hence, perturbing the NR/
Targeted Uncoupling of NR and PSD-95 Prevents PSD-95 interaction with Tat-NR2B9c ameliorates Ab-mediated
Premature Death and Memory Seficits in APP23 Mice toxicity in vitro.
Provided that disturbed NR/PSD-95 complexes with a reduced Next, we tested in vivo whether APP23 mice would also benefit
dendritic Fyn localization in Dtau74 and tau/ mice contribute from treatment with Tat-NR2B9c. A single dose of this peptide
to improved memory functions and survival of APP23.Dtau74 has previously been shown to confer virtually complete protec-
and APP23.tau/ mice, targeted perturbation of the NR/PSD- tion from excitotoxic damage in a rat model of stroke (Aarts
95 interaction, independent of tau or Fyn, should also decrease et al., 2002). First, we determined whether sufficient NR/
Ab toxicity. Therefore, we treated primary cortical cultures with PSD-95 uncoupling was achieved by intracerebroventricular
the Tat-NR2B9c peptide composed of carboxy-terminal amino (i.c.v.) Tat-NR2B9c treatment, using osmotic minipumps. We
acids of NR2b (including Y1472) fused to a HIV1-Tat peptide to delivered either Tat-NR2B9c or Tat-NR2BAA for 1 week and
achieve cell membrane permeability (Figure 6A). Tat-NR2B9c then performed coIP with a PSD-95 antibody (Figure 6D). This
has been shown previously to protect from NMDA-induced exci- revealed a reduced NR/PSD-95 interaction upon Tat-NR2B9c,
totoxicity (Aarts et al., 2002; Kornau et al., 1995). As a negative but not Tat-NR2BAA, treatment. The level of reduction was
control, we included Tat-NR2BAA in which critical amino acids similar to that found in Dtau74 and tau/ brains (Figure 6D).
were replaced by alanine (Aarts et al., 2002; Kornau et al., Sufficient uptake of peptides by the brain was further confirmed
1995). NMDA and Ab both induced pronounced cell death, while by protection from PTZ-induced seizures by Tat-NR2b9c, but
a combined NMDA/Ab treatment did not further increase cell not Tat-NR2BAA (Figure 6E). Next, we implanted minipumps

Cell 142, 387–397, August 6, 2010 ª2010 Elsevier Inc. 393


A wt B ∆tau74 C tau-/- Figure 7. Simplified Scheme of the
Proposed Mechanism Underlying Reduced
Ca2+ Ca2+ Ca2+ Excitotoxicity in Dtau74 and tau/ Mice
NMDAR NMDAR NMDAR Compared to the Wild-Type
NR2b NR2b NR2b
(A) Postsynaptic NMDA receptors (NRs) are het-
PSD PSD PSD eromeric complexes predominantly formed by
P
Fyn
P
Fyn
P
Fyn
subunits NR1, NR2A, and NR2B. The Src kinase
PSD-95 Tau PSD-95 Tau PSD-95
Fyn localizes to the postsynapse in a tau-depen-
dendritic spine

dent manner and associates with the postsynaptic


density (PSD; gray box), where it phosphorylates
(P) the NR subunit NR2b at Y1472 in the extreme
carboxy terminus. This phosphorylation facilitates
the interaction of NRs with the scaffolding protein
PSD-95. This interaction increases the stability of
NRs within the PSD and couples NRs to excito-
∆tau
toxic downstream signaling (skull). NR-mediated
currents (ESPC trace), however, do not depend
on this NR/PSD-95 interaction. Whether tau is
associated with the PSD via Fyn or another inter-
action partner remains to be elucidated.
(B) In Dtau74 mice, Dtau is excluded from entering dendrites. Since Fyn interacts with Dtau (red bar) in the cell body of neurons, it is therefore trapped and less
localized to dendrites. Also, phosphorylation of NR2b and the interaction of NRs and PSD-95 are markedly reduced. Hence, excitotoxic downstream signaling is
uncoupled from NRs and their stability within the PSD is reduced. As NR-mediated currents are not dependent on this interaction, they are not affected.
(C) As for Dtau74 mice, in tau/ mice, tau-dependent localization of Fyn to the postsynapse is also markedly reduced. NR2b phosphorylation and the interaction
of NRs and PSD-95 are decreased. Thus, excitotoxic downstream signaling is uncoupled from NRs, and their stability within the PSD is reduced. Again, NR-medi-
ated currents are not affected.

into 6-week-old APP23 mice for i.c.v. delivery of artificial cere- result in ‘‘trapping’’ of Fyn in the soma. Tau/ mice, in compar-
brospinal fluid (aCSF), with and without Tat-NR2B9c or Tat- ison, show a similar accumulation of Fyn, suggesting that post-
NR2BAA (Figure 6F). Mice in the aCSF and Tat-NR2BAA control synaptic Fyn targeting requires tau. This difference in mediating
groups died frequently (7 of 11 and 4 of 9, respectively), whereas aberrant sorting of Fyn between Dtau74 and tau/ mice
only 1 of 17 mice died in the Tat-NR2B9c group (Figure 6E). (Figure 7) may explain the additive effects on seizure suscepti-
Finally, we tested whether Tat-NR2B9c-treatment has long- bility and survival in Dtau.tau/ crosses. Reduced levels of
term effects on memory in APP23 mice. The T maze revealed postsynaptic Fyn in Dtau74 and tau/ mice are associated
comparable memory deficits in age-matched aCSF-treated with reduced phosphorylation of the Fyn-substrate NR2b at
and untreated APP23 mice (Figure 6F). However, treatment Y1472. Consistent with a critical role of Y1472 phosphorylation
with Tat-NR2B9c resulted in a significantly improved perfor- in facilitating the interaction of NRs with PSD-95 (Rong et al.,
mance. Thus, perturbing NR/PSD-95 interaction is sufficient 2001), this complex is reduced and destabilized in Dtau74 and
to prevent premature lethality and memory deficits in APP23 tau/ brains. Whether the Fyn-mediated stabilization of NR/
mice. PSD-95 complexes in the PSD under physiological conditions
involves a direct interaction with tau and what the exact mecha-
DISCUSSION nism(s) of tau-mediated dendritic Fyn localization are remains to
be established.
Dendritic Localization of Fyn Is Tau-Dependent In a rat model of stroke, targeted disruption of the NR/PSD-95
Our data reveal a dendritic function of the ‘‘axonal’’ protein tau, in interaction prevented excitotoxic damage and reduced the
targeting the kinase Fyn to the dendrite (Figure 7). We also found lesion size (Aarts et al., 2002). Consistent with this, reduced
an association of tau with the PSD complex by using coIP, PSD NR/PSD-95 complexes in Dtau74 and tau/ mice were associ-
purification, and immunohistochemistry with enhanced antigen ated with a reduced susceptibility to excitotoxicity. Interestingly,
retrieval. It is important to note that levels of tau in the dendritic NR-mediated currents were not affected in Dtau74 and tau/
compartment are much lower than in axons, suggesting that mice, which is in line with normal synaptic activity upon treat-
under physiological conditions a major function of tau is in axonal ment with Tat-NR2B9c (Aarts et al., 2002). Normal NR-mediated
MT stabilization and regulation of MT-dependent processes currents in Dtau74 and tau/ mice may be explained by
(Dixit et al., 2008; Weingarten et al., 1975). Here, we show that reduced, but not totally depleted, synaptic Fyn in Dtau74 and
the additional role of tau in dendrites becomes pivotal in disease, tau/ mice, comparable to the situation in heterozygous fyn-
in particular in mediating early Ab toxicity. deficient mice that have no overt deficits (Yagi et al., 1993), while
In both Dtau74 and tau/ mice, dendritic targeting of Fyn is in homozygous fyn-deficient mice these are pronounced (Grant
significantly reduced, as revealed by immunohistochemistry et al., 1992).
and synaptosomal purification and confirmed in primary
neurons. In Dtau74 mice, this is due to a competition of Dtau Dtau and tau/ Prevent Deficits of APP23 Mice
with endogenous tau in the interaction with Fyn. Both the abun- Excitotoxicity is increasingly recognized as a mechanism of how
dance of Dtau in the cell body and its exclusion from dendrites Ab exerts toxicity in AD. Accordingly, we found that crossing

394 Cell 142, 387–397, August 6, 2010 ª2010 Elsevier Inc.


of Dtau74 and tau/ mice, both characterized by reduced EXPERIMENTAL PROCEDURES
susceptibility to excitotoxicity, with Ab-forming APP23 mice
Animals
ameliorated premature mortality and memory deficits of APP23
APP23 and pR5 transgenic and tau/ mice have been generated previously
mice. In contrast, early lethality was more pronounced in (Götz et al., 2001a; Sturchler-Pierrat et al., 1997; Tucker et al., 2001). The
APP23 mice crossed with pR5. Similarly, tau deficiency or Dtau generation of Dtau74 mice is described in the Extended Experimental Proce-
expression conferred protection from Ab-induced toxicity in dures. Two- to three-month-old mice were analyzed in age- and sex-matched
primary neuronal cultures. However, Ab levels and plaque forma- groups, unless stated otherwise. All animal experiments were approved by the
tion, as well as endogenous tau phosphorylation (in APP23.D Animal Ethics Committee of the University of Sydney.
tau74), were comparable in APP23 mice, suggesting an alterna-
Histology, Western Blotting, IP, and Synaptosome Preparation
tive mechanism for protection. Interestingly, in APP23 mice we
Detailed protocols are provided in the Extended Experimental Procedures.
found both increased postsynaptic Fyn and Y1472 phosphoryla-
tion of NR2b that was completely reverted in APP23.Dtau74 and Electrophysiology
APP23.tau/ mice. Further reduction of post-synaptic Fyn in Electrophysiological recording were done in acute hippocampal slices
APP23.Dtau74.tau/ mice suggests additional tau-indepen- obtained from 4- to 8-week-old wild-type, Dtau74, tau/, and Dtau74.tau/
dent mechanisms in dendritic Fyn localization, which are partially mice as described in detail in the Extended Experimental Procedures.
competed with by Dtau. Consistent with a role for Fyn in Ab
pathology, Fyn transgenic mice present with seizures and Experimental Seizures
Seizures were induced by intraperitoneal (i.p.) injection of (50 mg/kg body
premature mortality (Kojima et al., 1998). This is exacerbated in
weight) pentylenetetrazole (PTZ; Sigma) as described (Roberson et al.,
Fyn/APPmut double-transgenic mice (Chin et al., 2004). More- 2007). Where indicated, mice were injected i.p. with (0.1 mg/kg body weight)
over, APP-associated mortality is reduced on a fyn/ back- MK801 (Sigma) 30 min prior to PTZ administration. Mice were video moni-
ground (Chin et al., 2004). Hence, our findings in APP23.Dtau74 tored, and seizure severity was rated by an independent, blinded person, as
and APP23.tau/ mice are consistent with previous data (Chin follows: 0, no seizures; 1, immobility; 2, tail extension; 3, forelimb clonus; 4,
et al., 2005; Chin et al., 2004). Furthermore, they are in line generalized clonus; 5, bouncing seizures; 6, full extension; and 7, death.
with the recent observation that crossing of PDAPP mice onto
a tau/ background reverses Ab-associated defects (Roberson i.c.v. Treatment with Osmotic Pumps
Six-week-old APP23 and WT mice were anesthetised with ketamine/xylazine,
et al., 2007). and i.c.v. delivery cannulas (Alzet; brain infusion kit #3 with one spacer) were
Mechanistically, our data suggest that stable NR/PSD-95 implanted with a stereotaxic frame (KOPF Instruments) at the following
complex formation is required for Ab toxicity in APP23 mice. coordinates according to the bregma: AP, 0.25 mm; ML, 1 mm; and DV,
This is likely to contribute to disease together with other tau- 2.5 mm. Osmotic mini pumps (Alzet; model #1004) were filled with aCSF
dependent and -independent mechanisms of Ab toxicity. In (Alzet) with and without Tat-NR2B9c or Tat-NR2BAA peptide (750 mM) and
support of our findings, we used a tau/Fyn-independent equilibrated in 0.9% NaCl at 37 C for 48 hr. They were attached to the i.c.v.
cannula tubing and subcutaneously implanted at the back. After 28 days,
approach to disrupt this interaction, by delivering the Tat-
the pumps were replaced with a second batch of pumps via a small skin inci-
NR2B9c peptide to young APP23 mice. This peptide has been sion for another 28 days. Then, they were removed and the tubing was ligated.
shown to protect from excitotoxicity in vitro and in vivo.
We show specifically that perturbing the NR/PSD-95 interac- Statistics
tion with the Tat-NR2B9c peptide improves survival and Statistics was done with the Prizm 4 software (GraphPad) with Student’s t or
memory functions of APP23 mice. The data suggest that disrup- two-way ANOVA test. Values are given as mean ± standard error.
tion of the NR/PSD-95 interaction is sufficient to prevent
Ab toxicity involving NR signaling. Remarkably, Tat-NR2B9c- SUPPLEMENTAL INFORMATION
treated APP23 mice survived long term, suggesting that treat-
Supplemental Information includes Extended Experimental Procedures and
ment within a short therapeutic window is sufficient to prevent
four figures and can be found with this article online at doi:10.1016/j.cell.
lethality. 2010.06.036.
In summary, we reveal a dendritic role for the ‘‘axonal’’
protein tau in postsynaptic targeting of Fyn. This involves inter- ACKNOWLEDGMENTS
action of Fyn with the tau projection domain (Lee et al., 1998).
Accordingly, dominant negative effects of Dtau expression or The authors thank Yves-Alain Barde for providing tau/ mice and him as well
tau deficiency result in reduced postsynaptic Fyn, decreased as Hanns Möhler and Nikolas Haass for helpful comments. This research was
supported by grants from the University of Sydney, National Health and
phosphorylation of its substrate NR2b and instability of NR/
Medical Research Council, Australian Research Council, and Deutsche
PSD-95 complexes (in Dtau74 and tau/ mice). Importantly, Forschungsgemeinschaft. J.G. is a Medical Foundation Fellow.
this additional function of tau appears to be pivotal for medi-
ating Ab toxicity, in that premature lethality, memory deficits, Received: December 14, 2009
and seizure susceptibility of APP23 mice were mitigated in Revised: April 6, 2010
APP23.Dtau74 and APP23.tau/ mice. Hence, reduction of Accepted: May 28, 2010
Published online: July 22, 2010
tau levels or targeting of tau-dependent mechanisms, such as
the Fyn-mediated interaction of NRs and PSD-95, are suitable
REFERENCES
strategies in the treatment of AD and related disorders, high-
lighting tau as an attractive drug target, in addition to Ab Aarts, M., Liu, Y., Liu, L., Besshoh, S., Arundine, M., Gurd, J.W., Wang, Y.T.,
(Ashe, 2007). Salter, M.W., and Tymianski, M. (2002). Treatment of ischemic brain damage

Cell 142, 387–397, August 6, 2010 ª2010 Elsevier Inc. 395


by perturbing NMDA receptor- PSD-95 protein interactions. Science 298, Hsiao, K., Chapman, P., Nilsen, S., Eckman, C., Harigaya, Y., Younkin, S.,
846–850. Yang, F., and Cole, G. (1996). Correlative memory deficits, Abeta elevation,
Ashe, K.H. (2007). A tale about tau. N. Engl. J. Med. 357, 933–935. and amyloid plaques in transgenic mice. Science 274, 99–102.
Kelly, P.H., Bondolfi, L., Hunziker, D., Schlecht, H.P., Carver, K., Maguire, E.,
Ballatore, C., Lee, V.M., and Trojanowski, J.Q. (2007). Tau-mediated neurode-
Abramowski, D., Wiederhold, K.H., Sturchler-Pierrat, C., Jucker, M., et al.
generation in Alzheimer’s disease and related disorders. Nat. Rev. Neurosci. 8,
(2003). Progressive age-related impairment of cognitive behavior in APP23
663–672.
transgenic mice. Neurobiol. Aging 24, 365–378.
Busche, M.A., Eichhoff, G., Adelsberger, H., Abramowski, D., Wiederhold, Kim, E., and Sheng, M. (2004). PDZ domain proteins of synapses. Nat. Rev.
K.H., Haass, C., Staufenbiel, M., Konnerth, A., and Garaschuk, O. (2008). Neurosci. 5, 771–781.
Clusters of hyperactive neurons near amyloid plaques in a mouse model of
Kojima, N., Ishibashi, H., Obata, K., and Kandel, E.R. (1998). Higher seizure
Alzheimer’s disease. Science 321, 1686–1689.
susceptibility and enhanced tyrosine phosphorylation of N-methyl-D-aspar-
Butner, K.A., and Kirschner, M.W. (1991). Tau protein binds to microtubules tate receptor subunit 2B in fyn transgenic mice. Learn. Mem. 5, 429–445.
through a flexible array of distributed weak sites. J. Cell Biol. 115, 717–730.
Kornau, H.C., Schenker, L.T., Kennedy, M.B., and Seeburg, P.H. (1995).
Calhoun, M.E., Wiederhold, K.H., Abramowski, D., Phinney, A.L., Probst, A., Domain interaction between NMDA receptor subunits and the postsynaptic
Sturchler-Pierrat, C., Staufenbiel, M., Sommer, B., and Jucker, M. (1998). density protein PSD-95. Science 269, 1737–1740.
Neuron loss in APP transgenic mice. Nature 395, 755–756. Ksiezak-Reding, H., and Yen, S.H. (1991). Structural stability of paired helical
Chin, J., Palop, J.J., Yu, G.Q., Kojima, N., Masliah, E., and Mucke, L. (2004). filaments requires microtubule-binding domains of tau: a model for self-asso-
Fyn kinase modulates synaptotoxicity, but not aberrant sprouting, in human ciation. Neuron 6, 717–728.
amyloid precursor protein transgenic mice. J. Neurosci. 24, 4692–4697. Kuo, Y.M., Beach, T.G., Sue, L.I., Scott, S., Layne, K.J., Kokjohn, T.A.,
Chin, J., Palop, J.J., Puoliväli, J., Massaro, C., Bien-Ly, N., Gerstein, H., Kalback, W.M., Luehrs, D.C., Vishnivetskaya, T.A., Abramowski, D., et al.
Scearce-Levie, K., Masliah, E., and Mucke, L. (2005). Fyn kinase induces (2001). The evolution of A beta peptide burden in the APP23 transgenic
synaptic and cognitive impairments in a transgenic mouse model of mice: implications for A beta deposition in Alzheimer disease. Mol. Med. 7,
Alzheimer’s disease. J. Neurosci. 25, 9694–9703. 609–618.
Lalonde, R., Dumont, M., Staufenbiel, M., and Strazielle, C. (2005). Neurobe-
Chishti, M.A., Yang, D.S., Janus, C., Phinney, A.L., Horne, P., Pearson, J.,
havioral characterization of APP23 transgenic mice with the SHIRPA primary
Strome, R., Zuker, N., Loukides, J., French, J., et al. (2001). Early-onset
screen. Behav. Brain Res. 157, 91–98.
amyloid deposition and cognitive deficits in transgenic mice expressing
a double mutant form of amyloid precursor protein 695. J. Biol. Chem. 276, Lee, G., Cowan, N., and Kirschner, M. (1988). The primary structure and
21562–21570. heterogeneity of tau protein from mouse brain. Science 239, 285–288.

Crowther, T., Goedert, M., and Wischik, C.M. (1989). The repeat region of Lee, G., Newman, S.T., Gard, D.L., Band, H., and Panchamoorthy, G. (1998).
microtubule-associated protein tau forms part of the core of the paired helical Tau interacts with src-family non-receptor tyrosine kinases. J. Cell Sci. 111,
filament of Alzheimer’s disease. Ann. Med. 21, 127–132. 3167–3177.
Leissring, M.A., Farris, W., Chang, A.Y., Walsh, D.M., Wu, X., Sun, X., Frosch,
Dixit, R., Ross, J.L., Goldman, Y.E., and Holzbaur, E.L. (2008). Differential
M.P., and Selkoe, D.J. (2003). Enhanced proteolysis of beta-amyloid in APP
regulation of dynein and kinesin motor proteins by tau. Science 319,
transgenic mice prevents plaque formation, secondary pathology, and
1086–1089.
premature death. Neuron 40, 1087–1093.
Drechsel, D.N., Hyman, A.A., Cobb, M.H., and Kirschner, M.W. (1992). Modu-
Lewis, J., Dickson, D.W., Lin, W.L., Chisholm, L., Corral, A., Jones, G., Yen,
lation of the dynamic instability of tubulin assembly by the microtubule-
S.H., Sahara, N., Skipper, L., Yager, D., et al. (2001). Enhanced neurofibrillary
associated protein tau. Mol. Biol. Cell 3, 1141–1154.
degeneration in transgenic mice expressing mutant tau and APP. Science 293,
El Khoury, J., Toft, M., Hickman, S.E., Means, T.K., Terada, K., Geula, C., and 1487–1491.
Luster, A.D. (2007). Ccr2 deficiency impairs microglial accumulation and Maas, T., Eidenmüller, J., and Brandt, R. (2000). Interaction of tau with the
accelerates progression of Alzheimer-like disease. Nat. Med. 13, 432–438. neural membrane cortex is regulated by phosphorylation at sites that are
Geschwind, D.H. (2003). Tau phosphorylation, tangles, and neurodegenera- modified in paired helical filaments. J. Biol. Chem. 275, 15733–15740.
tion: the chicken or the egg? Neuron 40, 457–460. Minkeviciene, R., Rheims, S., Dobszay, M.B., Zilberter, M., Hartikainen, J.,
Goedert, M., Wischik, C.M., Crowther, R.A., Walker, J.E., and Klug, A. (1988). Fülöp, L., Penke, B., Zilberter, Y., Harkany, T., Pitkänen, A., and Tanila, H.
Cloning and sequencing of the cDNA encoding a core protein of the paired (2009). Amyloid beta-induced neuronal hyperexcitability triggers progressive
helical filament of Alzheimer disease: identification as the microtubule-associ- epilepsy. J. Neurosci. 29, 3453–3462.
ated protein tau. Proc. Natl. Acad. Sci. USA 85, 4051–4055. Mucke, L., Masliah, E., Yu, G.Q., Mallory, M., Rockenstein, E.M., Tatsuno, G.,
Hu, K., Kholodenko, D., Johnson-Wood, K., and McConlogue, L. (2000). High-
Götz, J., and Ittner, L.M. (2008). Animal models of Alzheimer’s disease and
level neuronal expression of abeta 1-42 in wild-type human amyloid protein
frontotemporal dementia. Nat. Rev. Neurosci. 9, 532–544.
precursor transgenic mice: synaptotoxicity without plaque formation.
Götz, J., Chen, F., Barmettler, R., and Nitsch, R.M. (2001a). Tau filament J. Neurosci. 20, 4050–4058.
formation in transgenic mice expressing P301L tau. J. Biol. Chem. 276,
Nakazawa, T., Komai, S., Tezuka, T., Hisatsune, C., Umemori, H., Semba, K.,
529–534.
Mishina, M., Manabe, T., and Yamamoto, T. (2001). Characterization of Fyn-
Götz, J., Chen, F., van Dorpe, J., and Nitsch, R.M. (2001b). Formation of neuro- mediated tyrosine phosphorylation sites on GluR epsilon 2 (NR2B) subunit of
fibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. the N-methyl-D-aspartate receptor. J. Biol. Chem. 276, 693–699.
Science 293, 1491–1495. Palop, J.J., and Mucke, L. (2009). Epilepsy and cognitive impairments in
Grant, S.G., O’Dell, T.J., Karl, K.A., Stein, P.L., Soriano, P., and Kandel, E.R. Alzheimer disease. Arch. Neurol. 66, 435–440.
(1992). Impaired long-term potentiation, spatial learning, and hippocampal Palop, J.J., Chin, J., Roberson, E.D., Wang, J., Thwin, M.T., Bien-Ly, N., Yoo,
development in fyn mutant mice. Science 258, 1903–1910. J., Ho, K.O., Yu, G.Q., Kreitzer, A., et al. (2007). Aberrant excitatory neuronal
Haass, C., Schlossmacher, M.G., Hung, A.Y., Vigo-Pelfrey, C., Mellon, A., activity and compensatory remodeling of inhibitory hippocampal circuits in
Ostaszewski, B.L., Lieberburg, I., Koo, E.H., Schenk, D., Teplow, D.B., et al. mouse models of Alzheimer’s disease. Neuron 55, 697–711.
(1992). Amyloid beta-peptide is produced by cultured cells during normal Phillips, G.R., Huang, J.K., Wang, Y., Tanaka, H., Shapiro, L., Zhang, W., Shan,
metabolism. Nature 359, 322–325. W.S., Arndt, K., Frank, M., Gordon, R.E., et al. (2001). The presynaptic particle

396 Cell 142, 387–397, August 6, 2010 ª2010 Elsevier Inc.


web: ultrastructure, composition, dissolution, and reconstitution. Neuron 32, Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair
63–77. synaptic plasticity and memory. Nat. Med. 14, 837–842.
Roberson, E.D., Scearce-Levie, K., Palop, J.J., Yan, F., Cheng, I.H., Wu, T., Sturchler-Pierrat, C., Abramowski, D., Duke, M., Wiederhold, K.H., Mistl, C.,
Gerstein, H., Yu, G.Q., and Mucke, L. (2007). Reducing endogenous tau Rothacher, S., Ledermann, B., Bürki, K., Frey, P., Paganetti, P.A., et al.
ameliorates amyloid beta-induced deficits in an Alzheimer’s disease mouse (1997). Two amyloid precursor protein transgenic mouse models with Alz-
model. Science 316, 750–754. heimer disease-like pathology. Proc. Natl. Acad. Sci. USA 94, 13287–13292.
Roche, K.W., Standley, S., McCallum, J., Dune Ly, C., Ehlers, M.D., and Went- Tezuka, T., Umemori, H., Akiyama, T., Nakanishi, S., and Yamamoto, T. (1999).
hold, R.J. (2001). Molecular determinants of NMDA receptor internalization. PSD-95 promotes Fyn-mediated tyrosine phosphorylation of the N-methyl-D-
Nat. Neurosci. 4, 794–802. aspartate receptor subunit NR2A. Proc. Natl. Acad. Sci. USA 96, 435–440.
Rong, Y., Lu, X., Bernard, A., Khrestchatisky, M., and Baudry, M. (2001). Tyro- Tucker, K.L., Meyer, M., and Barde, Y.A. (2001). Neurotrophins are required for
sine phosphorylation of ionotropic glutamate receptors by Fyn or Src differen- nerve growth during development. Nat. Neurosci. 4, 29–37.
tially modulates their susceptibility to calpain and enhances their binding to Van Dam, D., D’Hooge, R., Staufenbiel, M., Van Ginneken, C., Van Meir, F., and
spectrin and PSD-95. J. Neurochem. 79, 382–390. De Deyn, P.P. (2003). Age-dependent cognitive decline in the APP23 model
Röskam, S., Neff, F., Schwarting, R., Bacher, M., and Dodel, R. (2010). APP precedes amyloid deposition. Eur. J. Neurosci. 17, 388–396.
transgenic mice: the effect of active and passive immunotherapy in cognitive Weingarten, M.D., Lockwood, A.H., Hwo, S.Y., and Kirschner, M.W. (1975).
tasks. Neurosci. Biobehav. Rev. 34, 487–499. A protein factor essential for microtubule assembly. Proc. Natl. Acad. Sci.
Salter, M.W., and Kalia, L.V. (2004). Src kinases: a hub for NMDA receptor USA 72, 1858–1862.
regulation. Nat. Rev. Neurosci. 5, 317–328.
Yagi, T., Aizawa, S., Tokunaga, T., Shigetani, Y., Takeda, N., and Ikawa, Y.
Selkoe, D.J. (1997). Alzheimer’s disease: genotypes, phenotypes, and treat- (1993). A role for Fyn tyrosine kinase in the suckling behaviour of neonatal
ments. Science 275, 630–631. mice. Nature 366, 742–745.
Selkoe, D.J. (2002). Alzheimer’s disease is a synaptic failure. Science 298, Zhao, L., Ma, Q.L., Calon, F., Harris-White, M.E., Yang, F., Lim, G.P., Morihara,
789–791. T., Ubeda, O.J., Ambegaokar, S., Hansen, J.E., et al. (2006). Role of p21-acti-
Shankar, G.M., Li, S., Mehta, T.H., Garcia-Munoz, A., Shepardson, N.E., vated kinase pathway defects in the cognitive deficits of Alzheimer disease.
Smith, I., Brett, F.M., Farrell, M.A., Rowan, M.J., Lemere, C.A., et al. (2008). Nat. Neurosci. 9, 234–242.

Cell 142, 387–397, August 6, 2010 ª2010 Elsevier Inc. 397


Single-Stranded DNA Transposition
Is Coupled to Host Replication
Bao Ton-Hoang,1,* Cécile Pasternak,2 Patricia Siguier,1 Catherine Guynet,1 Alison Burgess Hickman,3 Fred Dyda,3
Suzanne Sommer,2 and Michael Chandler1,*
1Laboratoire de Microbiologie et Génétique Moléculaires, Centre National de Recherche Scientifique, Unité Mixte de Recherche 5100,

118 Route de Narbonne, F31062 Toulouse Cedex, France


2Université Paris-Sud, Centre National de Recherche Scientifique, Unité Mixte de Recherche 8621, Laboratoire de Recherche Correspondant

du Commissariat à l’Energie Atomique 42V, Institut de Génétique et Microbiologie, Bâtiment 409, Orsay, France
3Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda,

MD 20892, USA
*Correspondence: bao.tonhoang@ibcg.biotoul.fr (B.T-H.), michael.chandler@ibcg.biotoul.fr (M.C.)
DOI 10.1016/j.cell.2010.06.034

SUMMARY genetic tools in identifying specific gene regulatory regions by


insertion and are being developed as delivery systems for gene
DNA transposition has contributed significantly to therapy applications.
evolution of eukaryotes and prokaryotes. Insertion A variety of structurally and mechanistically distinct enzymes
sequences (ISs) are the simplest prokaryotic trans- (transposases) have evolved to carry out transposition by several
posons and are divided into families on the basis different pathways (Turlan and Chandler, 2000; Curcio and
of their organization and transposition mechanism. Derbyshire, 2003). They all possess an endonuclease activity
allowing them to cleave, excise, and insert transposon DNA
Here, we describe a link between transposition of
into a new location. Depending on the system (Curcio and Derby-
IS608 and ISDra2, both members of the IS200/ shire, 2003), different types of nucleophile can be used by trans-
IS605 family, which uses obligatory single-stranded posases to attack a phosphorus atom of a backbone phospho-
DNA intermediates, and the host replication fork. diester bond and cleave DNA. These include water (generally
Replication direction through the IS plays a crucial activated by enzyme-bound metal ions), a hydroxyl group at
role in excision: activity is maximal when the ‘‘top’’ the 50 or 30 end of a DNA strand, or a hydroxyl group of an amino
IS strand is located on the lagging-strand template. acid of the transposase itself, such as serine or tyrosine.
Excision is stimulated upon transient inactivation of Many mobile DNA elements move using a ‘‘cut-and-paste’’
replicative helicase function or inhibition of Okazaki mechanism by excision of a double-stranded copy from one
fragment synthesis. IS608 insertions also exhibit an genomic location and insertion at another. Recently, a family of
orientation preference for the lagging-strand tem- bacterial insertion sequences (ISs), the IS200/IS605 family, has
been found that uses a completely different pathway and an
plate and insertion can be specifically directed to
unusual transposase with a catalytic tyrosine (a Y1 transposase).
stalled replication forks. An in silico genomic Studies of one member, IS608 (Figure 1A), provided a detailed
approach provides evidence that dissemination of picture of their transposition (Ton-Hoang et al., 2005; Ronning
other IS200/IS605 family members is also linked to et al., 2005; Guynet et al., 2008; Barabas et al., 2008). In vitro,
host replication. this requires single-stranded DNA (ssDNA) substrates and is
strand specific: only the ‘‘top’’ strand is recognized by the
INTRODUCTION element-encoded transposase, TnpA, and is cleaved and trans-
ferred, whereas the ‘‘bottom’’ strand does not transpose. Exci-
DNA transposition involves movement of discrete DNA seg- sion of the top strand as a transposon circle with joined left
ments (transposons) from one genomic location to another. and right ends is accompanied by rejoining of the DNA flanks.
It occurs in all kingdoms of life and has contributed significantly The circle junction then undergoes TnpA-catalyzed integration
to evolution of eukaryotes and prokaryotes. Transposable into an ssDNA target in a sequence-specific reaction. Insertion
elements can represent a significant proportion of their host involves transfer of both the 50 and 30 ends of the single-strand
genomes (Biémont and Vieira, 2006). They have been particularly circle junction into the ssDNA target. The left (50 ) IS608
well studied in bacteria where they are major motors of broad end always inserts specifically just 30 of the tetranucleotide,
genome remodeling, play an important role in horizontal gene 50 -TTAC-30 (Kersulyte et al., 2002), which is also essential for
transfer, and can sequester and transmit a variety of genes subsequent transposition (Ton-Hoang et al., 2005).
involved in accessory cell functions, such as resistance to anti- The obligatorily single-stranded nature of IS200/IS605 trans-
microbial agents, catabolism of unusual compounds, and path- position in vitro raises the possibility that it is limited in vivo by
ogenicity, virulence, or symbiosis. They are also important as the availability of its ssDNA substrates. A number of cellular

398 Cell 142, 398–408, August 6, 2010 ª2010 Elsevier Inc.


processes generate or occur using ssDNA, including DNA repair,
natural transformation, conjugative plasmid transfer, single-
stranded phage infection, and replication (where the DNA
serving as the template for Okazaki fragment synthesis on the
lagging strand of the replication fork is single stranded).
Here, we investigate the link between IS608 transposition and
the availability of ssDNA during replication in vivo. Our results
demonstrate that transposition of the IS200/IS605 family is
closely integrated into the host cell cycle and takes advantage
of the presence of ssDNA on the lagging-strand template at
the replication fork for dissemination. We also show that IS608
transposition is affected by perturbing the fork: transitory inacti-
vation of crucial replication proteins increased excision from the
lagging-strand template, and stalling of the fork resulted in inser-
tions directed to the lagging strand of the blocked fork.
Our results also suggest that insertion and excision of the
related element, ISDra2, also depends on the lagging-strand
template in its host, the radiation-resistant bacterium Deinococ-
cus radiodurans, and that this dependency can be abolished
with irradiation. We have extended our analysis to a number of
related IS200/IS605 elements in a variety of sequenced bacterial
genomes. The results of this in silico analysis are also consistent
with a strong bias of insertion into the lagging-strand template in
these organisms. Together, the results demonstrate the impor-
tance of the lagging-strand template for IS608 and ISDra2
activity and suggest that all IS200/IS605 family members have
evolved a mode of transposition that exploits ssDNA at the repli-
cation fork.

RESULTS

IS608 Excision Depends on the Direction of Replication


To investigate whether replication direction affects IS608 trans-
position, we used a plasmid assay in E. coli to monitor the exci-
sion step of transposition (Ton-Hoang et al., 2005). In this assay,
the IS-carrying plasmids included an IS608 derivative in which
the tnpA and tnpB genes (Figure 1A) were replaced by a chloram-
phenicol resistance (CmR) cassette (Figure 1C). In one case, the
active (top) IS608 strand was located in the lagging-strand
template (pBS102), and in the second, the replication origin
Figure 1. IS608 Excision Depends on the Direction of Replication was inverted (Figures 1B and 1C), placing the transposionally
(A) IS608 organization and a simplified transposition model. Gray arrows, tnpA active top strand on the leading-strand template (pBS144).
and tnpB orfs; red and blue boxes, left (LE) and right (RE) ends (color code A second compatible plasmid supplied TnpA in trans under
retained throughout).
(Ai) Schematized single-stranded IS608 showing secondary structures of LE
and RE, the flanking TTAC, and cleavage positions at the ends (vertical black strand as part of the lagging-strand template. Right: pBS144 with the active
arrows). IS608 strand as part of the leading-strand template. Ori, pBR322 origin of repli-
(Aii) Excision and ssDNA circle formation with an RE-LE junction and a sealed cation; Cm, bla, SpSm, chloramphenicol, b-lactamase and, streptomycin/
donor joint (black line) retaining TTAC. spectinomycin resistance genes; Plac, lac promoter; tnpA-his, C-terminal
(Aiii) TnpA brings together the transposon junction with a new TTAC-carrying his6-tagged tnpA gene. Directions of DNA replication and transcription are
target (dotted black line). Vertical black arrows, points of cleavage and strand indicated. Agarose gel (0.8%) showing separation of plasmid DNA from over-
transfer. night cultures of strains carrying pBS102 + pBS21 (lane 2) and pBS144 +
(Aiv) IS608 insertion into the target. pBS121 (lane 3). Lane 1, 1 kb standard.
(B) Orientation of the IS608 derivative with respect to replication direction. The (D) Mating-out assays. Left-hand column, transposon donor plasmid; middle,
disposition of the IS608 active (top) strand with respect to replication direction presence or absence of TnpA (relevant plasmids in parentheses); right,
is shown when the fork approaches from one direction (left) when it is part of measured transposition frequencies (standard error in parentheses; n > 3).
the lagging-strand template or the other (right) when it is part of the leading Plasmids pBS102ter1 and pBS144ter1 carry a single set of origin proximal
strand. This is described in more detail in Figure S1 and its legend. terminators; pBS102ter2 and pBS144ter2 carry two sets of terminators flank-
(C) Excision measured directly in vivo by the appearance of ‘‘donor joint’’ plas- ing both transposon ends (Extended Experimental Procedures).
mids, deleted for the IS608 derivative. Left: pBS102 with the active IS608 See also Figure S1A.

Cell 142, 398–408, August 6, 2010 ª2010 Elsevier Inc. 399


control of plac (Figure 1C). After overnight growth, IS excision when the growth protocol was modified to include a 30 min
was monitored by detection of reclosed donor backbone mole- temperature shift to 42 C (and further incubation of 3 hr at
cules from which the IS had been deleted. 30 C to allow replication to recover), excision increased about
As shown in Figure 1C, when the IS608 active strand was 10- and 7-fold in the dnaB and dnaG mutants, respectively,
located on the lagging-strand template, the donor backbone compared to the wild-type (column d). Omission of the 42 C
species could be clearly identified along with the parental step resulted in indistinguishable basal levels for wild-type and
plasmid and the plasmid used to supply transposase (lane 2). mutant hosts (column c). Thus, inactivation of DnaB helicase
However, when the replication origin was inverted and the active function or inhibition of initiation of Okazaki fragment synthesis
IS strand was located on the leading strand, formation of the with a dnaGts mutation stimulated the excision step of transpo-
excised donor backbone species was only barely detectable sition, consistent with the notion that ssDNA at the replication
(lane 3). fork is a substrate for IS excision.
This effect of replication direction on IS608 transposition was
also observed in mating-out assays (Galas and Chandler, 1982) Effect of Transposon Size on Excision
that measure overall transposition frequency. We monitored If excision occurs at the replication fork and requires ssDNA, it
transposition was monitored by following movement of IS608 seemed possible that IS length might influence excision
from a nonmobilizable donor plasmid into a conjugative plas- frequency since the probability that both IS ends are within the
mid. When the IS608 active strand was on the lagging-strand single-stranded region of the lagging-strand template should
template (Figure 1D, line 2), the transposition frequency was decrease with increasing IS length. We therefore examined the
5.6 3 105, but when it was on the leading-strand template, effect of IS length in the excision assay by using a set of IS608
the frequency dropped 27-fold to 2.1 3 106 (line 3). To ensure derivatives with varying spacing between the left end (LE) and
that this was not due to possible changes in transcription right end (RE) and found that excision decreased strongly as
resulting from the inversion introduced during cloning to a function of increasing IS length and that these frequencies
switch the orientation of the replication origin (where bla was were strongly modified in a strain carrying a dnaGts mutation.
inverted together with ori), we inserted transcriptional termina- Increasing the IS length from 0.3 to 2 kb, resulted in a 7- to
tors (Simons et al., 1987) on either one (lines 4–5) or both (lines 10-fold decrease in excision frequency with a log-linear relation-
6–7) sides of the IS608 derivatives to insulate them from ship, consistent with the notion that excision occurs more effi-
impinging transcription. In these cases, the observed effect of ciently when both ends are located within the short single
replication direction on transposition frequency remained lagging-strand region of about 1.5–2 kb upstream of the first
unchanged. complete Okazaki fragment at the replication fork (see Johnson
and O’Donnell, 2005, for review). As the IS length was further
Effect of Mutant Primase and Replicative Helicase increased, excision decreased only slightly, at least up to a trans-
on IS Excision poson length of 4 kb (Figure 2C) with a possible inflection
Since IS608 excision is sensitive to replication direction, we between 1.5 and 2 kb.
asked whether it was affected by perturbation of the replication The length of ssDNA on the lagging-strand template depends
fork. We used two temperature-sensitive replication mutants: on the initiation frequency of Okazaki fragment synthesis, in turn
dnaG308ts, encoding a mutant DNA primase, DnaG (Wechsler determined by DnaG. Progressive inactivation of DnaG activity
and Gross, 1971), and dnaB8ts, encoding a mutant of the essen- by growth of the dnaGts mutant at increasing but sublethal
tial replication fork DNA helicase, DnaB (Carl, 1970). Replication temperatures should reduce this frequency and increase the
in these mutants occurs at 30 C but is interrupted after a shift to mean length of ssDNA at the replication fork upstream of the first
the restrictive temperature of 42 C. Inhibition of either DnaG or complete Okazaki fragment. We therefore analyzed excision of
DnaB activity is expected to increase the amount of ssDNA at the IS608-derived transposons in wild-type and dnaGts mutants
the fork, principally on the lagging-strand template (Louarn, at different temperatures (Figure 2D). While profiles were indis-
1974; Fouser and Bird, 1983; Belle et al., 2007) (see also the tinguishable for the wild-type strain at 30 C and 33 C, the
Discussion). dnaGts mutant exhibited a generally higher excision frequency
To test the effect of inhibition of DnaG and DnaB activities, we at 30 C and showed a lower length dependent slope, revealing
used a genetic screen in which a b-lactamase gene is interrupted that the replication fork is affected by the mutation even at the
by an IS608 derivative (pAM1, Figure S2; Experimental Proce- normal permissive temperature. However, an inflection still
dures). TnpA-catalyzed precise excision (using TnpA provided appeared to occur. At 33 C, excision increased significantly,
in trans) results in reconstitution of the b-lactamase gene particularly for the longer transposons.
(Figure 2A) and the appearance of ampicillin resistant (ApR) To further examine this, we cloned the wild-type dnaG allele
colonies. downstream of the tnpAIS608 gene in the TnpA-providing plasmid
As shown in Figures 2Bi, 2Bii, and 2Biii for the wild-type and so that both were under control of the same promoter (Experi-
dnaB and dnaG mutants, respectively, after overnight growth mental Procedures). When this plasmid, pBS179, was intro-
at 30 C (a permissive temperature) without TnpA induction, the duced into the dnaGts strain, it clearly suppressed the dnaGts
frequency of ApR colonies was low for the wild-type and mutant defect at 33 C (Figure 2E). Moreover, when introduced into the
hosts (column a). Induction of TnpA expression resulted in wild-type dnaG strain, the excision frequencies were even
a nearly 3-fold increase in excision with little difference between further reduced and the length-dependant slope was increased
wild-type, dnaBts, and dnaGts strains (column b). However, (Figure 2F).

400 Cell 142, 398–408, August 6, 2010 ª2010 Elsevier Inc.


Figure 2. Excision Frequency: Effects of dnaBts and dnaGts Mutants and IS Length
(A) Schematic of the excision assay. The relevant features of the generic IS-carrying donor plasmid, pAM1, and the product after IS excision, DpAM1, are shown
together with the plasmid used to supply transposase, pBS135. These are described in detail in Figure S1 and its legend.
(B) Transitory inactivation of DnaB or DnaG. Stimulation factors for excision from wild-type, dnaBts, and dnaGts strains: i, ii, and iii, respectively. Values are
normalized to those of an overnight culture without IPTG at 30 C for each strain (0.8 3 102, 0.6 3 102, and 2.2 3 102 corresponding to column a), overnight
cultures diluted at 30 C with IPTG for 4 hr (column b), TnpA induction at 30 C (45 min) and incubation at 30 C (3 hr 30 min) without IPTG (column c), and TnpA
induction at 30 C (45 min) followed by a shift to 42 C without IPTG (30 min) and then at 30 C (3 hr) (column d).
(C) Effect of IS length on excision frequency. IS608 derivatives are 0.3, 0.5, 0.8, 1.1, 1.4, 1.9, 3, and 4 kb long. The curve shows results expressed as ApRTpRKmR /
TcRKmR with DH5a at 37 C. Error bars are the standard deviation (SD) of more than four independent experiments.
(D) Effect of dnaG on IS length-dependant excision. MC4100, 30 C (dark blue) and 33 C (yellow); MC4100dnaGts, 30 C (light blue); and MC4100dnaGts+dnaGwt,
33 C (purple).
(E) Effect of DnaGwt overexpression on excision frequency in dnaGts strains. MC4100dnaGts, 33 C (purple), and MC4100dnaGts+dnaGwt, 33 C (light blue).
(F) Effect of DnaGwt overexpression on excision frequency in wild-type strains. MC4100, 33 C (yellow), and MC4100+dnaGwt, 33 C (purple).
See also Figure S1B.

IS608 Insertion into the E. coli Chromosome lagging-strand template, they should occur in one orientation
The circular E. coli chromosome replicates bidirectionally from on one side of ori and in the opposite orientation on the other.
the replication origin, oriC. If IS608 insertions target the To test this, we isolated IS608 insertions in the E. coli

Cell 142, 398–408, August 6, 2010 ª2010 Elsevier Inc. 401


strand, the lagging-strand template, a result that has strong
statistical support (Figure 3 legend).
In all cases, both plasmid and chromosome insertions
occurred 30 to a TTAC target, as expected. As these are distrib-
uted equally on both strands of the chromosome and the TnpA
donor plasmid (data not shown), the observed strand biases
for insertion cannot be explained by a bias in target sequence
distribution.

Targeting Stalled Plasmid Replication Forks:


Tus-Ter System
Since our accumulating data suggested that IS608 targets
ssDNA at the replication fork, we asked whether insertion could
also be observed into forks stalled at a predefined location. For
this, we used the E. coli Tus/Ter system in which a Ter site, when
bound by the protein Tus, strongly reduces replication fork
progression through Ter in the nonpermissive orientation (Ternp)
(see Bierne et al., 1994) but not in the opposite, permissive,
orientation (Terp) (Neylon et al., 2005).
The assay (Figure 4A and Figure S1) used a suicide conjuga-
tion system (Demarre et al., 2005) in which the unidirectionally
replicating target plasmid was carried by a recipient strain with
an inactivated chromosomal tus gene. The target plasmid
Figure 3. Insertions into the E. coli Chromosome and into a Resident carried a functional tus gene and a Ter site in either the permis-
Plasmid sive or nonpermissive orientation (Figures 4B and Extended
(A) Experimental system. The temperature-sensitive transposon donor
Experimental Procedures). The plasmid-based tus gene was
plasmid, pBS156 (middle), and the plasmid used for supplying TnpA,
pBS135 (left), are shown together with a schematic of the host chromosome
transitorily induced only for the duration of the experiment to
(right). Growth at the nonpermissive temperature results in loss of pBS156 avoid plasmid loss. We also inserted a stretch of DNA with
and IS insertion into the chromosome or pBS135. This is described in detail several spaced copies of the TTAC target tetranucleotide on
in Figure S1 and its legend. both strands upstream of the Ter sites.
(B) Chromosomal insertions. The chromosome is shown linearized at its Mapping of the IS insertion sites revealed that all occurred at
terminus. Red spot, origin of replication, ori; rrn, ribosomal RNA genes; black
TTAC sequences and were distributed over the entire length of
arrows, positions of insertions (Table S1). Multiple insertions are indicated by
both target plasmids, largely in an orientation expected for inser-
a number above the larger arrows. Replication occurs in both directions into
the left and right replicores, blue and orange, respectively: the lagging-strand tion into the lagging-strand template (Figure 4B, black arrow-
template of the left (bottom) and right (top) replicores. Fisher’s exact test gives heads). The overall distribution was the same regardless of Ter
a p value of 7.122e6. site orientation. As for the other target plasmids used here, we
(C) Plasmid insertions. Replication of the p15A-based plasmid occurs from left confirmed that TTAC sequences were distributed roughly
to right. Orange triangle, origin of replication; KmR, tnpA, and lacIQ, kanamycin equally on both DNA strands (data not shown).
resistance, transposase and lac repressor genes, respectively. Arrows indi-
An additional set of insertions was observed in the target
cate their direction of transcription. Vertical arrowheads show the position of
the insertions obtained from the same experiments as in (A) above. plasmid carrying Ternp when compared to that with Terp (Figures
See also Figure S1C and Table S1. 4B and 4C). To precisely map these, we used PCR analysis with
primers complementary to a sequence upstream of the Ter sites
and either LE (for lagging-strand insertions) or RE (for leading-
chromosome using a temperature-sensitive plasmid as the IS strand insertions). When the target plasmid carried Ternp, ampli-
donor and supplying TnpA in trans (Figure 3A). Insertions were fication products from four independent experiments (Figure 4C,
localized by an arbitrary PCR procedure (Experimental Proce- lanes 1–4) revealed insertions adjacent to the Ter site on the
dures) followed by DNA sequencing. We observed a dramatic lagging-strand template (Figure 4B, filled red arrowheads; Fig-
skew in strand specificity of IS608 insertion relative to the origin ure 4C), although occasionally an insertion was observed on
of replication, oriC. the opposite strand (lane 5). Although there was a degree of vari-
Insertions either in the left or right replicores were in the ability in the insertion distribution between experiments (com-
orientation expected for transposition into the lagging-strand pare lanes 1–4), a consistently strong signal was obtained from
template (Figure 3B). It is interesting to note that while insertions the TTAC site located at 63 bp from Ternp, suggesting that this
were distributed around the chromosome, many appeared in the is the preferred insertion site. However, insertions were also
vicinity of the highly transcribed rRNA genes (Table S1). observed at the closest site, located only 26 bp from Ter
We also obtained insertions into the TnpA donor plasmid in the (Figure 4C, lanes 1, 3, and 4).
same experiment. In contrast to the chromosome, this plasmid With Terp, no strong Tus-dependent insertions were observed
replicates unidirectionally and the IS608 insertion pattern was (Figure 4D). In four independent experiments we only once iso-
completely (Figure 3C) different. All occurred into only one lated an insertion in this region (in the plasmid population from

402 Cell 142, 398–408, August 6, 2010 ª2010 Elsevier Inc.


experiment 1, lane 1), and this had occurred into a TTAC site
within the Ter sequence itself rather than immediately upstream.
Thus, the lagging strand of replication forks blocked by Tus is
a preferential target for IS608 insertions and these can occur in
close proximity to the Tus binding site, Ter.

Another IS200/IS605 Family Member: ISDra2


ISDra2 is an IS200/IS605 family member from the highly radiation
resistant D. radiodurans. It has a similar organization to IS608
and inserts specifically 30 to a TTGAT pentanucleotide (Islam
et al., 2003). Like IS608, it transposes using ssDNA intermedi-
ates (Pasternak et al., 2010). We had observed specific stimula-
tion of ISDra2 transposition upon g- or UV-irradiation (Mennecier
et al., 2006; Pasternak et al., 2010) related to the availability of
ssDNA generated during radiation-induced fragmentation and
reassembly of the host genome, called extended synthesis
dependent strand annealing (ESDSA) (Zahradka et al., 2006).
To determine whether spontaneous ISDra2 transposition is
influenced by host chromosome replication, we measured
ISDra2 excision in D. radiodurans as a function of replication
direction through the IS. D. radiodurans replication is thought
to be bidirectional (Hendrickson and Lawrence, 2006). We con-
structed D. radiodurans strains with an ISDra2 derivative,
ISDra2-113 (Pasternak et al., 2010), placed in one or the other
orientation at the same chromosomal location (Extended Exper-
imental Procedures). Excision of these ISs reconstitutes a func-
tional TcR gene. Consistent with our results with IS608, no exci-
sion (<4.8 3 109; Figure 5A) was detected without transposase,
whereas in the presence of TnpAISDra2, relatively high excision
levels (2.1 3 103) were observed when the IS derivative was
in one orientation but was reduced about 30-fold to 6.7 3 105
in the other (Figure 5A).
However, the orientation bias of excision was lost in g-irradi-
ated D. radiodurans strains: after g-irradiation, excision of
ISDra2-113 reached the same and maximal level of about 2 3
102 for both orientations relative to the direction of replication
(Figure 5A). Since there should be no strand bias in generating
ssDNA after irradiation during ESDSA, the lack of excision and
insertion bias in irradiated D. radiodurans suggests that insertion
Figure 4. IS608 Insertions into Tus/Ter Stalled Replication Forks targets are no longer limited to those rendered accessible during
(A) Experimental design. The suicide conjugative IS-carrying donor plasmid, the replication.
pBS167b, is shown (top). It cannot propagate in the recipient, and IS insertions
Finally, we isolated 23 independent spontaneous insertions of
were monitored into the Ter-carrying target plasmid, pBS172-3, as described
in detail in Figure S1 and its legend. Plasmids are described in the Extended ISDra2-113 in D. radiodurans chromosome I by scoring for TcR
Experimental Procedures. CmR colonies. All occurred into a TTGAT target and exhibited
(B) Insertions with Ter in permissive and nonpermissive orientations. Replica- an orientation bias (18/23) on the lagging-strand template (Fig-
tion from ori is from left to right; gray boxes, araC repressor, tus, and bla ure 5B and Table S2) without irradiation. In contrast, 21 inser-
genes; *, Ter-proximal TTAC sequences; horizontal red arrowhead, Ternp tions isolated from g-irradiated D. radiodurans occurred essen-
(nonpermissive, pBS172); horizontal black arrowhead, Terp (permissive,
tially equally on both strands (Figure 5C and Table S3).
pBS173); black vertical arrowheads, IS608 insertions; red vertical arrowheads,
multiple insertions upstream of Ternp or an insertion inside of Terp. The position
of the oligonucleotides used to localize insertions is also shown. Genomic Analysis Reveals Orientation Bias
(C) Detailed analysis of insertions in the Ternp region. PCR amplification prod- in Other IS200/IS605 Family Members
ucts were separated on a 5% acrylamide gel. The distribution and orientation In view of the strong orientation bias observed for IS608 and
of insertions close to Ter were determined in four independent experiments ISDra2 insertions, we wondered whether similar patterns might
(LE, lanes 1–4; RE, lanes 5–8). Distribution of TTAC on the lagging-strand
be found in other bacterial genomes as vestiges of members
and leading-strand template are shown in the cartoon (left and right, respec-
of the IS200/IS605 family occur widely (ISfinder: http://www-is.
tively).
(D) Detailed analysis of insertions in the Terp region. Identical experiment as biotoul.fr). We identified several genomes with a significant
shown in (C) with Terp. number of copies of various members of this family and deter-
See also Figure S1D. mined the orientation of insertion relative to the origin of

Cell 142, 398–408, August 6, 2010 ª2010 Elsevier Inc. 403


(Pestoides F) (Figure 6G) are clearly different and show a complex
pattern with no apparent relationship to the replication direction.
However, these Y. pestis strains revealed aberrant GC skews
resulting from a series of inversions and rearrangements
throughout the chromosome (Song et al., 2004; Garcia et al.,
2007). When these are taken into account, an astoundingly
close correlation emerges between GC skew and IS1541
orientation.
Similar reasoning may explain those insertions that initially
appear in contradiction with a lagging-strand template targeting
preference. For example, of the five IS200 insertions in S. enter-
ica strain CT18 oriented in the opposite direction (Figure 6A), one
is located in a short chromosome region with an inverted GC-
skew compared to the neighboring sequences. This implies
that this region has undergone inversion and that the associated
IS insertion likely had occurred prior to this event. The insertion
pattern observed in S. enterica (Ty2) (Deng et al., 2003;
Figure 6B) is quite similar to that of S. enterica CT18; both carry
an identical number of IS200 copies. The differences in IS200
Figure 5. Excision and Insertion of ISDra2-113 in Deinococcus radio- distribution between these two strains can be entirely explained
durans by a large interreplicore inversion between S. enterica (Ty2) and
(A) ISDra2-113 excision frequencies depend on whether the active (top) IS- S. enterica CT18. These genomic results strongly support the
Dra2-113 strand is located on the lagging- or leading-strand template. The
idea that insertion into the lagging-strand template of the chro-
middle column indicates whether TnpA was provided and whether the cells
were subjected to g-irradiation. Strain constructions are presented in the mosomes of their host is a general characteristic of IS200/
Extended Experimental Procedures. IS608 family members.
(B) Spontaneous ISDra2-113 insertions into the D. radiodurans chromosome I.
The origin (ori, red ellipse) and direction of replication of the D. radiodurans DISCUSSION
chromosome was assumed to be that proposed by Hendrickson and Law-
rence (2006). All insertions occurred 30 to TTGAT pentanucleotide sequences.
The transposition pathway of IS200/IS605 IS family members
Detailed mapping is presented in Table S2. Fisher’s exact test gave a p value of
0.01108. Multiple insertions are indicated by a number above the larger
involves ssDNA substrates and intermediates. In vitro IS excision
arrows. requires both transposon ends to be single stranded, and inser-
(C) ISDra2-113 insertions into the D. radiodurans chromosome I after g-irradi- tion of the excised single-stranded circular transposon interme-
ation. Detailed mapping is presented in Table S3. Fisher’s exact test gave diate requires access to an ssDNA target (Guynet et al., 2009).
a p value of 0.1344, suggesting that the pattern is random. We now demonstrate that excision and insertion of two family
See also Tables S2 and S3.
members, IS608 and ISDra2, occur preferentially in the lagging-
strand DNA template in vivo.
replication (as defined by GC skew). The genomic ISs exhibited Excision is dependent on replication direction through the IS: it
a strong orientation bias, supporting our notion that the lagging- is high when the active IS strand is on the lagging-strand
strand template provides the most attractive ssDNA for IS template but substantially lower when part of the leading-strand
targeting. template. We also examined insertions of a plasmid-localized
The results for representative bacterial genomes are shown in IS608 into normally replicating E. coli chromosomes and sponta-
Figure 6. For example, S. enterica CT18 carries 27 full copies of neous insertions of ISDra2 into D. radiodurans chromosome I.
IS200 (Deng et al., 2003) (Figure 6A). Thirteen of these are on They were largely directed to the lagging-strand template, result-
one replicore and 14 on the other, while all but five occur in ing in a skew of strand-specific insertion on either side of the
the orientation expected for insertion into the lagging-strand replication origin in these bidirectionally replicating chromo-
template. Another example is Yersinia pseudotuberculosis somes. For a unidirectionally replicating E. coli plasmid, inser-
(IP31758) (Chain et al., 2004) (Figure 6C) with 16 full IS1541 tions occurred into only one strand. Importantly, the orientation
copies (nine in the left and seven in the right replicore), 13 of effect for ISDra2 insertion was abolished when transposition
these in an orientation consistent with insertion into the was triggered by g-irradiation, accompanied by an increase in
lagging-strand template. Photobacterium profundum SS9 (Vezzi transposition frequency, consistent with the observation that
et al., 2005; Figure 6D) carries 28 ISPpr13 copies distributed g-irradiation induces a repair pathway resulting in massive
between its two chromosomes, with 26 in the expected orienta- amounts of ssDNA with no obvious strand bias.
tion, and in Shewanella woodyi with 13 copies of ISShwo2, all IS608 insertions into the E. coli chromosome were fairly evenly
except one are in the expected orientation (Karpinets et al., distributed but a significant number occurred in the highly tran-
2010; Figure 6E). Fisher’s exact test provided strong statistical scribed rrn genes (which are oriented in the sense of replication),
support (Figure 6 legend). suggesting that high transcription levels might also provide
Interestingly, the distribution of the 42 IS1541 copies in accessible ssDNA for IS608 insertion, e.g., by generating R loops
Y. pestis (Microtus) (Figure 6F) and the 50 copies of it in Y. pestis or by affecting replication fork passage. Replication in E. coli

404 Cell 142, 398–408, August 6, 2010 ª2010 Elsevier Inc.


Figure 6. Orientation of IS200/IS605 Family Members in Bacterial Genomes
Overall GC skew (G – C / G + C) is indicated in blue and orange. Black line, calculated GC skew with Artemis (http://www.sanger.ac.uk/Software/Artemis/) with
a 10 kb window; red ellipse, origin of replication; vertical arrow heads (above or below the genome depending on their orientation), individual ISs.
(A) S. enterica (typhi) CT18 (NC_003198); IS200, 709 bp (Fisher’s exact test p value, 0.002054).
(B) S. enterica (typhi) Ty2 (NC_004631); IS200, 709 bp (p value, 0.004799).
(C) Y. pseudotuberculosis IP31758 (NC_009708); IS1541, 708–709 bp (p value, 0.02144).
(D) P. profundum SS9 (NC_006371 and NC_006370); ISPr13, 596 bp (chromosome 1 p value, 0.0005139; chromosome 2 p value, 0.002806).
(E) S. woodyi (NC_010506); ISShwo2, 613 bp (p value, 0.02297).
(F) Y. pestis Microtus 91001 (NC_005810); IS1541, 711 bp (p value, 1.139e06).
(G) Y. pestis Pestoides F (NC_009381); IS1541D, 711 bp (p value, 2.479e06).
In (F) and (G), p values were calculated using the potential number of target sequences taking into account the regions of GC skew inversion.

has been estimated to be approximately 20-fold faster than is sion after transitory inactivation of both dnaGts and dnaBts
transcription (800 nt/s versus 20 to 50 nt/s) (Kornberg and Baker, mutants.
1992). Replication forks may stall at transfer RNA and other If excision occurs at the replication fork and requires ssDNA,
highly expressed genes, possibly because transcription the probability that both ends are within the single-stranded
complexes collide ‘‘head on’’ with the forks. Replication forks region of the lagging-strand template should decrease with
also stall upon codirectional encounters with RNA polymerase increasing IS length, and thus the size of the IS should influence
(Elı́as-Arnanz and Salas, 1997; Mirkin et al., 2006), possibly as excision frequency exactly as we observe. Moreover, the effi-
a result of a trapped RNA polymerase not readily displaced ciency of excision was generally higher in the dnaGts mutant
from DNA by fork progression. even at the permissive growth temperature of 30 C and the slope
That ssDNA at the replication fork facilitates IS608 transposi- of the curve was less steep. At the sublethal temperature of
tion is reinforced by studies which perturb the fork using temper- 33 C, excision was even higher and the length dependence
ature sensitive DnaG (primase) or DnaB (helicase) mutants. even less marked. This is consistent with an increase in ssDNA
DnaG inactivation prevents initiation of Okazaki fragment syn- length of on the lagging-strand template resulting from a lower
thesis, increasing the average length of ssDNA upstream of the Okazaki fragment initiation frequency in the dnaGts mutant
first complete Okazaki fragment on the lagging-strand template. even at temperatures permissive for growth and suggests that
(Louarn, 1974; Fouser and Bird, 1983). DnaB inactivation results the slope is a function of the ssDNA length available on the
in accumulation of large amounts of ssDNA mostly likely arising lagging-strand template.
from degradation of both the nascent DNA and leading-strand We also investigated the effect of DnaG overproduction.
template or from uncoupled leading-strand synthesis (Belle Expression of a cloned wild-type dnaG gene not only sup-
et al., 2007). We observed a significant stimulation of IS608 exci- pressed the dnaGts phenotype but also resulted in an even

Cell 142, 398–408, August 6, 2010 ª2010 Elsevier Inc. 405


more pronounced length dependence in the wild-type back- that transposition is infrequent or that transposition events
ground, suggesting that, as observed in vitro (Zechner et al., become genetically fixed in the population.
1992; Sanders et al., 2010), DnaG concentration controls the IS200/IS605 family members are not alone in showing asym-
frequency of initiation and Okazaki fragment size. More impor- metric strand preference in insertion. Other transposable
tantly, this result would suggest that DnaG is not saturating at elements such as Tn7 and IS903 also appear to do so (Peters
the normal replication fork in vivo. and Craig, 2001; Hu and Derbyshire, 1998). Tn7 is targeted to
While the slopes of the curves presumably reflect the length of replication forks during conjugative plasmid transfer and inserts
ssDNA on the lagging-strand template, the explanation for the in a specific orientation. The transposon-encoded TnsE protein
apparent inflection of the curves for the longer transposons is is instrumental in targeting the transpososome to the junction
less clear. It is possible that we are observing the combined between single- and double-stranded DNA by interaction with
effects of two phenomena: for example the initial probability the b clamp component of the replication apparatus (Parks
that both ends are in a single-stranded form and the probability et al., 2009; Chandler, 2009). Insertion of Tn7 into a replication
that both ends find each other. Further analysis is required to fork likely occurs within the dsDNA covered by an Okazaki frag-
determine the exact reason behind this behavior. ment. IS903 insertion bias in the conjugative F plasmid might
Although, for simplicity, we describe the lagging-strand also reflect targeting to the conjugative replication fork. It is
template as single stranded at the fork, it is important to note worthwhile noting that IS10 and IS50 have also exploited host
that it is not naked but protected by proteins such as single- replication: both are activated by transient formation of hemime-
strand binding protein (Ssb) (Shereda et al., 2008). This implies thylated DNA after fork passage (Roberts et al., 1985; Yin et al.,
that the transposition machinery can access the DNA through 1988; Dodson and Berg, 1989).
the protecting protein and raises the question of whether TnpA However, there are major mechanistic differences between
can recognize Ssb or other components of the replisome. Exper- Tn7, IS903, and members of the IS200/IS605 family, suggesting
iments to investigate this are in progress. that different pathways are at play. Perhaps most importantly,
We also used the natural Tus/Ter replication fork termination Tn7 transposes using a dsDNA intermediate in contrast to the
system (Neylon et al., 2005; Kaplan and Bastia, 2009) to examine ssDNA species used by IS608 and ISDra2. For Tn7 and many
whether blocked forks might also attract IS608 insertions. The other transposons with dsDNA intermediates, strand transfer
Tus-Ter complex forms a transient barrier to the replicative heli- into a dsDNA target occurs using the 30 OH groups on each com-
case, DnaB, when a fork arrives in the nonpermissive direction plementary strand at each end. Insertion of the first strand into
(Neylon et al., 2005). When provided with appropriate target tet- a single-strand target would leave a fatal break. Thus, whereas
ranucleotides, Tus-dependent IS608 insertions readily occurred an overarching theme in DNA transposition may be the exploita-
close (26–77 nt) to the Ter site on the lagging-strand template, tion of replication forks, different elements do so in different ways.
consistent with nucleotide resolution mapping of the terminated The unique excision and insertion properties of the IS200/
nascent DNA in vitro and in vivo showing that the final lagging- IS605 family may make them useful tools for probing ssDNA
strand primer sites are arrested 50–70 nucleotides upstream of structures in vivo. The relationship between excision and IS
Ter (Hill and Marians, 1990; Mohanty et al., 1998). While our length might be used to determine the effect of various factors
data are consistent with the idea that stalled forks favor IS608 on the state of the replication fork in vivo. For example, treat-
insertion, we do not yet know whether the ssDNA substrates ments leading to reduced fork velocity, Okazaki fragment
are directly available at blocked forks or are generated during synthesis, uncoupling of lagging from leading-strand synthesis,
their processing, e.g., during replication restart or repair of ds or simply forks blocked by different factors could all be probed
breaks caused by replication arrest (Michel et al., 1997; Bierne using an excision assay as an experimental readout. We are
and Michel, 1994). aware that the topology of the replication fork of small, multicopy
To determine whether other IS200/IS605 family members plasmids may differ in some respects from that of the chromo-
might locate suitable ssDNA substrates for transposition, we some, and this system may also be used to explore these
annotated several complete bacterial genomes for ISs and iden- possible differences. We are at present testing these possibilities
tified several that harbor multiple copies of these family experimentally.
members. In the majority, these ISs showed a similar orientation
bias to IS608 and ISDra2 relative to replication direction. Thus, EXPERIMENTAL PROCEDURES
targeting of the ssDNA available on the lagging-strand template
Bacterial Strains and Media
appears to be a general theme among IS200/IS605 family
Bacterial strains are described in the Extended Experimental Procedures; see
members. also Figure S2 and Table S4. E. coli cultures were grown in Luria broth (LB) sup-
Replication direction and therefore identification of the lagging plemented where necessary with chloramphenicol (Cm, 10 or 30 mg/ml), kana-
strand is generally implied from GC skew, the preference for G mycin (Km, 20 mg/ml), ampicillin (Ap, 100 mg/ml), gentamycin (Gm, 5 mg/ml),
over C on the leading strand thought to be the result of differen- spectinomycin (Sp, 30 mg/ml) and streptomycin (Sm, 20 mg/ml), tetracycline
tial repair (Lobry, 1996; Grigoriev, 1998). More strictly speaking, (Tc, 15 mg/ml), or 2,6-Diaminoheplanediole acid (DAP, 0.006%). D. radiodurans
media and growth conditions were as described (Pasternak et al., 2010; Bona-
we observed that IS orientation was correlated with the GC skew
cossa de Almeida et al., 2002).
of the region into which they were inserted rather than with repli-
cation direction per se, suggesting that the insertions predated Plasmids
the genome rearrangements whose scars are revealed by Details of plasmid constructions are available upon request; see Figure S3,
changes in GC skew. This has two important implications: either Table S4, and the Extended Experimental Procedures for schematics of

406 Cell 142, 398–408, August 6, 2010 ª2010 Elsevier Inc.


plasmids used and additional information. The dnaG-carrying plasmid pBS179 SUPPLEMENTAL INFORMATION
was constructed by insertion of a DNA fragment carrying the wild-type dnaG
allele with its natural ribosome binding site isolated by PCR with flanking Supplemental Information includes Extended Experimental Procedures, three
oligonucleotides, dnaGN, and dnaGC into the SphI site of transposase figures, and four tables and can be found with this article online at doi:10.1016/
supplying plasmid downstream of the tnpA gene, placing both genes under j.cell.2010.06.034.
control of plac. The clone was verified by sequencing and complements the
dnaGts allele. ACKNOWLEDGMENTS

Excision Assay with pAM1 Derivatives We would like to thank members of the ‘‘Mobile Genetic Elements’’ group,
Effect of dnaG308 and dnaB8 on Excision A. Bailone, P. Polard, and D. Lane for discussions, G. Coste and B. Marty
E. coli MG4100 wild-type and dnaG308 and dnaB8 mutant strains were grown for expert technical assistance, L. Lavatine for guiding us through the
overnight at 30 C, the permissive temperature, in LB+KmTc, and cultures were mysteries of statistics, A. Varani for identifying a representative oligonucleotide
diluted at 30 C and grown with 0.5 mM IPTG for 4 hr or 45 min. IPTG was sequence used in the PCR mapping for the D. radiodurans genome, and the
removed by centrifugation and cells were resuspended in prewarmed medium Institut Curie for the use of the 137Cs irradiation system. This work was sup-
at 42 C for 30 min and incubated at 30 C for 3 hr. As a control, the 42 C step ported by: intramural funding from the Centre National de Recherche Scienti-
was omitted. Cells were harvested and dilutions plated on LA+KmTc and fique (France), in its later stages by ANR grant Mobigen (M.C. and S.S.), Euro-
LA+KmTcAp. pean contract LSHM-CT-2005-019023 (M.C.), and the Commissariat à
Effect of Transposon Length l’Energie Atomique and Electricité de France (France; S.S.). At the National
E. coli DH5a harboring pBS135 and pAM1-derivative plasmids was grown Institutes of Health, this work was supported by the Intramural Program of
overnight at 37 C in LB+KmTc. Cultures were diluted in fresh medium at the National Institute of Diabetes and Digestive and Kidney Diseases.
37 C with KmTc and 0.5 mM IPTG to induce TnpA expression. Cells were
harvested after 5 hr and dilutions plated on LA+KmTc and LA+KmTcAp. Received: December 23, 2009
Effect of Transposon Length in the dnaG308 Mutant Strain Revised: April 3, 2010
Overnight cultures of MC4100 and MC4100 dnaGts were grown at 30 C and Accepted: May 17, 2010
diluted in fresh medium at 30 C or 33 C containing KmTc and 0.5 mM IPTG. Published: August 5, 2010
Cells were harvested after 5 hr and dilutions plated on LA+KmTc and
LA+KmTcAp at 30 C. REFERENCES

Barabas, O., Ronning, D.R., Guynet, C., Hickman, A.B., Ton-Hoang, B.,
Tus/Ter Experiments Chandler, M., and Dyda, F. (2008). Mechanism of IS200/IS605 family DNA
Overnight donor and recipient strains were grown in LB+DAP+SpSmCm transposases: activation and transposon-directed target site selection. Cell
and LB+GmKmAp, respectively. Glucose (0.5%) was added to fully repress 132, 208–220.
the para used to drive Tus expression. Cultures were diluted in fresh LB medium
Belle, J.J., Casey, A., Courcelle, C.T., and Courcelle, J. (2007). Inactivation of
without antibiotics (with DAP for donor cells). At an OD600 of 0.5, donor
the DnaB helicase leads to the collapse and degradation of the replication fork:
cells were incubated without agitation, the recipient was diluted to an OD600
a comparison to UV-induced arrest. J. Bacteriol. 189, 5452–5462.
of 0.15, and TnpA was induced with 0.5 mM IPTG. After 60 min, Tus
was induced for 30 min by addition of 0.08% arabinose. Donor and recipient Biémont, C., and Vieira, C. (2006). Genetics: junk DNA as an evolutionary force.
strains were mixed, incubated for 2 hr and plated on Cm-containing LA plates Nature 443, 521–524.
supplemented with 0.5% glucose. Bulk plasmid DNA was isolated and the Bierne, H., and Michel, B. (1994). When replication forks stop. Mol. Microbiol.
distribution of insertions in the population was analyzed by PCR with 13, 17–23.
six sets of primers: B248, B249, tus as forward primers, and LE and RE Bierne, H., Ehrlich, S.D., and Michel, B. (1994). Flanking sequences affect
as reverse primers (Extended Experimental Procedures) to determine the replication arrest at the Escherichia coli terminator TerB in vivo. J. Bacteriol.
insertion orientation. PCR amplification used Phusion DNA Polymerase 176, 4165–4167.
(Finnzyme) in GC buffer as follows: 30 s 98 C, 35 cycles of 10 s 98 C, 30 s
Bonacossa de Almeida, C., Coste, G., Sommer, S., and Bailone, A. (2002).
64 C, and 1 min 72 C. Fifty nanograms of bulk plasmid DNA were used for
Quantification of RecA protein in Deinococcus radiodurans reveals involve-
each reaction.
ment of RecA, but not LexA, in its regulation. Mol. Genet. Genomics 268,
28–41.
Sequencing IS608 Insertions in the E.coli Chromosome Carl, P.L. (1970). Escherichia coli mutants with temperature-sensitive
IS608 insertions into the E. coli chromosome and coresident plasmid pBS135 synthesis of DNA. Mol. Gen. Genet. 109, 107–122.
were sequenced by arbitrary PCR as described (Guynet et al., 2009).
Chain, P.S., Carniel, E., Larimer, F.W., Lamerdin, J., Stoutland, P.O., Regala,
W.M., Georgescu, A.M., Vergez, L.M., Land, M.L., Motin, V.L., et al. (2004).
Measurement of In Vivo Spontaneous and g-Induced Excision Insights into the evolution of Yersinia pestis through whole-genome compar-
Frequencies of ISDra2-113 ison with Yersinia pseudotuberculosis. Proc. Natl. Acad. Sci. USA 101,
Excision frequencies were determined with individual CmR TcS colonies puri- 13826–13831.
fied from strains GY14302 or GY14303 (Extended Experimental Procedures) Chandler, M. (2009). Clamping down on transposon targeting. Cell 138,
grown with or without TnpA expression in trans as described (Pasternak 621–623.
et al., 2010). The mean and standard deviations were calculated from six inde-
Curcio, M.J., and Derbyshire, K.M. (2003). The outs and ins of transposition:
pendent experiments. ISDra2 insertion sites were mapped by arbitrary-primed
from Mu to Kangaroo. Nat. Rev. Mol. Cell Biol. 4, 865–877.
(AP)-PCR with DyNazyme EXT DNA polymerase (Finnzymes) as described
(Extended Experimental Procedures). Demarre, G., Guérout, A.M., Matsumoto-Mashimo, C., Rowe-Magnus, D.A.,
Statistical Analysis of Insertion Orientation Marlière, P., and Mazel, D. (2005). A new family of mobilizable suicide plasmids
This was performed with Fisher’s exact test with the ‘‘R’’ statistical package based on broad host range R388 plasmid (IncW) and RP4 plasmid (IncPalpha)
(http://www.r-project.org/) taking into account the number of potential target conjugative machineries and their cognate Escherichia coli host strains. Res.
sites on the leading and lagging strands. For the Yersinia species, the number Microbiol. 156, 245–255.
of potential target sites and of insertion sites was calculated taking into Deng, W., Liou, S.R., Plunkett, G., 3rd, Mayhew, G.F., Rose, D.J., Burland, V.,
account GC skew inversion. Kodoyianni, V., Schwartz, D.C., and Blattner, F.R. (2003). Comparative

Cell 142, 398–408, August 6, 2010 ª2010 Elsevier Inc. 407


genomics of Salmonella enterica serovar Typhi strains Ty2 and CT18. J. Bac- Michel, B., Ehrlich, S.D., and Uzest, M. (1997). DNA double-strand breaks
teriol. 185, 2330–2337. caused by replication arrest. EMBO J. 16, 430–438.
Dodson, K.W., and Berg, D.E. (1989). Factors affecting transposition activity of Mirkin, E.V., Castro Roa, D., Nudler, E., and Mirkin, S.M. (2006). Transcription
IS50 and Tn5 ends. Gene 76, 207–213. regulatory elements are punctuation marks for DNA replication. Proc. Natl.
Elı́as-Arnanz, M., and Salas, M. (1997). Bacteriophage phi29 DNA replication Acad. Sci. USA 103, 7276–7281.
arrest caused by codirectional collisions with the transcription machinery. Mohanty, B.K., Sahoo, T., and Bastia, D. (1998). Mechanistic studies on the
EMBO J. 16, 5775–5783. impact of transcription on sequence-specific termination of DNA replication
Fouser, L., and Bird, R.E. (1983). Accumulation of ColE1 early replicative inter- and vice versa. J. Biol. Chem. 273, 3051–3059.
mediates catalyzed by extracts of Escherichia coli dnaG mutant strains. Neylon, C., Kralicek, A.V., Hill, T.M., and Dixon, N.E. (2005). Replication termi-
J. Bacteriol. 154, 1174–1183. nation in Escherichia coli: structure and antihelicase activity of the Tus-Ter
complex. Microbiol. Mol. Biol. Rev. 69, 501–526.
Galas, D.J., and Chandler, M. (1982). Structure and stability of Tn9-mediated
cointegrates. Evidence for two pathways of transposition. J. Mol. Biol. 154, Parks, A.R., Li, Z., Shi, Q., Owens, R.M., Jin, M.M., and Peters, J.E. (2009).
245–272. Transposition into replicating DNA occurs through interaction with the proces-
sivity factor. Cell 138, 685–695.
Garcia, E., Worsham, P., Bearden, S., Malfatti, S., Lang, D., Larimer, F.,
Lindler, L., and Chain, P. (2007). Pestoides F, an atypical Yersinia pestis strain Pasternak, C., Ton-Hoang, B., Coste, G., Bailone, A., Chandler, M., and Som-
from the former Soviet Union. Adv. Exp. Med. Biol. 603, 17–22. mer, S. (2010). Irradiation-induced Deinococcus radiodurans genome frag-
mentation triggers transposition of a single resident insertion sequence.
Grigoriev, A. (1998). Analyzing genomes with cumulative skew diagrams.
PLoS Genet. 6, e1000799.
Nucleic Acids Res. 26, 2286–2290.
Peters, J.E., and Craig, N.L. (2001). Tn7 recognizes transposition target struc-
Guynet, C., Hickman, A.B., Barabas, O., Dyda, F., Chandler, M., and Ton-
tures associated with DNA replication using the DNA-binding protein TnsE.
Hoang, B. (2008). In vitro reconstitution of a single-stranded transposition
Genes Dev. 15, 737–747.
mechanism of IS608. Mol. Cell 29, 302–312.
Roberts, D., Hoopes, B.C., McClure, W.R., and Kleckner, N. (1985). IS10 trans-
Guynet, C., Achard, A., Hoang, B.T., Barabas, O., Hickman, A.B., Dyda, F., and
position is regulated by DNA adenine methylation. Cell 43, 117–130.
Chandler, M. (2009). Resetting the site: redirecting integration of an insertion
sequence in a predictable way. Mol. Cell 34, 612–619. Ronning, D.R., Guynet, C., Ton-Hoang, B., Perez, Z.N., Ghirlando, R., Chan-
dler, M., and Dyda, F. (2005). Active site sharing and subterminal hairpin recog-
Hendrickson, H., and Lawrence, J.G. (2006). Selection for chromosome archi-
nition in a new class of DNA transposases. Mol. Cell 20, 143–154.
tecture in bacteria. J. Mol. Evol. 62, 615–629.
Sanders, G.M., Dallmann, H.G., and McHenry, C.S. (2010). Reconstitution of
Hill, T.M., and Marians, K.J. (1990). Escherichia coli Tus protein acts to arrest
the B. subtilis replisome with 13 proteins including two distinct replicases.
the progression of DNA replication forks in vitro. Proc. Natl. Acad. Sci. USA 87,
Mol. Cell 37, 273–281.
2481–2485.
Shereda, R.D., Kozlov, A.G., Lohman, T.M., Cox, M.M., and Keck, J.L. (2008).
Hu, W.Y., and Derbyshire, K.M. (1998). Target choice and orientation prefer- SSB as an organizer/mobilizer of genome maintenance complexes. Crit. Rev.
ence of the insertion sequence IS903. J. Bacteriol. 180, 3039–3048. Biochem. Mol. Biol. 43, 289–318.
Islam, S.M., Hua, Y., Ohba, H., Satoh, K., Kikuchi, M., Yanagisawa, T., and Simons, R.W., Houman, F., and Kleckner, N. (1987). Improved single and
Narumi, I. (2003). Characterization and distribution of IS8301 in the radioresist- multicopy lac-based cloning vectors for protein and operon fusions. Gene
ant bacterium Deinococcus radiodurans. Genes Genet. Syst. 78, 319–327. 53, 85–96.
Johnson, A., and O’Donnell, M. (2005). Cellular DNA replicases: components Song, Y., Tong, Z., Wang, J., Wang, L., Guo, Z., Han, Y., Zhang, J., Pei, D.,
and dynamics at the replication fork. Annu. Rev. Biochem. 74, 283–315. Zhou, D., Qin, H., et al. (2004). Complete genome sequence of Yersinia pestis
Kaplan, D.L., and Bastia, D. (2009). Mechanisms of polar arrest of a replication strain 91001, an isolate avirulent to humans. DNA Res. 11, 179–197.
fork. Mol. Microbiol. 72, 279–285. Ton-Hoang, B., Guynet, C., Ronning, D.R., Cointin-Marty, B., Dyda, F., and
Karpinets, T.V., Obraztsova, A.Y., Wang, Y., Schmoyer, D.D., Kora, G.H., Park, Chandler, M. (2005). Transposition of ISHp608, member of an unusual family
B.H., Serres, M.H., Romine, M.F., Land, M.L., Kothe, T.B., et al. (2010). of bacterial insertion sequences. EMBO J. 24, 3325–3338.
Conserved synteny at the protein family level reveals genes underlying Shewa- Turlan, C., and Chandler, M. (2000). Playing second fiddle: second-strand pro-
nella species’ cold tolerance and predicts their novel phenotypes. Funct. In- cessing and liberation of transposable elements from donor DNA. Trends
tegr. Genomics 10, 97–110. Microbiol. 8, 268–274.
Kersulyte, D., Velapatiño, B., Dailide, G., Mukhopadhyay, A.K., Ito, Y., Vezzi, A., Campanaro, S., D’Angelo, M., Simonato, F., Vitulo, N., Lauro, F.M.,
Cahuayme, L., Parkinson, A.J., Gilman, R.H., and Berg, D.E. (2002). Transpos- Cestaro, A., Malacrida, G., Simionati, B., Cannata, N., et al. (2005). Life at
able element ISHp608 of Helicobacter pylori: nonrandom geographic distribu- depth: Photobacterium profundum genome sequence and expression anal-
tion, functional organization, and insertion specificity. J. Bacteriol. 184, ysis. Science 307, 1459–1461.
992–1002.
Wechsler, J.A., and Gross, J.D. (1971). Escherichia coli mutants temperature-
Kornberg, A., and Baker, T.A. (1992). DNA Replication (New York: W.H. sensitive for DNA synthesis. Mol. Gen. Genet. 113, 273–284.
Freeman). Yin, J.C., Krebs, M.P., and Reznikoff, W.S. (1988). Effect of dam methylation
Lobry, J.R. (1996). Asymmetric substitution patterns in the two DNA strands of on Tn5 transposition. J. Mol. Biol. 199, 35–45.
bacteria. Mol. Biol. Evol. 13, 660–665. Zahradka, K., Slade, D., Bailone, A., Sommer, S., Averbeck, D., Petranovic, M.,
Louarn, J.M. (1974). Size distribution and molecular polarity of nascent DNA in Lindner, A.B., and Radman, M. (2006). Reassembly of shattered chromo-
a temperature-sensitive dna G mutant of Escherichia coli. Mol. Gen. Genet. somes in Deinococcus radiodurans. Nature 443, 569–573.
133, 193–200. Zechner, E.L., Wu, C.A., and Marians, K.J. (1992). Coordinated leading- and
Mennecier, S., Servant, P., Coste, G., Bailone, A., and Sommer, S. (2006). lagging-strand synthesis at the Escherichia coli DNA replication fork. II.
Mutagenesis via IS transposition in Deinococcus radiodurans. Mol. Microbiol. Frequency of primer synthesis and efficiency of primer utilization control
59, 317–325. Okazaki fragment size. J. Biol. Chem. 267, 4045–4053.

408 Cell 142, 398–408, August 6, 2010 ª2010 Elsevier Inc.


A Large Intergenic Noncoding RNA
Induced by p53 Mediates Global Gene
Repression in the p53 Response
Maite Huarte,1,2,* Mitchell Guttman,1,3 David Feldser,3,4 Manuel Garber,1 Magdalena J. Koziol,1,2
Daniela Kenzelmann-Broz,5,6 Ahmad M. Khalil,1,2 Or Zuk,1 Ido Amit,1 Michal Rabani,1 Laura D. Attardi,5,6 Aviv Regev,1,3
Eric S. Lander,1,3,7 Tyler Jacks,3,4 and John L. Rinn1,2,*
1The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
2Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
3Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
4The Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA
5Department of Radiation Oncology
6Department of and Genetics

Stanford University School of Medicine, Stanford, CA 94305, USA


7Department of Systems Biology, Harvard Medical School, Boston, MA 02114, USA

*Correspondence: mhuarte@broadinstitute.org (M.H.), jrinn@broadinstitute.org (J.L.R.)


DOI 10.1016/j.cell.2010.06.040

SUMMARY 4 trimethylated (H3K4me3) promoter region and histone 3-lysine


36 trimethylation (H3K36me3) corresponding to the elongated
Recently, more than 1000 large intergenic noncoding transcript (Guttman et al., 2009). These lincRNAs show clear
RNAs (lincRNAs) have been reported. These RNAs evolutionary conservation, implying that they are functional
are evolutionarily conserved in mammalian genomes (Guttman et al., 2009; Ponjavic et al., 2007).
and thus presumably function in diverse biologi- In an attempt to understand the potential biological roles of
cal processes. Here, we report the identification of lincRNAs, a method to infer putative function based on correla-
tion in expression between lincRNAs and protein-coding genes
lincRNAs that are regulated by p53. One of these
was developed. These studies led to preliminary hypotheses
lincRNAs (lincRNA-p21) serves as a repressor in about the involvement of lincRNAs in diverse biological pro-
p53-dependent transcriptional responses. Inhibition cesses, from stem cell pluripotency to cell-cycle regulation
of lincRNA-p21 affects the expression of hundreds (Guttman et al., 2009). In particular, we observed a group of
of gene targets enriched for genes normally re- lincRNAs that are strongly associated with the p53 transcrip-
pressed by p53. The observed transcriptional repres- tional pathway. p53 is an important tumor suppressor gene
sion by lincRNA-p21 is mediated through the phys- involved in maintaining genomic integrity (Vazquez et al., 2008).
ical association with hnRNP-K. This interaction is In response to DNA damage, p53 becomes stabilized and trig-
required for proper genomic localization of hnRNP- gers a transcriptional response that causes either cell arrest or
K at repressed genes and regulation of p53 mediates apoptosis (Riley et al., 2008).
apoptosis. We propose a model whereby transcrip- The p53 transcriptional response involves both activation and
repression of numerous genes. While p53 is known to transcrip-
tion factors activate lincRNAs that serve as key
tionally activate numerous genes, the mechanisms by which p53
repressors by physically associating with repressive leads to gene repression have remained elusive. We recently
complexes and modulate their localization to sets of reported evidence that many lincRNAs are physically associated
previously active genes. with repressive chromatin modifying complexes and suggested
that they may serve as repressors in transcriptional regulatory
INTRODUCTION networks (Khalil et al., 2009). We therefore hypothesized that
p53 may repress genes in part by directly activating lincRNAs,
It has become increasingly clear that mammalian genomes which in turn regulate downstream transcriptional repression.
encode numerous large noncoding RNAs (Mercer et al., 2009; Here, we show that lincRNAs play a key regulatory role in the
Ponting et al., 2009; Mattick, 2009; Ponjavic et al., 2007). It p53 transcriptional response. By exploiting multiple independent
has been recently reported the identification of more than 1000 cell-based systems, we identify lincRNAs that are transcriptional
large intergenic noncoding RNAs (lincRNAs) in the mouse targets of p53. Moreover, we find that one of these p53-activated
genome (Carninci, 2008; Guttman et al., 2009). The approach lincRNAs—termed lincRNA-p21—serves as a transcriptional
to identify lincRNAs was by searching for a chromatin signature repressor in the p53 pathway and plays a role in triggering apo-
of actively transcribed genes, consisting of a histone 3-lysine ptosis. We further demonstrate that lincRNA-p21 binds to

Cell 142, 409–419, August 6, 2010 ª2010 Elsevier Inc. 409


A +/- Cre Doxorubicin
Figure 1. Several LincRNAs Are p53 Tran-
scriptional Targets
(A) Experiment layout to monitor p53-dependent
LSL/LSL
p53 mRNAs transcription. p53-restored (+Cre) or not restored
MEFs
0 3 6 9 (Cre) p53LSL/LSL MEFs were treated with
DNA damage time (hours)
500 nM dox for 0, 3, 6, and 9 hr (upper left).
+ Cre KRAS (p53LSL/LSL) tumor cells were treated with
LSL/LSL
hydroxytamoxifen for p53 restoration for 0, 8, 16,
KRAS p53 LincRNAs
24, 40, or 48 hr (lower left). RNA was subjected
Lung Tumor 0 8 16 24 40 48
time (hours)
microarray
to microarray analysis of mRNAs and lincRNAs.
p53 restoration profiling (B and C) Venn diagrams showing the number of
shared and distinct mRNAs (B) or lincRNAs (C)
B C induced in a p53-dependent manner in the MEF
MEF MEF or KRAS systems.
MEF & KRAS MEF & KRAS
645 KRAS 995 38 KRAS 32 (D and E) mRNAs (D) and lincRNAs (E) activated by
289 11 p53 induction (FDR < 0.05) in MEF or KRAS
system. Colors represent transcripts above (red)
D E or below (blue) the global median scaled to
0h 3h 6h 9h 0h 8h 16h 24h 40h48h 0h 3h 6h 9h 0h 8h 16h 24h 40h48h
8-fold activation or repression, respectively.
(F) Promoter region, conserved p53 binding
motif, promoter orientation, and p53-dependent
645 mRNAs

995 mRNAs

32 lincRNAs
38 lincRNAs

fold induction in reporter assays of lincRNA


promoters induced in a p53-dependent manner
(values are average of at least three biological
replicates [±STD]; p values are determined by
t test).
(G) p53-dependent induction of lincRNA pro-
-8 +8 -8 +8 moters requires the consensus p53 binding ele-
ments. Relative firefly luciferase expression
driven by promoters with p53 consensus motif
F induction (lincRNA-p21, lincRNA-Mkln1) or with deleted
Name H3K4me3 sequence strand fold induction P value
motif (DlincRNA-p21 and DlincRNA-Mkln1) in
lincRNA-Mkln1 chr6:31061148-31061169 GAACATGCCTGGCACTTGCAAA - 13.5 +/-1.6 5.9E-06
p53-restored p53LSL/LSL (p53+/+) or p53LSL/LSL
lincRNA-p21 chr17:29214831-29220756 GGCCTTGCCCGGGCTTGCCT - 24.6 +/-2.3 1.3E-05
cdkn1a chr17:28818329-28822000 CTGGGCATGTCTGGGCAGCGATC + 16.8 +/-1.2 3.7E-05 (p53/) cells. Values are relative to p53/ and
normalized by renilla levels (average of three repli-
cates ±STD).
G H (H) p53 specifically binds to p53 motifs in lincRNA
147 promoters. p53 ChIP enrichment in p53+/+ and
(Normalized Firefly Luciferase Activity)

30
117 p53/ MEFs on regions with p53 motifs (lincRNA-
Relative Reporter Induction

25 35 p21, lincRNA-Mkln1, Cdkn1a) or two irrelevant


Relative Fold Enrichment

+/+ +/+
p53 30 p53
20 -/- -/- regions (controls). Enrichment values are relative
(p53 ChIP/IgG)

p53 p53
25
15 20
to IgG and average of 3 replicates (±STD).
15 See also Figure S1 and Table S1.
10
10
5
5
0 0
l
21

n1

a
21

n1

n1

l-1

l-2
ro
n1

n1
p2
nt

ro

ro
kl
p

kl

kl
dk

dk
A-

A-

co

A-

nt

nt
M

M
C

co

co
A-

A-
N

A-
N
cR

cR

cR
N

N
cR

cR

cR
lin

in

lin
Δl

lin

in

lin
Δl

hnRNP-K. This interaction is required for proper localization of experimental systems that allow us to monitor gene expression
hnRNP-K and transcriptional repression of p53-regulated genes. changes at different times after p53 induction (Ventura et al., 2007).
Together, these results reveal insights into the p53 transcrip- The first system uses mouse embryonic fibroblasts (MEFs)
tional response and lead us to propose that lincRNAs may serve derived from mice where the endogenous p53 locus is inacti-
as key regulatory hubs in transcriptional pathways. vated by insertion of a transcriptional termination site flanked
by loxP sites (LSL) in the first intron. This endogenous p53 locus
RESULTS (p53 LSL/LSL) is restorable by removal of the stop element by Cre
recombination (Ventura et al., 2007). The p53 LSL/LSL MEFs were
Numerous LincRNAs Are Activated treated with AdenoCre virus expressing the Cre recombinase to
in a p53-Dependent Manner reconstitute the normal p53 allele or AdenoGFP control virus to
As a first attempt to dissect the functional mechanisms of maintain the inactive p53 LSL/LSL allele. Then we compared the
lincRNAs, we focused on a strong association in the expression transcriptional response between the p53-reconstituted and
patterns of certain lincRNAs and genes in the p53 pathway p53 LSL/LSL MEFs after 0, 3, 6, and 9 hr of DNA damage treatment
(Guttman et al., 2009). In order to determine whether these with doxorubicin (we will refer to this system as ‘‘MEFs’’)
lincRNAs are regulated by p53, we employed two independent (Figure 1A). The second system uses a lung tumor cell line

410 Cell 142, 409–419, August 6, 2010 ª2010 Elsevier Inc.


derived from mice expressing an oncogenic K-Ras mutation transcriptional reporter assays for these lincRNAs. Specifically,
(K-RasG12D) and a restorable p53 knockout allele (p53 LSL/LSL), we cloned their promoters (as defined by the H3K4me3 peaks
similar to that described above (D.F. and T.J., unpublished data). [Guttman et al., 2009]) into a luciferase reporter vector (Experi-
We compared the transcriptional response at different times mental Procedures) and transfected the constructs along with
(0, 8, 16, 24, 40, and 48 hr) after restoration of p53 expression a vector to normalize transfection efficiency. Both promoters
by Cre recombination (Experimental Procedures) (we will refer showed significant induction of firefly luciferase in p53-wild-
to this system as ‘‘KRAS’’) (Figure 1A). type but not in p53 null cells (p < 0.01) (Figures 1F and 1G).
To assess the transcriptional responses in each of these To determine whether the canonical p53-binding motif is
systems, we isolated total RNA at each time point before required for the observed transactivation, we repeated these
and after p53 restoration and performed DNA microarray anal- experiments in the absence of the p53-binding motif. Mutant
ysis to monitor protein-coding gene expression levels. In the promoters resulted in the abolition of the observed transactiva-
MEF system and KRAS systems, we identified a total of 1067 tion for both lincRNA-p21 and lincRNA-Mkln1 in p53+/+ cells
(645 activated, 422 repressed) and 1955 (995 activated, 960 (Figure 1F). Finally, we performed chromatin immunoprecipita-
repressed) genes, respectively, that were regulated in a p53- tion (ChIP) experiments to determine whether p53 directly binds
dependent manner (false discovery rate [FDR] < 0.05) (Figures to the sites containing the consensus motifs in vivo. Indeed, p53
1B and 1D and Table S1, parts A and B, available online). The is bound to the site containing the consensus motif in the
sets of p53-induced genes identified in the two systems showed promoters of both lincRNA-p21 and lincRNA-Mkln1 in p53+/+
significant (p < 108) overlap, including such canonical p53 but not p53/ MEFs treated with doxorubicin, and it is not
targets as Cdnk1a, Mdm2, Perp, and Fas (Table S1, parts A enriched at negative control sites of irrelevant regions (Figure 1H
and B). There are also a number of p53-induced genes unique and Extended Experimental Procedures). Together, these
to each system, likely reflecting specific properties of each results demonstrate that lincRNA-p21 and lincRNA-Mkln1 are
cell-type (Levine et al., 2006) (Figure S1C and Table S1, parts bona fide p53 transcriptional targets.
A and B).
Functional analysis of the classes of genes that are enriched LincRNA-p21 Is Induced by p53 in Different Cell Systems
among the genes regulated by p53 in both the MEF and KRAS We were intrigued by the p53 transcriptional target lincRNA-p21,
systems showed strong enrichment for known p53-regulated which resides 15 Kb upstream of the gene encoding the critical
processes, such as the cell cycle and apoptosis (Figures S1A cell-cycle regulator Cdkn1a (also known as p21), a canonical
and S1B). Moreover, gene set enrichment analysis (GSEA) (Sub- transcriptional target of p53 (Riley et al., 2008) (Figures 2A–2C,
ramanian et al., 2005) of previously published microarray anal- Figure S2A, and Table S1, parts C–E). Given the proximity of
yses revealed a significant overlap with the p53 regulated genes lincRNA-p21 to the Cdkn1a gene, we sought to ensure that the
identified here (Table S1, parts H and I). Together these results lincRNA transcript is distinct from that of the Cdkn1a gene. To
demonstrate that these two systems are largely reflective of this end, we cloned the full-length transcriptional unit of lincRNA-
canonical p53 transcriptional responses. p21 using the 50 and 30 RACE method (Schaefer, 1995); the
We next examined lincRNAs regulated by p53 in these two transcript contains two exons comprising 3.1 Kb (Figure 2B).
systems across the same time course, by analogously using In support of lincRNA-p21 being an independent transcript,
a custom tiling microarray representing 400 lincRNAs and lincRNA-p21 is transcribed in the opposite orientation from the
analyzing the data with previously described statistical methods Cdkn1a gene. Furthermore, the analysis of chromatin structure
(Guttman et al., 2009) (Experimental Procedures). We found 38 in mouse embryonic stem cells (mESCs) (Mikkelsen et al., 2007)
and 32 lincRNAs induced by p53 in the MEF and KRAS systems, indicates that these are distinct genes with distinct promoters
respectively (Figures 1C and 1E and Table S1, parts C–G). Inter- (Figure S2A).
estingly, 11 lincRNAs were induced by p53 in both model We next examined the transcriptional regulation of lincRNA-
systems (Figure 1C and Table S1, part C), many more than p21 in two additional cancer-derived cell lines. Specifically, we
expected by chance (p < 106). These results confirm that, in irradiated p53LSL/LSL mice to induce lymphomas and sarcomas
a manner similar to canonical p53 protein coding gene targets, and then restored p53 expression in tumor-derived cells (Exper-
numerous lincRNAs are temporally regulated by p53. imental Procedures) (Ventura et al., 2007). In cells derived in both
tumor types, lincRNA-p21 was strongly induced after p53 resto-
LincRNAs Are Direct Transcriptional Targets of p53 ration. Moreover, the induction of lincRNA-p21 followed similar
We sought to identify lincRNAs that might be canonical p53 kinetics as those of p53 and Cdnk1a, consistent with lincRNA-
target genes. As a first approach, we examined the promoters p21 being a primary transcriptional target of p53 (Figure 2D
of p53-induced lincRNA for enrichment of evolutionarily con- and Figure S2B).
served p53-binding motifs (Garber et al., 2009) (Extended We further investigated the orthologous lincRNA-p21 locus in
Experimental Procedures). The promoters of the p53-induced the human genome. We first mapped the promoter (H3K4me3
lincRNAs were highly enriched for conserved p53 motifs relative domain) of lincRNA-p21 to human genome. Interestingly, this
to the promoters of all lincRNAs (p < 0.01). We selected two region corresponds to one of four intergenic p53-binding sites
lincRNAs whose promoter regions contain highly conserved identified from a study by Wei et al. (2006) (Figure 2B) performing
canonical p53-binding motifs (el-Deiry et al., 1992; Funk et al., p53 ChIP followed by sequencing. Next, we mapped the
1992); we termed these lincRNA-p21 and lincRNA-Mkln1 (with lincRNA-p21 exonic structures to the human genome to deter-
the names referring to the neighboring gene). We next performed mine whether this region is expressed and induced by DNA

Cell 142, 409–419, August 6, 2010 ª2010 Elsevier Inc. 411


A 75 kb Figure 2. LincRNA-p21: A p53 Target Gene
Induced in Different Tumor Models
(A) Schematic representation of the chromosomal
Chr17 Sfrs3 lincRNA-p21 Cdkn1a location of the lincRNA-p21 gene locus. Arrow-
heads indicate the orientation of transcription.
(B) Promoter and transcript structure of lincRNA-
B C p21 gene locus. Chromatin structure at the
lincRNA-p21 locus is shown as mESC ChIP-Seq
p53 motif

GA G A G C TG
A C T C C GGGC A TC 6
human lincRNA-p21 data (Mikkelsen et al., 2007); for each histone
modification (green, H3K4me3; blue, H3K36me3),

Relative RNA level


G G C

A C
G
A C G
T C
T T T G A A
T T
T C
A A G G T
G T A C G G
CT C
T
5
T C
T T
G
T A A A C
T
C T C A A A

PET overlap ChIP-seq results are plotted as number of DNA


4 Untreated
density
Dox
fragments obtained at each position relative to
* 153 bp
500 bp 3
the genomic average. Red stars indicate the posi-
PETS 2
tion of the p53-binding motif. The promoter region
1
where p53 ChIP-PET fragments (black segments)
0
RT-PCR -RT map is enlarged (Wei et al., 2006). PET overlap

H3K4Me3
[ *
density (gray) and p53 motif sequence are shown.
The structure of the full-length lincRNA-p21 is rep-
resented with red boxes as exons and arrowed

H3K36Me3
[ lines as the intronic sequence.
(C) Human lincRNA-p21 is induced by DNA
damage. Relative RNA levels of human lincRNA-

RNA
[ lincRNA-p21
3073 bp
p21 determined by qRT-PCR (RT-PCR) or qPCR
(RT) from untreated human fibroblasts or 500 nM
DOX-treated for 14 hr. PCR primers map on the
human region orthologus to the first exon of the
mouse gene.
D LUNG TUMOR SARCOMA LYMPHOMA (D) LincRNA-p21 is induced by p53 in different
Relative RNA level

tumor cell lines. LincRNA-p21, p53, and Cdkn1a


Relative RNA level

Relative RNA level

2 1.2 1.2
1.7 1 1 relative RNA levels at different times after p53
1.3 0.8 0.8 lincRNA-p21
1 0.6 0.6 Cdkn1a restoration.
p53
0.7 0.4 0.4 Values in (C) and (D) are the median of four tech-
0.3 0.2 0.2
nical replicates (±STD).
0 0 0
0 8 16 24 40 48 0 12 24 36 48 0 12 24 36 48 72 See also Figure S2.
time after p53 restoration time after p53 restoration time after p53 restoration
(hours) (hours) (hours)

damage in human fibroblasts. Indeed, qRT-PCR showed that the pool was effective at knocking down its intended target genes
orthologous 50 exon region (adjacent to observed p53 ChIP in p53LSL/LSL restored MEFs (Figures 3A and 3B). We then
binding site by Wei et al.) of human lincRNA-p21 is expressed used microarray analysis to examine the broader transcriptional
and strongly induced in human fibroblasts upon DNA damage consequences of knockdown of p53 and lincRNA-p21. We iden-
(Figure 2C and Extended Experimental Procedures). tified 1520 and 1370 genes that change upon knockdown of p53
Collectively, these results provide evidence that both the and lincRNA-p21, respectively (relative to nontargeting control
human and mouse lincRNA-p21 promoters are bound by p53 siRNA, FDR < 0.05). We observed a remarkable overlap of
resulting in transcriptional activation in response to DNA 930 genes in both the lincRNA-p21 and p53 knockdowns, vastly
damage. Moreover, lincRNA-p21 is induced by p53 in diverse more than would be expected by chance (p < 10200) (Figure 3C,
biological contexts, including multiple different tumor types Figure S3A, and Table S2). Strikingly, 80% (745/930) of the
(Figure 2D and Figure S2B), suggesting that lincRNA-p21 plays common genes are derepressed in response to both p53 and
a role in the p53 pathway. lincRNA-p21 knockdown, much higher proportion than ex-
pected by chance (p < 1010) (Figure 3C and Table S2) when
LincRNA-p21 as a Repressor in the p53 Pathway compared to all genes affected by the p53 knockdown (Fig-
We next investigated the consequence of the loss of lincRNA- ure S3A). This observation suggests that lincRNA-p21 partici-
p21 function in the context of the p53 response. We reasoned pates in downstream p53 dependent transcriptional repression.
that, if lincRNA-p21 plays a role in carrying out the p53 transcrip- To demonstrate that the observed derepression upon
tional response, then inhibition of lincRNA-p21 would show lincRNA-p21 knockdown is indeed p53 dependent and is not
effects that overlap with inhibition of p53 itself. To test this due to off target effects of the RNAi-mediated knockdown, we
hypothesis, we performed RNA interference (RNAi)-mediated performed several additional experiments and analyses. First,
knockdown of lincRNA-p21 and p53 separately and monitored we repeated the knockdown experiments with four individual
the resulting changes in mRNA levels by DNA microarray siRNAs targeting lincRNA-p21, transfected separately rather
analysis. than in a pool and confirmed the derepression effect on select
Toward this end, we first designed a pool of small interfering target genes (Figure S3F and Table S2). Second, we confirmed
RNA (siRNA) duplexes targeting lincRNA-p21, a pool targeting that the same genes that were derepressed in the lincRNA-p21
p53 or nontargeting control sequences. We validated that each and p53 knockdown experiments correspond to genes that are

412 Cell 142, 409–419, August 6, 2010 ª2010 Elsevier Inc.


A B C Figure 3. LincRNA-p21 Is a Global
Repressor of Genes in the p53 Pathway
1.2 siRNA (A) RNAi-mediated knockdown of lincRNA-p21
Relative RNA level

1
p53 l NA and p53. Relative RNA levels determined by
0.8 lincRNA-p21 tro cR 1 p53
on lin -p2 53 p53 lincRNA
0.6 c p 1520
&
lincRNA-p21 -p21 qRT-PCR in p53-reconstitued p53LSL/LSL MEFs
0.4 p53 930 1370
transfected with the indicated siRNAs and treated
0.2
βActin with DOX (median of four technical replicates
0
siRNA siRNA siRNA
control lincRNA-p21 p53
±STD).
(B) p53 protein levels after lincRNA-p21 and p53
knockdown from cells treated as in (A). bActin
D levels are shown as loading control.
MEF KRAS
(C) Many genes are corepressed by lincRNA-p21
0.4
* 0.4 and p53. Top: Venn diagram of differentially ex-
*
Enrichment Score

Enrichment Score

0.2 0.2 80% pressed genes (FDR < 0.05) upon p53 knockdown
0.0 0 (left) or lincRNA-p21 knockdown (right); cells were
-0.2
treated as in (A) and subjected to microarray anal-
-0.2
Repressed Induced Repressed Induced ysis. Bottom: expression level of genes in lincRNA-
-0.4 by p53 by p53 -0.4
by p53 by p53
20%
p21 and p53 siRNA-treated cells relative to control
* FDR<0.001 * FDR<0.002 siRNA experiments. Expression values are dis-
siRNA siRNA siRNA P< 10 -10
control p53 lincRNA-p21
played in shades of red or blue relative to the
global median expression value across all experi-
-8 +8
ments (linear scale).
E p53 (D) Genes derepressed by lincRNA-p21 and p53
knockdown overlap with the genes repressed by
p53 restoration in the MEF and KRAS systems.
The black line represents the observed enrichment
LinRNA-p21
score profile of genes in the lincRNA-p21/p53
derepressed gene set to the MEF or KRAS gene
sets, respectively.
Cabc1 Fas Bax Phka2 Noxa Perp Stat3 Atf2 Bcl2l3 G2e3 Vcan Mtap4 Mapk3 CyclinD2 Cyclin G Cdk4 Reprimo Mdm2 Cdkn1a Cdkn1b Cdkn2a Wt1 Brca1
(E) Genes corregulated by lincRNA-p21 and p53
APOPTOSIS CELL CYCLE ARREST
are part of the p53 biological response. Examples
of genes affected by lincRNA-p21 and/or p53
siRNA-knockdown (FDR < 0.05). Downregulated
and upregulated genes are indicated with blue
arrows and red lines respectively.
See also Figure S3 and Table S2.

normally repressed upon p53 induction in both the KRAS and increase in viability after DNA damage of cells treated with
MEF systems, in the absence of RNAi treatment (GSEA FDR < siRNAs targeting either lincRNA-p21 or p53 compared to those
0.002) (Figure 3D). Third, we demonstrated that enforced treated with the control siRNA pool (Figures 4A and 4B). The
expression of lincRNA-p21 (Experimental Procedures) also per- increase in viability was greater for knockdown of p53, but was
turbed the expression of genes that are normally regulated by still highly significant for knockdown of lincRNA-p21 (p < 0.01).
p53 in both the KRAS and MEF systems (GSEA FDR < 0.01) We observed similar results when using three individual siRNA
(Figure S3H). Finally, we repeated the siRNA experiments in duplexes targeting lincRNA-p21, as well as two different control
the absence of p53 (dox/-AdCre) and demonstrated that dere- siRNA pools (Figure 4B and Figures S4A–S4C). These results
pression of these genes did not occur upon siRNA-mediated suggest that lincRNA-p21 plays a physiological role in regulating
knockdown in the absence of p53 (Figure S3I). Collectively, cell viability upon DNA damage in this system, although they do
these results indicate that lincRNA-p21 acts to repress many not discriminate whether the effect is due to misregulation of the
genes in p53-dependent transcriptional response. cell cycle or apoptosis.
To distinguish between these two possibilities, we first exam-
lincRNA-p21 Regulates Apoptosis ined whether cell-cycle regulation in response to DNA damage is
The activation of the p53 pathway has two major phenotypic affected by knockdown of p53 and lincRNAp-21. Specifically,
outcomes: growth arrest and apoptosis (Levine et al., 2006). we assayed 5-bromo-2-deoxyuridine (BrdU) incorporation and
Consistent with this, our microarray analysis demonstrates that propidium iodide staining of the cells by fluorescence-activated
p53 and lincRNA-p21 both regulate a number of apoptosis cell sorting (FACS) analysis. Consistent with the ability of p53
and cell-cycle regulator genes (Figure 3E, Figure S3G, and to inhibit cell-cycle progression, knockdown of p53 caused a
Table S2, parts A and B). Thus, we aimed to determine the phys- significant increase in BrdU incorporation in response to DNA
iological role of lincRNA-p21 in these processes. damage (p < 0.01). In contrast, knockdown of lincRNA-p21
Toward this end, we used RNAi-mediated knockdown of showed no significant changes in either BrdU levels or in the
lincRNA-p21 in dox-treated or untreated primary MEFs. We simi- percentages of cells in any of the cell-cycle phases (S, G1, or
larly performed RNAi-mediated knockdown of p53 (as a positive G2) with or without dox treatment (Figure 4C). These results
control) or used the nontargeting siRNA pool (as a negative suggest that lincRNA-p21 does not substantially contribute to
control) under the same conditions. We observed a significant cell-cycle arrest upon DNA damage.

Cell 142, 409–419, August 6, 2010 ª2010 Elsevier Inc. 413


A B C Figure 4. LincRNA-p21 Is Required for
untreated
6
untreated
4 Proper Apoptotic Induction
Relative cell number

Fold change relative


(A) Increased cell viability of lincRNA-p21-

to siRNA control
5 G1
3
4 G2
siRNA p53 S depleted cells. Relative number of siRNA-trans-
3 2
siRNA control fected MEFs treated with 400 nM DOX from
2 siRNA lincRNA-p21
siRNA control-1 siRNA control-2 1
1 24 hr after transfection (right) or untreated (left)
0 0 62 20 18 59 2417 55 22 23
0 24 48 72 siRNA siRNA siRNA determined by MTT assay.
time post transfection (hours) control lincRNA-p21 p53
(B) Knockdown of lincRNA-p21 with individual
DOX siRNAs increases cell viability. Images of MEFs
siRNA p53-1 siRNA p53-2 4 treated with different individual siRNAs after

Fold change relative


DOX

to siRNA control
Relative cell number

2.0
3 G1 * 48 hr of DOX treatment (72 hr after transfection).
1.6 G2
siRNA p53 2 S (C) LincRNA-p21 knockdown doesn’t affect cell-
1.2
siRNA control
0.8 siRNA lincRNA-p21 1 cycle regulation. Relative cell numbers in each
0.4
siRNA siRNA 0 57 34 9 56 33 11 29 44 27 cell-cycle phase determined by FACS of BrdU
0 lincRNA-p21-1 lincRNA-p21-2 siRNA siRNA siRNA
0 24 48 72 control lincRNA-p21 p53
incorporation and PI staining of MEFs treated as
time post transfection (hours)
in (A). Numbers inside bars represent percentages
D E of cells in each phase.
30 (D) LincRNA-p21 knockdown causes a decrease
in cellular apoptosis. p53-reconstituted p53LSL/LSL

% of apoptotic cells
25
siRNA control-1 siRNA control-2 siRNA p53 20
1.81 14.6 1.9 14.3 1.9 5.96
MEFs transfected with three individual siRNAs
15
10
* * targeting lincRNA-p21 (bottom), two independent
5 control siRNAs (upper left and middle) or a siRNA
0 pool targeting p53 (upper right). Twenty-four hours
7-AAD
7-AAD
7-AAD

si p53
lin
si

si NA
after transfection, cells were treated with 400 nM

R
R tro

R -p
cR
81.9 1.64 83.1 1.26 90.9 1.26
co

N
N l

N 2

A
n
A

A 1
Annexin-V Annexin-V Annexin-V
F doxorubicin and 14 hr later were harvested and
siRNA lincRNA-p21-1 siRNA lincRNA-p21-2 siRNA lincRNA-p21-3 subjected to FACS analysis. The x axis represents
siRNA
1.87 9.39 1.8 6.56 1.93 5.5
Annexin-V and the y axis 7-AAD staining. The
21 A
-p RN
l
ro

cp
3
nt

percentage of cells in each quadrant is indicated.


p5
co

lin

Cleaved (E) Decreased apoptosis caused by lincRNA-p21


caspase 3
7-AAD
7-AAD
7-AAD

knockdown. Percentage of Annexin-V-positive


87.1 1.61 91.3 0.92 92.49 1.35 βActin
Annexin-V Annexin-V Annexin-V cells (FACS) at 38 hr after transfection (14 hr of
G 400 nM DOX treatment) in MEFs treated as in (A).
DOX
(F) LincRNA-p21 knockdown in p53-reconstituted
untreated
2.5 p53LSL/LSL MEFs causes decrease in Caspase 3
Relative cell number

25
Relative cell number

20 control vector
2
control vector
cleavage. Levels of cleaved Caspase 3 or control
15 lincRNA-p21
1.5
lincRNA-p21 bActin in p53 reconstituted-p53LSL/LSL MEFs
10 1
treated with the indicated siRNA pools and
5 0.5
500 nM DOX for 14 hr.
0 0
0 24 48 72 0 24 48 72 (G) Decreased cell viability caused by lincRNA-p21
time after selection (hours) time after selection (hours)
overexpression. Relative numbers of LKR cells
overexpressing lincRNA-p21 or control plasmid
determined by MTT assay.
H DOX I DOX
8
(H) Overexpression of lincRNA-p21 causes cellular
% of apoptotic cells

7 * 70
60
control vector
apoptosis under DNA damage induction. Per-
6 50 lincRNA-p21
% of cells

5 centage of Annexin-V-positive LKR cells overex-


4 40
3 30
2 20 pressing lincRNA-p21 or control vector treated
1 10 with 500 nM DOX.
0 0
control lincRNA-p21 G1 G2 S (I) LincRNA overexpression doesn’t affect cell-
cycle regulation. Cell-cycle analysis of DOX-
treated LKR cells overexpressing lincRNA-p21 or
control plasmid.
All values are the average of 3 biological replicates
(±STD). * p < 0.01 relative to controls.
Also see Figure S4.

We then examined the impact of lincRNA-p21 and p53 knock- p21 would result in an increased apoptosis. Indeed, lincRNA-
downs on apoptosis. To this end, we assayed the proportion of the p21 overexpression in a lung cancer cell line harboring a KRAS
cell population undergoing apoptosis by measuring Annexin-V by mutation (referred to as LKR) and in NIH/3T3 MEFs caused a sig-
FACS analysis. We observed a significant decrease in the number nificant decrease in cell viability (Experimental Procedures, Fig-
of apoptotic cells after DNA damage in both the lincRNA-p21 and ure 4G, and Figure S4E). This decrease in viability was due to
p53 depleted cells relative to the siRNA control (p < 0.01) (Figures increased apoptosis in response to DNA damage (p < 0.01) and
4D and 4E). We also observed a decrease in Caspase 3 cleavage not to an effect in cell-cycle regulation (Figures 4H and 4I and
after knockdown of both p53 or lincRNA-p21, relative to controls Figure S4G). Together, these results demonstrate a reproducible
(Figure 4F). We next sought to determine whether, conversely to and similar reduction of apoptotic cells in response to DNA
lincRNA-p21 knockdown, the enforced expression of lincRNA- damage in both lincRNA-p21 and p53 knockdown experiments.

414 Cell 142, 409–419, August 6, 2010 ª2010 Elsevier Inc.


A Biotinylated
B se p2
1
D Figure 5. LincRNA-p21 Physically Interacts
RNA en A-
its A
an RN cRN with hnRNP-K
lin Native RIP
7 (A) Schematic representation of RNA pulldown

Fold Enrichment (RIP/IgG)


Nuclear 6
experiments to identify associated proteins. Bioti-
lincRNA-p21
extract 5 βActin
4
nylated lincRNA-p21 or antisense RNA were incu-
Gapdh
3 bated with nuclear extracts, targeted with strepta-
2 vidin beads, washed, and associated proteins
1
60 kDa hnRNP-K resolved in a gel. Specific bands were cutout and
0
E identified by mass spectrometry.
(B) LincRNA-p21 and hnRNP-K specifically
X-linked RIP interact in vitro. SDS-PAGE gel of proteins bound
Streptavidin 40 to lincRNA-p21 (right lane) or antisense RNA (left

Fold Enrichment (RIP/IgG)


lincRNA-p21
beads p53
30 βActin
lane). The highlighted region was submitted for
mass spectrometry identifying hnRNP-K as the
C 20
hnRNP-K band unique to lincRNA-p21.
10 (C) Western blot analysis of the specific associa-
Mass tion of hnRNP-K with lincRNA-p21. A nonspecific
Nono 0
Spectrometry
hnRNP-K hnRNP-K protein (NONO) is shown as a control.
antibody 1 antibody 2
(D) Association between endogenous lincRNA-
p21 and hnRNP-K in the nucleus of DNA damaged
MEFs in native conditions. RNA Immunoprecipita-
F 1 2 3 4 5 6 7 8 9 10 G tion (RIP) enrichment is determined as RNA asso-
16 * *
* * ciated to hnRNP-K IP relative to IgG control.
% apoptoptotic cells

hnRNP-K
12
(E) Physical association between lincRNA-p21 and
8 hnRNP-K after chemical crosslinking of life cells.
RNA
4 hnRNP-K was immunoprecipitated from nuclear
extracts of formaldehyde-crosslinked DNA-dam-
0
5’ 3’ length
aged MEFs, and associated RNAs were detected
ve

5’

Fu

3’

3’

1 778 nt
ct

77

26

18
ll
or

le
8

33

89

2 938 nt by RT-qPCR. The relative enrichment is calcu-


ng
nt

nt

nt
th

3 1459 nt
4 2125 nt
hnRNP-K Apoptosis
lated as in (D) and is the median of three techni-
5 3073 nt
6 3073 nt (antisense) Interaction Induction cal replicates of a representative experiment
7 2844 nt 5’ 778 nt + - (±STD).
8 2633 nt Full length + +
9
10
1889 nt
735 nt 3’ 2633 nt - - (F) LincRNA-p21 binds hnRNP-K through its 50
3’ 1889 nt - - terminal region. RNAs corresponding to dif-
ferent fragments of lincRNA-p21 or its antisense
sequence (middle and bottom) were treated as in (A) and associated hnRNP-K was detected by western blot (top).
(G) Percentage of Annexin-V-positive LKR cells overexpressing the indicated lincRNA-p21 fragments or empty vector as control (average of three replicates
[±STD]). * p < 0.001.
See also Figure S5.

Although MEFs typically respond to DNA damage by under- strate that lincRNA-p21 plays an important role in the p53-
going cell-cycle arrest rather than apoptosis (Kuerbitz et al., dependent induction of cell death.
1992), several additional lines of evidence are consistent with
the observed apoptosis phenotype in response to knockdown LincRNA-p21 Functions through Interaction
on p53 and lincRNA-p21. First, certain critical cell-cycle regula- with hnRNP-K
tors, such as Cdkn1a/p21, Cdkn2a, and Reprimo, are regulated We next wanted to investigate the mechanism by which
by p53 but not lincRNA-p21. For example, knockdown of lincRNA-p21 mediates transcriptional repression. We have
lincRNA-p21 perturbs neither the transcript levels of Cdkn1a/ recently reported that many lincRNAs regulate gene expression
p21 nor the protein stability (Figure S3E); this may explain why through their interaction with several chromatin regulatory com-
lincRNA-p21 knockdown is insufficient to cause a cell-cycle plexes (Khalil et al., 2009). Thus, we hypothesized that lincRNA-
phenotype, yet the p53 knockdown is. Second, we observed p21 could affect gene expression in a similar manner.
that both lincRNA-p21 and p53 knockdowns resulted in the To test this, we first performed nuclear fractionation experi-
repression of apoptosis genes (Noxa and Perp) and derepres- ments and confirmed that lincRNA-p21 is enriched in the nucleus
sion of cell survival genes (Bcl2l3, Stat3, and Atf2, among others) (Figure S5A). We next sought to identify proteins that are associ-
(Figure 3E and Table S2). Moreover, the decrease of apoptotic ated with lincRNA-p21 by an RNA-pulldown experiment. Specif-
cells in response to knockdown of lincRNA-p21 was comparable ically, we incubated in vitro-synthesized biotinylated lincRNA-
to that caused by knockdown of p53 (Figures 4D and 4E and p21 and antisense lincRNA-p21 transcripts (negative control)
Figures S4D and S4E). Third, the apoptosis phenotype is depen- with nuclear cell extracts and isolated coprecipitated proteins
dent on the dosage of dox-induced DNA damage (Figure S4D). with streptavidin beads (Experimental Procedures). We resolved
Thus, the apoptosis response is both p53 dependent and the RNA-associated proteins on a SDS-PAGE gel, cut out the
lincRNA-p21 dependent, with this dependence confirmed in bands specific to lincRNA-p21, and subjected them to mass
multiple cell types and conditions (Figures 4B, 4D, 4F, and 4H spectrometry (Figures 5A and 5B). In all six biological replicates,
and Figures S4A–S4C). Collectively, these observations demon- mass-spectrometry analysis identified heterogeneous nuclear

Cell 142, 409–419, August 6, 2010 ª2010 Elsevier Inc. 415


ribonucleoprotein K (hnRNP-K) as specifically associated with knockdowns (FDR < 0.05) (Figure 6 and Figure S6). Remarkably,
the sense (but not antisense) strand of lincRNA-p21. We inde- 83% of these common genes were derepressed in all three
pendently verified this interaction by western blot analysis knockdown experiments (Figure 6A and Figure S6D). The genes
(Figure 5C). hnRNP-K has been shown to play various roles in previously identified as coregulated by lincRNA-p21 and
the p53 pathway (Kim et al., 2008; Moumen et al., 2005). Interest- p53 also were strongly enriched (GSEA FDR < 104) among
ingly, among these roles, Kim et al. (2008) demonstrated that those regulated by hnRNP-K (Figure 6B and Table S3). Thus,
hnRNP-K is a component of a repressor complex that acts in lincRNA-p21 and hnRNP-K play roles in repressing a significant
the p53 pathway, consistent with our evidence that lincRNA- common set of genes in the p53-dependent response to DNA
p21 plays a role in global repression in this pathway. damage.
To further validate the interaction between lincRNA-p21 and We further reasoned that if hnRNP-K is involved in the repres-
hnRNP-K in our cell-based systems, we performed RNA immu- sion of genes corepressed by p53 and lincRNA-p21, then
noprecipitation (RIP) with an antibody against hnRNP-K from hnRNP-K might also be physically bound to the promoters of
nuclear extracts of MEFs subjected to DNA damage. We these genes. To test this, we performed ChIP experiments with
observed an enrichment of lincRNA-p21 (but not other unrelated antibodies against hnRNP-K, followed by hybridization to DNA
RNAs) with hnRNP-K antibody as compared to the nonspecific tiling microarrays covering 30,000 gene promoters. We identified
antibody (IgG control) (Figure 5D). We further performed analo- 1621 promoter regions with significant occupancy by hnRNP-K
gous RIP experiments with formaldehyde crosslinked cells (FDR < 0.05) (Figure 6 and Table S3). Notably, these promoter
followed by stringent washing conditions (Ule et al., 2005) to regions exhibit a significant overlap with genes that were differ-
rule out potential nonspecific interactions. Consistent with entially expressed upon hnRNP-K knockdown (GSEA FDR <
a bona fide interaction, we observed a greater and very signifi- 0.001) (Figure S6E). Moreover, hnRNP-K localizes to a significant
cant enrichment of lincRNA-p21 in the hnRNP-K RIP relative to fraction (FDR < 0.001) of the genes corepressed by lincRNA-p21
the IgG control RIP with two hnRNP-K different antibodies and p53 (Figure 6C), suggesting that these are primary sites of
(Figure 5E). hnRNP-K regulation.
We further performed deletion-mapping experiments to deter- We next wanted to determine whether lincRNA-p21 plays
mine whether hnRNP-K interacts within a specific region of a role in hnRNP-K localization at promoters of p53-repressed
lincRNA-p21. To this end, we carried out RNA pulldown experi- genes. To this end, we determined whether siRNA-mediated
ments with truncated versions of lincRNA-p21 followed by knockdown of lincRNA-p21 affected the localization of hnRNP-K
western blot detection of bound hnRNP-K. These analyses after induction of p53. Specifically, we performed hnRNP-K
identified a 780 nt region at the 50 end of lincRNA-p21 required ChIP in dox-treated p53-restored p53LSL/LSL MEFs transfected
for the interaction with hnRNP-K (Figure 5F). Interestingly, RNA with either siRNAs targeting lincRNA-p21 or nontargeting con-
folding analyses of this region based on sequence conservation trol siRNAs. These experiments revealed that the depletion of
and compensatory changes across 14 mammalian species lincRNA-p21 causes a significant reduction in the association
(Hofacker, 2003) predict a highly stable 280 nt structure of of hnRNP-K at the promoter regions of genes that are normally
lincRNA-p21 with deep evolutionary conservation (Figures S5B repressed in a lincRNA-p21- and p53-dependent fashion, as
and S5C). Together, the RNA pulldown, native RIP, crosslinked determined by ChIP-qPCR (Figure 6D). Specifically, 12 of the
RIP, and deletion mapping results demonstrate a specific asso- 15 tested promoter regions exhibited loss of hnRNP-K enrich-
ciation between hnRNP-K and lincRNA-p21. ment, in two biological replicate experiments, upon depletion
We next sought to determine the functional relevance of the of lincRNA-p21. Thus, hnRNP-K is bound to the promoters of
interaction between lincRNA-p21 and hnRNP-K. To do so, we genes that are normally repressed in a p53- and lincRNA-p21-
monitored the ability of different truncated versions of lincRNA- dependent manner, and this localization requires lincRNA-p21.
p21 to induce cellular apoptosis when overexpressed in LKR Collectively, our results indicate that lincRNA-p21 is a direct
cells (Experimental Procedures). This revealed that the deletion p53 transcriptional target in response to DNA damage, acts to
of the 50 end of lincRNA-p21, which mediates the hnRNP-K inter- repress genes that are downregulated as part of the canonical
action, abolishes the ability of lincRNA-p21 to induce apoptosis p53 transcriptional response, is necessary for p53 dependent
(Figure 5G). Interestingly, the 780 nt fragment at the 50 end of apoptotic responses to DNA damage in our cell-based systems,
lincRNA-p21 alone does not induce apoptosis, indicating that and functions at least in part through interaction with hnRNP-K
this fragment is required but not sufficient for lincRNA-p21-medi- by modulating hnRNP-K localization.
ated cellular apoptosis.
We hypothesized that hnRNP-K is required for proper tran-
scriptional repression of target genes shared between p53 and DISCUSSION
lincRNA-p21. If so, knockdown of hnRNP-K should result in
derepression of these shared targets. We tested this hypothesis It is clear that mammalian genomes encode numerous large
by performing siRNA-mediated knockdown of hnRNP-K, noncoding RNAs (Carninci, 2008; Guttman et al., 2009; Mattick,
lincRNA-p21, and p53 in p53-restored p53LSL/LSL MEFs, treating 2009; Ponjavic et al., 2007). Here, we demonstrate that
the cells with dox and profiling the changes in gene expression numerous lincRNAs are key constituents in the p53-dependent
by microarray analysis. transcriptional pathway. Moreover, we observed that some of
Consistent with our previous data, we observed a strong over- these lincRNAs are bound by p53 in their promoter regions and
lap of 582 genes affected in the hnRNP-K, lincRNA-p21, and p53 sufficient to drive p53-dependent reporter activity that requires

416 Cell 142, 409–419, August 6, 2010 ª2010 Elsevier Inc.


A Figure 6. LincRNA-p21 and hnRNP-K Corepress Genes in the p53
lincRNA- Transcriptional Response
hnRNP-K
p21
582 (A) Many genes are coregulated by p53, lincRNA-p21, and hnRNP-K. Genes
affected by knockdown of lincRNA-p21, p53, or hnRNP-K in p53-restored-
p53 DNA-damaged p53LSL/LSL MEFs determined by microarray analysis (FDR <
0.05). Shades of red or blue represent expression values relative to global
median across experiments. Percentages of up- and downregulated genes
are indicated.
17% (B) Genes repressed by lincRNA-p21 are significantly enriched in genes
repressed by p53 and hnRNPK. GSEA comparing the genes upregulated on
knockdown of LincRNA-p21 and those upregulated upon knockdown of p53
582 genes

(left) or hnRNP-K (right). The black line represents the observed enrichment
score profile of genes in the lincRNA-p21 gene set to the p53 or hnRNP-K
83%
gene sets, respectively.
(C) hnRNP-K associates to promoters of genes corepressed by lincRNA-p21
and p53. Examples of promoters of genes repressed by p53 and lincRNA-
p21 (G2e3, Mtap4, Suv39h1, and Vcan) or repressed by lincRNA-p21
siRNA siRNA siRNA siRNA -80 but not p53 (Rb1) bound by hnRNP-K (blue) determined by ChIP-chip of
P< 10
B p53 lincRNA-p21 hnRNP-K control hnRNP-K in dox-trated p53-reconstituted p53LSL/LSL MEFs (FDR < 0.05).
-8 +8 Cdkn2a and Wt1 are negative controls (gray).
(D) hnRNP-K binding to lincRNA-p21 and p53 corepressed genes is depen-
Enrichment Score

Enrichment Score

p53 hnRNP-K
* dent on lincRNA-p21. Relative enrichment of hnRNP-K (ChIP-qPCR) in the
0.5 * 0.5 indicated promoter regions in p53-reconstituted p53LSL/LSL MEFs transfected
with siRNA lincRNA-p21 or siRNA control and dox-treated determined by
0.0 0.0
ChIP-qPCR (representative of two biological replicates shown ±STD).
* FDR<.001 *FDR<.001 (E) Proposed models for the function of licRNA-p21 in the p53 transcriptional
C response. Induction of p53 activates the transcription of lincRNA-p21 by
+3
binding to its promoter (upper left). LincRNA-p21 binds to hnRNP-K, and this
lincRNA-p21
hnRNP-K interaction imparts specificity to genes repressed by p53 induction (upper right).
/p53
bound 0
repressed
-2 See also Figure S6 and Table S3.
G2e3
P<0.001
Relative Binding Enrichment (hnRNP-K ChIP/IgG)

+3 +3

-2
0 0 the consensus p53-binding motif, suggesting that these
-2
Mtap4 Suv39h1 lincRNAs are bona fide p53 transcriptional targets.
+3 +3 Having discovered multiple lincRNAs in the p53 pathway, we
0 0 decided to focus on one such lincRNA in particular: lincRNA-
-2 -2
Rb1 Vcan p21. Intrigued by its properties (genomic location upstream of
+3 +3 p21, p53-dependent activation requiring the consensus p53
0 0 motif, which is bound by p53 and conserved p53-dependent
-2
-2
Cdkn2a Wt1
activation of this gene in both human and mouse cell-based
systems), we explored the functional roles of lincRNA-p21. Our
D studies revealed a role for lincRNA-p21 in a p53-dependent
115
45 39
apoptotic response after DNA damage.
14 35 20 31 We further observed that siRNA-mediated inhibition of lincRNA-
26
12 siRNA control
p21 affects the expression of hundreds of gene targets that are
Relative Fold Enrichment

siRNA lincRNA-p21 enriched for genes normally repressed by p53 in both the MEF
(hnRNPK ChIP/IgG)

10

8
and RAS cell-based systems. Strikingly, the vast majority of these
common target genes are derepressed upon inhibition of either
6
p53 or lincRNA-p21—suggesting that lincRNA-p21 functions as
4
a downstream repressor in the p53 transcriptional response.
2
We gained mechanistic clues into how lincRNA-p21 functions
0
to repress such a large subset of the p53 transcriptional
Vc

Jm

Zb

At

Lp

Pd

Zf

G a
xc

us

b1

sp

dk

ap
tp

p3
f2

p
an

tb

lim
jd

a4
r6

25

n2

dh
20

86
3

response by biochemical experiments that identified a specific


2

E interaction between lincRNA-p21 and hnRNP-K. This interaction


p53
POL II
is supported by RNA-pulldown, native RIP, crosslinked RIP, and
deletion-mapping experiments. Moreover, we identified a 780 nt
50 region of lincRNA-p21 that is required for hnRNP-K binding
hnRNP-K hnRNP-K and subsequent induction of apoptosis. Interestingly, this region
is much more highly conserved than the remainder of the tran-
? script. This suggests that patches of conservations, previously
determined to be unique to lincRNAs, (Guttman et al., 2009),
lincRNA-p21
may point to functional elements for binding interactions within

Cell 142, 409–419, August 6, 2010 ª2010 Elsevier Inc. 417


lincRNAs (as was also recently determined for Xist binding to EXPERIMENTAL PROCEDURES
PRC2) (Zhao et al., 2008).
Cell Lines and In Vivo Models
hnRNP-K is known to interact with other repressive complexes
KRAS lung tumor-derived cell lines were isolated from individual tumors (D.F.
such as the Histone H1.2 or members of the Polycomb-group and T.J., unpublished data). Isolation of matched p53+/+ and p53/ MEFs,
(PcG) (Kim et al., 2008; Denisenko and Bomsztyk, 1997). The p53LSL/LSL MEFs, lymphomas, and sarcomas, and p53 restoration were
physical interaction between lincRNA-p21 and hnRNP-K is likely done as described (Ventura et al., 2007). Primary WT MEFs and NIH/3T3
required for lincRNA-p21-mediated gene repression, as loss of MEF cells were purchased from ATCC. Transfection, infection, and treatment
hnRNP-K function results in the derepression of the same genes conditions are described in the Extended Experimental Procedures.
that are repressed by both p53 and lincRNA-p21. Importantly,
genome-wide ChIP-chip analysis revealed hnRNP-K binding at Promoter Reporter Assays
the promoters of these corepressed gene loci, suggestive of direct LincRNA promoters were cloned into pGL3-basic vector (Promega), and motif
regulation by hnRNP-K and lincRNA-p21. Moreover, we observed deletions were performed by mutagenesis. p53-reconstituted or control
p53LSL/LSL MEFs were transfected with 800 ng pGL3 and 30 ng TK-Renilla
a lincRNA-p21 dependent binding of hnRNP-K at several of these
plasmid per 24-well. Twenty-four hours later, cells were treated with 500 nM
corepressed promoter regions. While hnRNP-K has been previ- dox for 13 hr, and cell extracts were assayed for firefly and renilla luciferase
ously shown to activate one gene in the p53 pathway (Moumen activities (Promega E1910).
et al., 2005), our analyses suggest that it plays a much more
widespread role in repression. Together, these results implicate
LincRNA and Gene-Expression Profiling and Informatic Analyses
lincRNA-p21 as an important repressor in the p53 pathway, by RNA isolation, lincRNA expression profiling, and ChIP-chip analyses (Nimble-
interacting with and modulating the localization of hnRNP-K. gen arrays), as well as Affymetrix gene-expression profiling and analyses, were
Our results raise the possibility that many transcriptional performed as described (Guttman et al., 2009) (Extended Experimental Proce-
programs (beyond the p53-pathway) may involve inducing dures). Structure predictions were performed using the Vienna RNA package
protein factors that activate specific sets of downstream genes (Hofacker, 2003).
and lincRNAs that repress previously active sets of genes. The
notion of a noncoding RNA being involved in silencing-specific Antibodies
gene loci is consistent with our recent observation that many The following antibodies were used: anti-p53, Novocastra (NCL-p53-CM5p)
lincRNAs (including lincRNA-p21) bind to chromatin complexes (western blot) and Vector Labs (CM-5) (ChIP); anti-hnRNP-K, Santa Cruz
Biotechnology (sc-25373) (western blot) and Abcam (Ab70492 and Ab39975)
(such as PRC2) and are required to mediate repression at key
(ChIP and RIP); and control rabbit IgG Abcam (Ab37415-5) (RIP and ChIP-
gene loci (Khalil et al., 2009). Moreover, there are several exam- chip).
ples of lincRNAs involved in repression of known target genes—
including HOTAIR-dependent repression of HOXD genes (Rinn
Viability and Apoptosis Assays and Cell-Cycle Analysis
et al., 2007) and XIST, AIR, and KCNQ1OT1, involved in genomic MTT assays were performed with Cell Proliferation Kit I from Roche
imprinting and silencing of several genes in cis (Nagano et al., (11465007001). For apoptosis quantification, the Apoptosis Detection Kit I
2008; Pandey et al., 2008; Zhao et al., 2008). from BD Biosciences (cat#559763) and FACS (van Engeland et al., 1996)
The precise mechanism by which lincRNA-p21 contributes to were used. Cell-cycle analysis was performed as described (Brugarolas
repression at specific loci remains to be defined. Various possi- et al., 1995).
bilities include that (1) lincRNA-p21 might direct a protein
complex to specific loci by Crick-Watson base pairing; (2) Cloning, RNA Pulldown, Deletion Mapping, RIP, and ChIP
lincRNAs might act by forming DNA-DNA-RNA triple-helical 50 and 30 RACE cloning of lincRNA-p21 were performed from total RNA of dox-
structures, which do not require Crick-Watson base-pairing, treated MEFs with RLM-RACE Kit (Ambion). RNA pulldown and deletion
mapping were performed as described (Rinn et al., 2007) with 1 mg mESC
such as reported for a large noncoding RNA that forms a
nuclear extract and 50 pmol of biotinylated RNA. Mass spectrometry was per-
triple-helix upstream of the Dihydrofolate Reductase (DHFR) formed as described (Shevchenko et al., 1996). Native RIP was carried out as
promoter resulting in repression of DHFR (Martianov et al., described (Rinn et al., 2007). For crosslinked RIP, cells were crosslinked with
2007); or (3) lincRNAs might alter the binding specificity of 1% formaldehyde, antibody incubated overnight, recovered with protein G
DNA-binding proteins (such as hnRNP-K) to influence their target Dynabeads, and washed with RIPA buffer. After reverse crosslink, RNA was
preference (Figure 6D). Further experiments are needed to analyzed by qRT-PCR. p53 ChIP and hnRNP-K ChIP experiments were per-
formed as previously described (Rinn et al., 2007) (Extended Experimental
distinguish between these and other possibilities.
Procedures).
Aside from the general interest in gene regulation, we note that
lincRNA-p21 and several other lincRNAs function in an important
pathway for cancer. It is tempting to speculate that other RNA Interference and LincRNA-p21 Overexpression
siRNA transfections were done with 75 nM siRNA and Lipofectamine 2000
lincRNAs may also play key roles in numerous other tumor-
(Invitrogen). For overexpression, lincRNA-p21 or truncated forms were cloned
suppressor and oncogenic pathways, representing a hitherto into the pBABE vector. After transfection, cells were selected with 2 mg/ml
unknown paradigm in cellular transformation and metastasis. puromycin.
It will be important for future studies to determine whether
lincRNAs genes can serve as tumor suppressor genes or
ACCESSION NUMBERS
oncogenes.
In summary, lincRNAs may point to new mechanisms of gene The accession number for the full-length mouse lincRNA-p21 sequence
regulation, components in disease pathways and potential reported in this paper is HM210889 (bankit1350506). All primary data are avail-
targets for the development of therapies. able at the Gene Expression Omnibus (GSE21761).

418 Cell 142, 409–419, August 6, 2010 ª2010 Elsevier Inc.


SUPPLEMENTAL INFORMATION Martianov, I., Ramadass, A., Serra Barros, A., Chow, N., and Akoulitchev, A.
(2007). Repression of the human dihydrofolate reductase gene by a non-
Supplemental Information includes Extended Experimental Procedures, six coding interfering transcript. Nature 445, 666–670.
figures, and four tables and can be found with this article online at doi:10. Mattick, J.S. (2009). The genetic signatures of noncoding RNAs. PLoS Genet.
1016/j.cell.2010.06.040. 5, e1000459.
Mercer, T.R., Dinger, M.E., and Mattick, J.S. (2009). Long non-coding RNAs:
ACKNOWLEDGMENTS insights into functions. Nat. Rev. Genet. 10, 155–159.
Mikkelsen, T.S., Ku, M., Jaffe, D.B., Issac, B., Lieberman, E., Giannoukos, G.,
We would like to thank Loyal A. Goff (Massachusetts Institute of Technology Alvarez, P., Brockman, W., Kim, T.K., Koche, R.P., et al. (2007). Genome-wide
[MIT]) for bioinformatic support, Nadya Dimitrova (MIT) for input on the manu- maps of chromatin state in pluripotent and lineage-committed cells. Nature
script, David Garcia (MIT) for experimental assistance, and Sigrid Hart (Broad 448, 553–560.
Institute) for illustration support. J.L.R. is a Damon Runyon-Rachleff, Searle, Moumen, A., Masterson, P., O’Connor, M.J., and Jackson, S.P. (2005). hnRNP
and Smith Family Foundation Scholar. J.L.R. and A.R. are Richard Merkin K: an HDM2 target and transcriptional coactivator of p53 in response to DNA
Foundation Scholars. This work was supported by the National Institutes of damage. Cell 123, 1065–1078.
Health (NIH) Director’s New Innovator Award, Smith Family Foundation,
Nagano, T., Mitchell, J.A., Sanz, L.A., Pauler, F.M., Ferguson-Smith, A.C., Feil,
Damon Runyon Cancer Foundation, Searle Scholar Program, and NIH
R., and Fraser, P. (2008). The Air noncoding RNA epigenetically silences tran-
1R01CA119176-01.
scription by targeting G9a to chromatin. Science 322, 1717–1720.
Received: October 6, 2009 Pandey, R.R., Mondal, T., Mohammad, F., Enroth, S., Redrup, L., Komorowski,
Revised: April 6, 2010 J., Nagano, T., Mancini-Dinardo, D., and Kanduri, C. (2008). Kcnq1ot1 anti-
Accepted: June 3, 2010 sense noncoding RNA mediates lineage-specific transcriptional silencing
Published online: July 29, 2010 through chromatin-level regulation. Mol. Cell 32, 232–246.
Ponjavic, J., Ponting, C.P., and Lunter, G. (2007). Functionality or transcrip-
tional noise? Evidence for selection within long noncoding RNAs. Genome
REFERENCES
Res. 17, 556–565.
Brugarolas, J., Chandrasekaran, C., Gordon, J.I., Beach, D., Jacks, T., and Ponting, C.P., Oliver, P.L., and Reik, W. (2009). Evolution and functions of long
Hannon, G.J. (1995). Radiation-induced cell cycle arrest compromised by noncoding RNAs. Cell 136, 629–641.
p21 deficiency. Nature 377, 552–557. Riley, T., Sontag, E., Chen, P., and Levine, A. (2008). Transcriptional control of
Carninci, P. (2008). Non-coding RNA transcription: turning on neighbours. Nat. human p53-regulated genes. Nat. Rev. Mol. Cell Biol. 9, 402–412.
Cell Biol. 10, 1023–1024. Rinn, J.L., Kertesz, M., Wang, J.K., Squazzo, S.L., Xu, X., Brugmann, S.A.,
Denisenko, O.N., and Bomsztyk, K. (1997). The product of the murine homolog Goodnough, L.H., Helms, J.A., Farnham, P.J., Segal, E., and Chang, H.Y.
of the Drosophila extra sex combs gene displays transcriptional repressor (2007). Functional demarcation of active and silent chromatin domains in
activity. Mol. Cell. Biol. 17, 4707–4717. human HOX loci by noncoding RNAs. Cell 129, 1311–1323.
Schaefer, B.C. (1995). Revolutions in rapid amplification of cDNA ends: new
el-Deiry, W.S., Kern, S.E., Pietenpol, J.A., Kinzler, K.W., and Vogelstein, B.
strategies for polymerase chain reaction cloning of full-length cDNA ends.
(1992). Definition of a consensus binding site for p53. Nat. Genet. 1, 45–49.
Anal. Biochem. 227, 255–273.
Funk, W.D., Pak, D.T., Karas, R.H., Wright, W.E., and Shay, J.W. (1992). A tran-
Shevchenko, A., Wilm, M., Vorm, O., Jensen, O.N., Podtelejnikov, A.V., Neu-
scriptionally active DNA-binding site for human p53 protein complexes. Mol.
bauer, G., Shevchenko, A., Mortensen, P., and Mann, M. (1996). A strategy
Cell. Biol. 12, 2866–2871.
for identifying gel-separated proteins in sequence databases by MS alone.
Garber, M., Guttman, M., Clamp, M., Zody, M.C., Friedman, N., and Xie, X. Biochem. Soc. Trans. 24, 893–896.
(2009). Identifying novel constrained elements by exploiting biased substitu-
Subramanian, A., Tamayo, P., Mootha, V.K., Mukherjee, S., Ebert, B.L.,
tion patterns. Bioinformatics 25, i54–i62.
Gillette, M.A., Paulovich, A., Pomeroy, S.L., Golub, T.R., Lander, E.S., and
Guttman, M., Amit, I., Garber, M., French, C., Lin, M.F., Feldser, D., Huarte, M., Mesirov, J.P. (2005). Gene set enrichment analysis: a knowledge-based
Zuk, O., Carey, B.W., Cassady, J.P., et al. (2009). Chromatin signature reveals approach for interpreting genome-wide expression profiles. Proc. Natl.
over a thousand highly conserved large non-coding RNAs in mammals. Nature Acad. Sci. USA 102, 15545–15550.
458, 223–227. Ule, J., Jensen, K., Mele, A., and Darnell, R.B. (2005). CLIP: a method for iden-
Hofacker, I.L. (2003). Vienna RNA secondary structure server. Nucleic Acids tifying protein-RNA interaction sites in living cells. Methods 37, 376–386.
Res. 31, 3429–3431. van Engeland, M., Ramaekers, F.C., Schutte, B., and Reutelingsperger, C.P.
Khalil, A.M., Guttman, M., Huarte, M., Garber, M., Raj, A., Rivea Morales, D., (1996). A novel assay to measure loss of plasma membrane asymmetry during
Thomas, K., Presser, A., Bernstein, B.E., van Oudenaarden, A., et al. (2009). apoptosis of adherent cells in culture. Cytometry 24, 131–139.
Many human large intergenic noncoding RNAs associate with chromatin- Vazquez, A., Bond, E.E., Levine, A.J., and Bond, G.L. (2008). The genetics of
modifying complexes and affect gene expression. Proc. Natl. Acad. Sci. the p53 pathway, apoptosis and cancer therapy. Nat. Rev. Drug Discov. 7,
USA 106, 11667–11672. 979–987.
Kim, K., Choi, J., Heo, K., Kim, H., Levens, D., Kohno, K., Johnson, E.M., Ventura, A., Kirsch, D.G., McLaughlin, M.E., Tuveson, D.A., Grimm, J., Lintault,
Brock, H.W., and An, W. (2008). Isolation and characterization of a novel L., Newman, J., Reczek, E.E., Weissleder, R., and Jacks, T. (2007). Restoration
H1.2 complex that acts as a repressor of p53-mediated transcription. of p53 function leads to tumour regression in vivo. Nature 445, 661–665.
J. Biol. Chem. 283, 9113–9126.
Wei, C.L., Wu, Q., Vega, V.B., Chiu, K.P., Ng, P., Zhang, T., Shahab, A., Yong,
Kuerbitz, S.J., Plunkett, B.S., Walsh, W.V., and Kastan, M.B. (1992). Wild-type H.C., Fu, Y., Weng, Z., et al. (2006). A global map of p53 transcription-factor
p53 is a cell cycle checkpoint determinant following irradiation. Proc. Natl. binding sites in the human genome. Cell 124, 207–219.
Acad. Sci. USA 89, 7491–7495. Zhao, J., Sun, B.K., Erwin, J.A., Song, J.J., and Lee, J.T. (2008). Polycomb
Levine, A.J., Hu, W., and Feng, Z. (2006). The P53 pathway: what questions proteins targeted by a short repeat RNA to the mouse X chromosome. Science
remain to be explored? Cell Death Differ. 13, 1027–1036. 322, 750–756.

Cell 142, 409–419, August 6, 2010 ª2010 Elsevier Inc. 419


A Minimal Midzone Protein Module
Controls Formation and Length of
Antiparallel Microtubule Overlaps
Peter Bieling,1,2 Ivo A. Telley,1 and Thomas Surrey1,*
1CellBiology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
2Present address: Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco,
CA 94158, USA
*Correspondence: surrey@embl.de
DOI 10.1016/j.cell.2010.06.033

SUMMARY 2008). The midzone size in metazoans is typically 2–3 mm in


length (Mastronarde et al., 1993). Several proteins essential for
During cell division, microtubules are arranged in a spindle elongation, faithful chromosome segregation, and pro-
large bipolar structure, the mitotic spindle, to segre- per cytokinesis relocalize selectively to the central spindle as a
gate the duplicated chromosomes. Antiparallel mic- consequence of changes in their phosphorylation patterns in
rotubule overlaps in the spindle center are essential anaphase: PRC1 (protein required for cytokinesis 1), kinesin-4,
for establishing bipolarity and maintaining spindle the centralspindlin complex (containing a kinesin-6), the chro-
mosome passenger complex, and several other molecular
stability throughout mitosis. In anaphase, this anti-
motors, nonmotor microtubule-bundling proteins, and regula-
parallel microtubule array is tightly bundled forming tors (Glotzer, 2009). How the organization of the midzone is de-
the midzone, which serves as a hub for the recruit- termined by the midzone proteins is, however, not understood.
ment of proteins essential for late mitotic events. The conserved vertebrate-bundling protein PRC1 (Jiang et al.,
The molecular mechanism of midzone formation 1998; Kurasawa et al., 2004; Mollinari et al., 2002; Zhu and Jiang,
and the control of its size are not understood. Using 2005) and its orthologs (Ase1 in yeasts [Loiodice et al., 2005;
an in vitro reconstitution approach, we show here Schuyler et al., 2003], SPD-1 in C. elegans [Verbrugghe and
that PRC1 autonomously bundles antiparallel micro- White, 2004], Feo in D. melanogaster [Verni et al., 2004], and
tubules and recruits Xklp1, a kinesin-4, selectively to MAP65 in plants [Muller et al., 2004]) have emerged as key
overlapping antiparallel microtubules. The proces- players for midzone formation. Ase1 in budding yeast has been
sive motor Xklp1 controls overlap size by overlap proposed to be the core component of the spindle midzone.
Genetic experiments have demonstrated that localization of all
length-dependent microtubule growth inhibition.
midzone proteins depends on Ase1, whose localization is inde-
Our results mechanistically explain how the two pendent of most other midzone proteins (Khmelinskii et al.,
conserved, essential midzone proteins PRC1 and 2007). Interestingly, fission yeast Ase1 has been shown to bind
Xklp1 cooperate to constitute a minimal protein with some preference to antiparallel versus parallel microtubule
module capable of dynamically organizing the core bundles in vitro (Janson et al., 2007; Kapitein et al., 2008), indi-
structure of the central anaphase spindle. cating that recognition of antiparallel microtubules is an autono-
mous property of Ase1 in yeast.
INTRODUCTION In contrast to yeast, PRC1 in higher eukaryotes interacts with
motor proteins of the kinesin-4 type. These chromokinesin
The various architectures adopted by the microtubule cytoskel- motors relocalize to the central spindle in anaphase (Kurasawa
eton are determined by distinct combinations of mechanochem- et al., 2004; Kwon et al., 2004; Lee and Kim, 2004; Powers
ical protein activities. How global properties of a complex et al., 2004; Vernos et al., 1995; Zhu and Jiang, 2005). Correct
cytoskeletal system, for instance characteristic shape and size, localization of both PRC1 and kinesin-4 to the midzone depend
emerge from the local biochemical properties of the constituting on each other (Kurasawa et al., 2004; Zhu and Jiang, 2005; Zhu
molecules is an open question. During cell division, the microtu- et al., 2005). However, whether kinesin-4 transports PRC1 to the
bule cytoskeleton forms the mitotic spindle to segregate the midzone (Zhu and Jiang, 2005) or PRC1 selectively binds the
duplicated set of chromosomes (Walczak and Heald, 2008). central region of the anaphase spindle thereby recruiting kine-
Spindles consist of two antiparallel arrays of microtubules that sin-4 (Kurasawa et al., 2004) is presently unclear. Importantly,
overlap in the spindle center, an organization that ensures it is unknown how the size of the midzone is determined.
spindle function. At anaphase onset, the central spindle reorga- Whereas absence of PRC1 results in complete loss of midzone
nizes to form the midzone, a dense specialized array of antipar- microtubule bundles in mammalian cells (Kurasawa et al.,
allel microtubules (Glotzer, 2009; Khmelinskii and Schiebel, 2004; Mollinari et al., 2002; Zhu and Jiang, 2005), absence of

420 Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc.


kinesin-4 leads to increased anaphase spindle length (Kurasawa that the growth velocity of two microtubules of an antiparallel
et al., 2004; Zhu and Jiang, 2005). Interestingly, a fragment of overlap is similar to that of individual microtubules and therefore
Xenopus kinesin-4 has been shown to decrease microtubule not significantly affected by the strong accumulation of PRC1.
growth and shrinkage rates in vitro (Bringmann et al., 2004), an To investigate the mechanism by which vertebrate PRC1
activity different from other regulators of microtubule dynamics specifically recognizes antiparallel overlaps, we quantified its
(Howard and Hyman, 2007). Hence, kinesin-4 might contribute affinity and selectivity of binding to distinct microtubule configu-
to midzone formation by regulating midzone microtubule rations. We measured the average fluorescence intensity of
dynamics. The minimal set of protein activities required for PRC1–Alexa 647 in antiparallel overlaps as a function of the
specific organizational features of the spindle midzone and the PRC1–Alexa 647 concentration (Figure 1E, Figure S1A). PRC1
logic behind the combination of the required activities are, binding to antiparallel overlaps was strong and positively coop-
however, unknown. erative with a microscopic dissociation constant of 7.6 ± 0.4 nM
We have developed an in vitro reconstitution approach to (mean ± standard error of the mean [SEM]) and a Hill coefficient
investigate the combinatorial action of purified full-length of 1.9 ± 0.1 (Figure 1E). Under standard imaging conditions, we
Xenopus kinesin-4 (Xklp1) and Xenopus PRC1 on microtubule did not detect any accumulation of PRC1 on individual microtu-
organization and dynamics. We show that Xklp1 is recruited to bules or between parallel microtubules even at the highest PRC1
antiparallel microtubule overlaps by PRC1, which autonomously concentration used (31.5 nM) (Figure S1B). We estimate that
discriminates between parallel and antiparallel microtubules with PRC1 binds at least 28 times more strongly to antiparallel
high selectivity. Xklp1 ensures the maintenance of microtubule microtubule pairs than to individual or parallel microtubules
overlap size by selectively inhibiting the growth of overlapping (Extended Experimental Procedures), indicating considerably
microtubules in a manner that is dependent on overlap length. higher selectivity for antiparallel microtubules than previously
Our results demonstrate that only two proteins are necessary reported for Ase1 from fission yeast (Janson et al., 2007).
and sufficient for the selective formation and for adaptable length To investigate the mechanism of this high binding selectivity,
control of antiparallel microtubule overlaps in vitro, as required we imaged single PRC1 molecules at higher temporal resolution.
for the formation of the midzone in vivo. Many binding events of individual PRC1–Alexa 647 molecules
were detected in antiparallel overlaps (Figure 1F, right), whereas
RESULTS hardly any events were observed in parallel microtubule pairs
(Figure 1F, left). Individual PRC1 molecules were not statically
PRC1 Selectively Binds to Antiparallel Microtubule bound but diffused in antiparallel overlaps (Figures S1C and
Overlaps with Nanomolar Affinity S1D). Despite the high affinity for antiparallel microtubules,
We developed a fluorescence microscopy assay to visualize turnover of PRC1 in antiparallel overlaps was fast with a single-
dynamic microtubule encounters in vitro. We attached short, molecule half-life of around 1 s (Figure 1G, Figure S1E).
Alexa 568- and biotin-labeled, stabilized microtubule seeds Therefore, selective binding to antiparallel microtubules is an
under flow to glass slides, which were functionalized with a intrinsic property of the individual PRC1 dimer and it is very
NeutrAvidin-biotin-polyethylene glycol layer. The majority of dynamic. This appears to distinguish PRC1 from Ase1 whose
the seeds aligned, and microtubules growing from these seeds moderate selectivity for antiparallel microtubule binding was
in the presence of Alexa 568–tubulin occasionally encountered suggested to be a consequence of the formation of slowly
each other in a plus end-to-plus end configuration (Figure 1A). dissociating oligomers (Kapitein et al., 2008).
Multicolor time-lapse TIRF microscopy revealed that antiparallel
microtubules connected in the presence of 5 nM purified PRC1– Reconstitution of a Minimal Midzone with Stable
Alexa 647 as soon as their plus ends encountered, forming an Antiparallel Microtubule Overlaps In Vitro
antiparallel microtubule overlap (Figure 1B, Movie S1 available We next tested the additional effect of Xenopus kinesin-4 Xklp1
online). Interestingly, PRC1–Alexa 647 strongly accumulated in on microtubule overlap formation. In the presence of both 5 nM
the growing antiparallel overlap region under conditions at which purified PRC1–Alexa 647 and 15 nM purified Xklp1–GFP (Fig-
it did not localize significantly to individual microtubules or to ure S2), antiparallel microtubules connected to each other as
parallel microtubule pairs (Figure 1C, Movie S1). This demon- soon as their plus ends encountered, forming an antiparallel
strates that vertebrate PRC1 does not require an additional microtubule overlap that remarkably reached a constant size
factor to selectively target antiparallel microtubule configura- after awhile (Figure 2A, Movie S2, and Movie S3). PRC1–Alexa
tions. Autonomous binding to antiparallel microtubules, there- 647 and also Xklp1–GFP were observed to strongly accumulate
fore, appears to be conserved between PRC1 orthologs from selectively in antiparallel but not parallel overlap regions (Fig-
vertebrates and yeast (Janson et al., 2007). ure 2B, Movie S2). Xklp1 had only a minor positive effect on
The length of antiparallel microtubule overlaps increased the amount of overlap-associated PRC1 (Figure 2C). In contrast,
linearly with time after the microtubule encounter (Figure 1D, no Xklp1 was observed in the absence of PRC1 in antiparallel
top) indicating constant average overlap growth velocity. Over- microtubule overlaps (Figures 2D–2F). Microtubules often failed
lap length as measured from plus end to plus end of the two to form a tight bundle in the presence of Xklp1 only (Movie S4),
antiparallel microtubules increased approximately twice as fast a behavior never observed when PRC1 was present.
(4.9 ± 0.9 mm/min, mean ± standard deviation [SD]) as the length These TIRF microscopy data indicate that Xklp1 interacts
of single microtubules that were not part of an antiparallel micro- directly with PRC1, which we confirmed by pull-down experi-
tubule pair (2.3 ± 0.3 mm/min) (Figure 1D, bottom), demonstrating ments (Figure 2G, lane 2). Consequently, an N-terminal motor

Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc. 421


PRC1 Figure 1. PRC1 Preferentially Binds to and
A evanescent field bundling protein/complex B
microtubules Diffuses in Antiparallel Microtubule Over-
Alexa568-Tubulin
microtubule seed 0:00 laps
(A) Scheme of the experimental setup. Dynamic
0:23
microtubules were grown in the presence of free
0:47 Alexa 568-labeled tubulin (light blue) and fluores-
cently labeled midzone proteins (red) from stabi-
1:10 lized microtubule seeds (dark blue) attached to a
anti-parallel overlap PEG-passivated glass surface by means of
1:33
biotin–NeutrAvidin (yellow and green) links. Micro-
1:57 tubules occasionally encountering each other in a
plus end-to-plus end configuration forming an
antiparallel microtubule overlap and fluorescently
C PRC1 D
scheme microtubules PRC1 microtubules labeled midzone proteins were observed by multi-
antiparallel overlap color time-lapse TIRF microscopy.
15 free microtubules
(B) Time sequence of overlaid TIRF microscopy

length [µm]
10
images of PRC1–Alexa 647 (red, 5 nM) and a
dynamic Alexa 568–microtubule pair (blue) form-
5 ing an antiparallel overlap taken at the indicated
0
times in min:s. The time-lapse was recorded at 1
0 1 2 3 frame per 4.67 s. Scale bar, 10 mm.
time [min]

growth velocity [µm/min]


(C) TIRF images showing several microtubules
6 growing from immobilized microtubule seeds, re-
corded 4–9 min after start of the experiment
4
(top), and a kymograph (time-space plot) of a
2 selected microtubule pair (bottom) in the presence
0 of PRC1–Alexa 647. The kymograph begins
0 1 2
time [min]
3 1 min after start of the experiment. Left: sche-
matic illustration of the microtubule configurations
E F parallel overlap antiparallel overlap visible in the image and kymograph: microtubule
fluor. signal in AP overlap

scheme
segments (gray, plus ends labeled with ‘‘+’’)
2
growing from seeds (white) form antiparallel (red)
microtubule image
[x104 au]

before time-lapse or parallel (green) overlaps. Parallel overlaps


form from parallel seed bundles. Right: Overlaid
1
and single-channel TIRF microscopy images and
kymographs for PRC1–Alexa 647 and Alexa 568
0 microtubules as indicated. Concentrations and
0 10 20 30 frame rate as in (B). Horizontal scale bars, 10 mm.
PRC1 concentration [nM]
G 100 kymograph of
PRC1 time-lapse
Vertical scale bar, 1 min.
(D) Average length (top) and average instantaneous
growth velocity (bottom) of antiparallel microtubule
overlaps (red, n = 7) and of individual microtubules
1 − CDF

10-1 not part of a pair from the same experiment (blue,


n = 14) forming in the presence of PRC1–Alexa
647 as a function of time. For antiparallel microtu-
microtubule image bule overlaps t = 0 is the moment of the plus end-
10-2
0 2 4 6 8 10 12 after time-lapse to-plus end encounter. Error bars are standard
dwell time [s] PRC1 (single molecules)
deviation (SD). Concentrations are as in (B).
microtubules
(E) Average fluorescence signal of PRC1–Alexa
647 bound to antiparallel microtubule overlaps
plotted against the total PRC1–Alexa 647 concentration. Data (black points) were fitted either using the Hill equation (black, straight line) or a hyperbolic function
(red, dotted line). At least a total of 100 mm of antiparallel overlap were evaluated per condition. Error bars are standard error of the mean (SEM).
(F) Single-molecule imaging of PRC1–Alexa 647 in a parallel (left) and an antiparallel (right) microtubule pair. Top: schematic representation of the pairs. Below:
kymographs, positioned between TIRF microscopy images of microtubule overlaps before (top) and after (bottom) fast recording of individual PRC1–Alexa 647
molecules, showing the behavior of individual PRC1–Alexa 647 molecules (green). The PRC1–Alexa 647 concentration was 50 pM. Frame rate was 1 frame per
100 ms. Horizontal scale bar, 10 mm. Vertical scale bar, 5 s.
(G) Dwell time distribution plotted as 1  cumulative distribution function (CDF) of individual PRC1–Alexa 647 molecules in antiparallel overlaps (black, n = 687).
The red dashed curve is a biexponential fit with characteristic times t1 = 0.91 ± 0.02 s and t2 = 4.47 ± 0.24 s (mean ± 95% confidence interval [CI]), and relative
amplitudes 87% t1 and 13% t2. The black dashed curve is a monoexponential fit. The mean bleaching time for Alexa 647 was 32.0 ± 4.5 s. Consequently t1 is
practically unaffected by bleaching and the corrected t2 is 5.2 s.
See also Figure S1 and Movie S1.

fragment, Xklp506–GFP, that does not interact with PRC1 (Fig- interaction between Xklp1 and PRC1 is required for PRC1-depen-
ure 2G, lane 3) failed to accumulate in antiparallel microtubule dent recruitment of Xklp1 to antiparallel microtubule overlaps.
overlaps despite the presence of PRC1–Alexa 647, as observed The most striking observation in the presence of both midzone
by TIRF microscopy (Figure 2H, right). This shows that a direct proteins was, however, that the growth of microtubule plus ends

422 Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc.


being part of an antiparallel overlap slowed down until it ceased To gain a better understanding of the effect of Xklp1 on the
completely in all observed cases (n > 100) (Figure 2A, kymograph microtubule catastrophe frequency, we measured the microtu-
in 2B, 2H, left, Movie S2, Movie S3). The growth of overlap bule growth velocity at different tubulin concentrations with
microtubules gradually stopped within about 1 min after the and without 300 nM Xklp1 (Figure 3E). This allowed us to deter-
end-to-end encounter (Figure 2I, green). In contrast, individual mine the association and dissociation rate constants of guano-
microtubules not engaged in overlaps in the same experiment sine-50 -triphosphate (GTP)-tubulin at the growing plus end
continued growing with constant velocity of 2.1 ± 0.3 (mean ± (Drechsel et al., 1992; Walker et al., 1988). We found that
SD) mm/min (Figure 2I, blue). Inhibition of antiparallel microtubule 300 nM Xklp1 reduced the on-rate of GTP-tubulin by a factor
overlap growth was a consequence of the simultaneous pres- of about two and the off-rate by a factor of around three (Fig-
ence of PRC1 and full-length Xklp1. The presence of neither ure 3F). This shows that Xklp1 has the unique property of making
PRC1 alone (Figures 1B–1D) nor Xklp1 alone (Figure 2F) had a microtubules less dynamic by reducing the overall turnover of
significant effect on microtubule growth in antiparallel overlaps. GTP-tubulin at growing microtubule ends, distinguishing it
Hence, stable antiparallel overlaps with a constant size of several from other known mitotic regulators of microtubule dynamics
micrometers formed only in the presence of both midzone pro- (Brouhard et al., 2008; Helenius et al., 2006; Varga et al., 2006).
teins (Figures 2A, 2B, and 2I; Movie S3). Such antiparallel micro-
tubule overlaps maintained a constant size over long periods of Dynamic Control of Antiparallel Microtubule Overlap
time despite the complete inhibition of microtubule growth. This Length
is remarkable, given that slowdown of microtubule growth To investigate how the inhibitory effect of Xklp1 on tubulin turn-
usually results in frequent transitions to microtubule depolymer- over affects the length of antiparallel microtubule overlaps in the
ization (catastrophes) (Arnal et al., 2004; Janson et al., 2003; presence of PRC1, we varied the Xklp1:PRC1 ratio in antiparallel
Walker et al., 1988). microtubule encounter experiments, leaving the PRC1 concen-
Our results show that a ‘‘minimal midzone’’ can be reconsti- tration constant (Figure 4A, Movie S5). At Xklp1:PRC1 ratios lower
tuted in vitro with dynamic microtubules and two proteins only: than 1, microtubule plus-end growth persisted over long times
PRC1 is necessary and sufficient to recognize and bundle (>10 min) after formation of an antiparallel overlap, albeit at greatly
antiparallel microtubules. Xklp1 alone does not bind to antipar- reduced speeds as compared to microtubules that were not part
allel overlaps but is instead recruited by overlap-associated of an overlap (Movie S5). At Xklp1:PRC1 ratios R1, microtubule
PRC1. This recruitment results in local inhibition of microtubule plus ends within the antiparallel overlap stopped growing soon
growth without causing microtubule depolymerization, leading after their initial encounter leading to stable antiparallel overlaps
to antiparallel microtubule overlaps of constant size. with a distinct characteristic steady-state length for each
Xklp1:PRC1 ratio (Figure 4B). The average steady-state overlap
Xklp1 Inhibits Turnover of Tubulin at the Growing length decreased with increasing Xklp1 concentration, ranging
Microtubule End from 4.2 mm to 1.2 mm for ratios from 1 to 20, respectively (Fig-
To better understand how Xklp1 affects microtubule dynamics, ure 4B). Thus the concentration of Xklp1 determines the steady-
we measured the effect of varying concentrations of Xklp1 state length of the antiparallel microtubule overlap.
alone on the dynamics of individual microtubules. Similar to an Next, we investigated whether the steady-state overlap
N-terminal fragment of Xklp1 (Bringmann et al., 2004), full-length lengths could reversibly respond to changes in the Xklp1 con-
Xklp1 alone reduced the growth velocity of microtubule plus centration (Figure 4C). We preformed short overlaps at high
ends in a concentration-dependent manner (Figures 3A and 3B). Xklp1:PRC1 ratios, followed by reduction of the Xklp1 concen-
This effect could only be observed at Xklp1 concentrations of tration, keeping the PRC1 and tubulin concentrations constant.
several hundred nanomolar, well above the Xklp1 concentra- Strikingly, we observed that microtubules of short preformed
tions in the lower nanomolar range that lead to inhibition of overlaps started to grow again until they had reached the length
microtubule growth in antiparallel overlaps in the presence of characteristic for the new Xklp1:PRC1 ratio (Figures 4C–4E,
PRC1. Interestingly, the catastrophe frequency remained very Movie S6). These results demonstrate that the Xklp1:PRC1 ratio
low at increasing Xklp1 concentrations (Figure 3C), despite the can dynamically control the steady-state overlap length.
strong reduction in microtubule growth velocity, which is ex- Why do higher Xklp1:PRC1 ratios result in shorter antiparallel
pected to lead to more frequent catastrophes (Arnal et al., overlaps? The major contribution of Xklp1 in determining overlap
2004; Janson et al., 2003; Walker et al., 1988). The effect of length could be either its inhibitory effect on microtubule growth
Xklp1 on the catastrophe frequency is more evident when or, alternatively, a consequence of Xklp1 sliding apart antiparallel
comparing microtubules growing with similar velocities (Fig- microtubules, similar to kinesin-5 (Kapitein et al., 2005). Indeed,
ure 3D). The catastrophe frequencies of microtubules in the we observed that microtubules engaged in antiparallel overlaps
presence of Xklp1, plotted as a function of their growth velocities frequently buckled over time (Figure S3A), indicative of plus-
(Figure 3D, red), are significantly lower than those of microtu- end-directed microtubule sliding. The average sliding length
bules growing at similar velocities in the absence of Xklp1 at (Extended Experimental Procedures) was, however, in all cases
reduced tubulin concentrations (Figure 3D, blue). This shows short as compared to the overlap length at steady state (Figure 4B).
that Xklp1 inhibits catastrophes, explaining why local inhibition A plot of the individual sliding lengths of all microtubule pairs
of microtubule growth by PRC1-recruited Xklp1 in antiparallel against their individual overlap length also did not reveal any corre-
microtubule pairs (Figures 2A, 2B, and 2I) does not result in lation (Figure S3B). Therefore, antiparallel microtubule sliding
destabilization of the overlap. does not significantly contribute here to overlap length control.

Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc. 423


Xklp1 + PRC1
A microtubules C I
20

PRC1 signal in AP overlap


0:00

growth velocity [µm/min]


antiparallel overlap
7.5 free microtubules
0:23 15 6

[x103au]

length [µm]
0:47 10 5 4
1:10
5 2.5 2
1:33
0 0 0
1:57 - Xklp1 + Xklp1 0 1 2 3 0 1 2 3
time [min] time [min]

Xklp1 + PRC1
B scheme microtubules PRC1 Xklp1 microtubules

D Xklp1 E G Beads Input


microtubules M 1 2 3 M 1 2 3 M
250 kDa
8
Xklp1 signal in AP overlap

0:00 Xklp1-GFP
150 kDa

0:23 6
100 kDa
[x103au]

PRC1-SNAP
0:47 Xklp1506-GFP
4 75 kDa

1:10
2
tubulin 50 kDa
1:33
0 37 kDa
1:57 +PRC1 - PRC1

F Xklp1 H Xklp1 + PRC1 Xklp1506+ PRC1


scheme microtubules Xklp1 microtubules microtubules microtubules

Figure 2. A Minimal Protein System Forms Stable Antiparallel Overlaps In Vitro


(A) Time sequence of overlaid TIRF microscopy images of PRC1–Alexa 647 (red, 5 nM) and Xklp1–GFP (green, 15 nM) in a dynamic Alexa 568-labeled microtubule
pair (blue) forming an antiparallel overlap at the indicated times in min:s.
(B) TIRF images, recorded 4–9 min after the start of the experiment, showing several microtubules growing from immobilized microtubule seeds (top) and a kymo-
graph of a selected microtubule pair (bottom). Left: schematic illustration of the microtubule configurations. Color coding as in Figure 1C. Right: overlaid and
single-channel TIRF microscopy images and kymographs for PRC1–Alexa 647, Xklp1–GFP, and Alexa 568 microtubules as indicated. Concentrations as in (A).
(C) Average fluorescence intensity signal of PRC1–Alexa 647 bound to antiparallel microtubule overlaps in the presence (n = 45 overlaps) or absence (n = 18
overlaps) of Xklp1–GFP, obtained from intensity line scans. Error bars are SEM.
(D) Time sequence of overlaid TIRF images of 15 nM Xklp1–GFP (green) and a dynamic Alexa 568-microtubule pair (blue) forming an antiparallel overlap taken at
the indicated times in min:s.

424 Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc.


A Figure 3. Xklp1 Inhibits Turnover of Tubulin at the
Growing Microtubule End
(A) Representative kymographs of Alexa 568-labeled
microtubules growing in the absence or presence of
Xklp1–GFP (not shown) at the indicated concentrations.
Concentration of soluble tubulin is 17.5 mM. The frame
rate was 1 frame per 3 s. Horizontal scale bar, 10 mm.
Vertical scale bar, 1 min.
(B) Average microtubule growth velocity and (C) catas-
trophe frequency plotted against the total Xklp1–GFP
concentration. Sixty microtubules from two independent
experiments were analyzed per condition.
0 nM Xklp1 300 nM Xklp1 600 nM Xklp1
(D) Catastrophe frequency as a function of microtubule
growth velocity. Red data points are from experiments
B C D with 17.5 mM tubulin and varying Xklp1–GFP concentra-
3 3 tions, as shown in (B) and (C). Blue data points are from
growth velocity [µm/min]

-Xklp1
catastrophe frequency
catastrophe frequency

10 +Xklp1 experiments with varying tubulin concentrations (7, 12,


[10-2 events/min]
[10-2 events/min]

2 2 and 17.5 mM) in the absence of Xklp1–GFP.


(E) Average growth rate in the absence (blue circles) or in
5 the presence of 300 nM Xklp1–GFP (red circles) as a func-
1 1
tion of total tubulin concentration. A multiple linear regres-
sion model with a categorical factor g (+Xklp1: g = 0,
0 0 0 Xklp1: g = 1) revealed that both the abscissa and the
0 200 400 600 0 200 400 600 0 1 2 3
Xklp1 concentration [nM] Xklp1 concentration [nM] growth velocity [µm/min] slope were significantly different (p < 0.001, see Experi-
mental Procedures). The resulting regression lines repre-
sent vg = kon c – koff with the net assembly rate vg in tubulin
E F
dimers per second and the time constants for association
150

20

-Xklp1 kon koff


+Xklp1 [µM-1 s-1] [ s-1] kon in s1mM1 and dissociation koff in s1 of GTP-tubulin
10
growth rate [dimers/s]

4.03 ± 0.04 9.60 ± 0.85 at the growing microtubule end. Inset: regression lines
0

- Xklp1
100

[3.951 ; 4.101] [8.626 ; 10.571] and their 95% confidence intervals (dashed) at the origin,
-10

0 4 8
1.86 ± 0.02 3.08 ± 0.49 showing that not only kon (slope) but also koff (intersection
+ 300 nM Xklp1
[1.820 ; 1.906] [2.125 ; 4.027] with abscissa) is different for the two conditions. 185 to
50

210 microtubules from two independent experiments were


analyzed for each tubulin concentration per condition.
(F) Values for kon and koff (mean ± SEM, with 95% confi-
0

dence in brackets) as obtained from fits in (E).


0 5 10 15 20 25 30 35
tubulin concentration [µM]
Error bars in (B)–(E) are SD.

Xklp1 Is a Processive Motor estingly, accumulation of Xklp1 at these free microtubule ends
To elucidate the mechanism by which Xklp1 sets the size of also resulted in inhibition of microtubule growth (Movie S7).
antiparallel microtubule overlaps, we inspected the behavior This suggests that (1) it is the plus-end-associated fraction
of the motor more closely. Occasionally, antiparallel overlaps of Xklp1 that inhibits microtubule growth and (2) Xklp1 can
formed by lateral encounters between one microtubule end target the plus end by means of processive motility over con-
and one microtubule segment distant from the growing end siderable distances. This is surprising, given that an N-terminal
(Figure 5A). In such cases, Xklp1–GFP loaded onto the overlap fragment of Xklp1 has been proposed to be at most weakly
region by PRC1 also accumulated at the plus end of the processive based on enzymatic activity assays (Bringmann
microtubule extending beyond the overlap (Figure 5A). Inter- et al., 2004).

(E) Average fluorescence intensity signal of Xklp1–GFP bound to the antiparallel microtubule overlap in the presence (n = 42 overlaps) or absence of PRC1–Alexa
647, as obtained from intensity line scans. Error bars are SEM.
(F) Kymograph of the microtubule pair shown in (D). Color code as in (B).
(G) Pull-down of Xklp1 by PRC1–SNAP immobilized on beads. Coomassie-stained SDS-gel shows fractions bound to the beads after incubation with different
input solutions. Lanes 1: negative control with beads lacking PRC1–SNAP incubated with a mixture of 2 mM full-length Xklp1–GFP, 2 mM truncated Xklp1506–GFP,
and 5 mM soluble tubulin. Only weak nonspecific binding of the two Xklp1 proteins is seen. Lanes 2 and 3: beads with immobilized PRC1–SNAP incubated with
either 2 mM full-length Xklp1–GFP (lanes 2) or truncated 2 mM Xklp1506–GFP (lanes 3) in the presence of 5 mM soluble tubulin.
(H) Representative kymographs of antiparallel microtubule overlaps (blue) in the presence of PRC1–Alexa 647 (red, 5 nM) and either full-length Xklp1–GFP
(green, 15 nM) (left) or truncated Xklp1506–GFP (green, 15 nM) (right).
(I) Average length (left) and average instantaneous growth velocity (right) of antiparallel overlaps (green, n = 9) and individual microtubules that are not part of a pair
from the same experiment (blue, n = 18) formed in the presence of PRC1 and full-length Xklp1 as a function of time (conditions as in A and B). For comparison, the
length of microtubule overlaps formed in the presence of only PRC1 (from Figure 1D) is shown (red dashed line). t = 0 is the moment of plus end-to-plus end
encounter. Error bars are SD. Concentrations are as in (A). Horizontal scale bars are 10 mm. Vertical scale bars are 1 min. The frame rate for all time-lapse movies
was 1 frame per 4.67 s. Kymographs start 1 min after the start of the experiment.
See also Figure S2 and Movie S2, Movie S3, and Movie S4.

Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc. 425


A B Figure 4. Dynamic Control of Antiparallel Overlap
Xklp1 PRC1 microtubles
Length
overlap length
20 : 1 sliding length (A) Representative triple-channel TIRF microscopy images
Xklp1 : PRC1 of PRC1–Alexa 647 (red, 5 nM) and Xklp1–GFP (green,
4

length [µm]
100, 25, and 5 nM) at PRC1:Xklp1 ratios as indicated in
a dynamic Alexa 568–microtubule pair (blue) forming an
5:1
antiparallel overlap. Images were recorded 7–12 min after
Xklp1 : PRC1
2 start of the experiment. Scale bar, 10 mm.
(B) Average overlap length (red) or average sliding length
1:1 (blue) as a function of the Xklp1:PRC1 ratio. Error bars
Xklp1 : PRC1
0
are SD. Overlap lengths and sliding lengths were
1 2 3 5 10 20 measured for at least 15 overlaps from at least three
Xklp1:PRC1 ratio experiments per condition after steady state had been
reached. Yellow shading indicates the regime where no
steady-state length was established.
imaging
dilution

C SS 1 overlap growth SS 2 D (C) Reversibility experiment: Antiparallel microtubule over-


Xklp1 PRC1 microtubles laps were allowed to form at 100 nM Xklp1–GFP and 5 nM
PRC1–Alexa 647 (Xklp1:PRC1 ratio 20:1) and to reach
00:00 their steady-state length (black dot with SD bars in red
overlap length [µm]

area). Subsequently, Xklp1–GFP was diluted to 5 nM


4
01:22 keeping the PRC1–Alexa 647 and tubulin concentrations
constant (new Xklp1:PRC1 ratio 1:1, details in Experi-
02:44 mental Procedures). Quantification of overlap lengths
2
versus time for three examples (dark green, blue, and
red dots in green area) shows that after 6 min the over-
03:06
laps reach a new steady-state length characteristic for
0 the new, lower Xklp1 concentration (purple area).
0 3 6 9
time [min] 04:28 (D) Example time series of overlaid triple-channel TIRF
E microscopy images at the indicated times (min:s) after
06:00 Xklp1–GFP dilution and (E) corresponding kymographs
showing a representative growing overlap. The frame rate
was 1 frame per 4.67 s. Colors and horizontal scale bar as
in (A). Vertical scale bar, 1 min. See also Figure S3 and
Movie S5 and Movie S6.
merge PRC1 Xklp1

We therefore imaged single molecules of full-length Xklp1–GFP to the other microtubule of the pair (Figure 6A, left, Figure S5A,
on stabilized, individual microtubules in the absence of PRC1. We Movie S8). This highly mobile behavior of the motor was very
observed that at low ionic strength Xklp1–GFP dimers indeed different from that of single PRC1–Alexa 647 molecules (5 pM)
moved processively (Figure 5B) with an average run length of 1.2 in the presence of excess PRC1–Alexa 488 (5 nM) in the overlap,
mm (Figure 5C) and an average velocity of 0.8 mm/s (Figure 5F, which only diffused for very short distances (Figure 6A, right). The
red). Reduction in ionic strength was necessary because binding dwell time distribution of Xklp1–GFP in PRC1-containing antipar-
of Xklp1 to individual microtubules was very weak. Processivity allel overlaps was roughly exponential (Figure 6B, black dots)
was an intrinsic property of the dimerized motor domain, as with a calculated average dwell time of 5.9 s (dashed red line).
demonstrated by imaging a GFP-labeled motor fragment (Figures This demonstrates that Xklp1 dwells longer in a PRC1-decorated
5D–5G). Run length (0.9 mm, Figure 5E) and velocity (0.9 mm/s, antiparallel overlap as compared to on individual microtubules in
Figure 5F, black) of Xklp1506–GFP were in a similar range as the the absence of PRC1 at low ionic strength (Figure 6B, gray
values for the full-length motor. The longer dwell times of the circles), and also longer than the individual PRC1 molecules in
full-length motor (Figure S4A) are probably a consequence of the antiparallel overlap (Figure 6B, blue dots). This shows that
a secondary microtubule-binding site (Figure S4B). These results PRC1 increases the residence time of Xklp1 in the overlap prob-
demonstrate that Xklp1 is a processive motor, suggesting that it ably by offering additional binding sites. Mean squared displace-
might measure the overlap length by processive movement. ment (MSD) analysis confirmed that Xklp1 was very mobile in
PRC1-containing overlaps (Figure 6C, black dots), in contrast
Xklp1 Determines Overlap Length by a Processive to slowly diffusing PRC1 molecules (Figure 6C, blue dots;
Overlap Exploration Mechanism Figure S1C). However, Xklp1 explored shorter distances in
To test the hypothetical mechanism of processive overlap PRC1-containing overlaps than on individual microtubules in
length measurement, we imaged single full-length Xklp1–GFP the absence of PRC1 under low ionic strength conditions
molecules (500 pM) in the presence of excess PRC1–Alexa 647 (Figure 6C, gray dots). The MSD curve of Xklp1 in antiparallel over-
(5 nM) in dynamic antiparallel microtubule pairs at high time reso- laps could be well described using a ‘‘persistent random walk’’
lution. Strikingly, Xklp1–GFP was very mobile in PRC1-containing model (Figure 6C, dashed red line, Extended Experimental Proce-
antiparallel overlaps. Xklp1–GFP showed processive movement dures) (Othmer et al., 1988; Tranquillo and Lauffenburger, 1987). A
often abruptly changing direction, indicating a transition from one fit to the data yielded a characteristic persistence time between

426 Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc.


A Xklp1 PRC1 microtubles scheme
Figure 5. Xklp1 Is a Processive Motor
(A) Time sequence (min:s) of multichannel TIRF micros-
copy images of PRC1–Alexa 647 (red, 5 nM) and Xklp1–
GFP (green, 15 nM) in a dynamic Alexa 568-labeled
+ microtubule pair (blue) forming an antiparallel overlap by
a lateral encounter between one microtubule plus end
and one microtubule segment distant from its growing
end, as illustrated in the scheme (right). Scale bar, 5 mm.
+
(B) Representative kymograph of individual full-length
Xklp1–GFP dimers (green, 2.5 nM) on an individual stabi-
00:00 00:47 01:33 02:20 03:07 lized microtubule (red) at low ionic strength (see Extended
Experimental Procedures) imaged at high time resolution
(10 frames/s). Horizontal scale bars, 10 mm. Vertical
B Xklp1 microtubles
D Xklp1506 microtubles
F scale bar, 5 s.
35
Xklp1 (C) Histogram of the run length distribution of full-length
30 fit, v = 0.82 µm/s Xklp1–GFP with a monoexponential fit (red dashed);
Xklp1506
MSD [µm2] 25 fit, v = 0.93 µm/s mean run length was 1.20 mm (95% CI in brackets).
20 (D) Representative kymograph of individual Xklp1506–GFP
15 dimers (green, 20 nM) on an individual taxol-stabilized
microtubule (red) at very low ionic strength conditions
10
(see Extended Experimental Procedures). Imaging and
5
bars as in (B).
0
0 1 2 3 4 5 6 (E) Histogram of the run length distribution of Xklp1506–
time [s] GFP with a monoexponential fit (red dashed); mean run
C E G length was 0.91 mm (95% CI in brackets).
400 300 300
Xklp1 Xklp1506 (F) Mean squared displacement (MSD) curve of full-length
L = 1.20 µm L = 0.91 µm
300 [1.122 ; 1.273] [0.860 ; 0.967] Xklp1–GFP (red) and truncated Xklp1506–GFP (black) and
Xklp1
200 200 Xklp1506 parabolic fits (dashed lines, see Experimental Proce-
counts

counts
counts

dKin401
200 dures). Error bars are SEM.
(G) Histograms of the initial brightness of Xklp1–GFP,
100 100
100 Xklp1506–GFP, and dimeric Kin401–GFP as a control (Telley
et al., 2009), indicating that both fragment and full-length
0 0 0
Xklp1 are dimeric.
0 5 10 15 20 0 5 10 15 20 2 4 6 8 10 12 14 16 18 20 See also Figure S4 and Movie S7.
run length [µm] run length [µm] intensity [102 au]

directional switches of 2.3 s and a velocity of 0.5 mm/s for average exploratory range limits the maximum size of the
Xklp1 in the overlap. For time intervals larger than the persistence steady-state antiparallel microtubule overlap length to 5–6 mm
time, the movement of Xklp1 was effectively diffusive. (Figure 4B, Figure 6D). This defines a processive exploration
To estimate which distances Xklp1 can explore in PRC1- mechanism for antiparallel microtubule overlap length control.
decorated antiparallel microtubule overlaps, we calculated the
distribution of average exploratory distances (Figure 6D, black DISCUSSION
dots, Extended Experimental Procedures). This analysis reveals
that Xklp1 can explore average lengths of up to 6 mm. This is The Mechanism of Antiparallel Overlap Generation
in strong contrast to the low exploratory distances of PRC1 and Length Control by PRC1/Kinesin-4 Explains
(Figure 6D, blue dots), which can only move for distances of up Anaphase Spindle Characteristics In Vivo
to 0.2 mm. Strikingly, the average exploratory distances of We have elucidated a molecular mechanism capable of selective
individual Xklp1 molecules coincide well with the range of formation and adaptable length control of antiparallel microtubule
steady-state lengths of antiparallel microtubule overlaps (Fig- overlaps by two conserved midzone proteins. Central to this
ure 4B). This suggests that, by processive movement, Xklp1 rea- mechanism is the combination of four distinct molecular activities
ches the plus ends of antiparallel overlapping microtubules, (Figure 7): (1) bundling of microtubules with high selectivity for
where it can modulate microtubule dynamics. To test whether antiparallel orientation by PRC1, (2) recruitment of kinesin-4 by
the processive nature of Xklp1’s motility is indeed central to the PRC1 to antiparallel overlaps, and (3) processive microtubule
mechanism of steady-state length establishment, we replaced plus-end-directed motility together with (4) an inhibitory effect
adenosine-50 -triphosphate (ATP) by adenosine-50 -diphosphate on microtubule growth by the motor protein kinesin-4. The combi-
(ADP) in the TIRF microscopy experiment, thereby preventing nation of these activities constitutes a minimal two-component
processive motility of Xklp1 but not its recruitment by PRC1. We system capable of formation of a minimal midzone in vitro.
observed that under these conditions the two midzone proteins Our in vitro reconstitution explains why loss of either PRC1 or
failed to produce stable steady-state overlap lengths (Figure S6). kinesin-4 leads to nonequivalent defects in midzone formation
Instead, overlap microtubules continued growing despite the in vivo (Kurasawa et al., 2004; Mollinari et al., 2002; Zhu and
presence of PRC1 and Xklp1 in the overlap. We conclude that Jiang, 2005). We show that selective antiparallel microtubule
processive overlap exploration allows Xklp1 to reach and modu- crosslinking is an intrinsic property of vertebrate PRC1, an
late the behavior of microtubule plus ends and that its finite activity which appears to be evolutionarily conserved (Janson

Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc. 427


A Xklp1 in overlap PRC1 in overlap Figure 6. Xklp1 Determines Antiparallel Microtu-
bule Overlap Length by Processive Overlap
scheme Exploration
(A) Single-molecule imaging of either individual Xklp1–
microtubule/PRC1 image
before time-lapse GFP molecules (500 pM, green) in a PRC1–Alexa 647
(5 nM, blue) containing antiparallel microtubule overlap
(red) (right panel) or individual PRC1–Alexa 647 molecules
(5 pM, green) in a PRC1–Alexa 488 (5 nM, blue) containing
antiparallel microtubule overlap (red) (left panel). Scheme
(top) and triple-channel TIRF microscopy images of the
microtubule pair before (upper image) and after (lower
image) fast time-lapse imaging of individual Xklp1–GFP
or PRC1–Alexa 647 molecules, as illustrated in a kymo-
graph (middle). The frame rate was 10 s1. Horizontal
kymograph of single-
molecule time-lapse scale bar, 10 mm. Vertical scale bar, 5 s.
(B) Dwell time distribution (shown as 1 – CDF) of single
Xklp1–GFP molecules moving in antiparallel microtubule
overlap regions (conditions as in A, right; black dots, n =
226) and a monoexponential fit (dashed red); mean dwell
time is 5.85 ± 0.12 s (mean ± 95% CI). For comparison,
the distributions for Xklp1–GFP on single taxol-microtu-
bules (gray circles, under conditions as in Figure 5B) and
for PRC1–Alexa 647 in a PRC1–Alexa 488-containing anti-
parallel overlap (blue dots, under conditions as in A, left)
microtubule/PRC1 image are shown.
after time-lapse
(C) Mean squared displacement (MSD) curves for the same
Xklp1 (single molecules) PRC1 (single molecules) conditions as shown in (B), and a fit to the data of Xklp1–
microtubules microtubules
GFP in antiparallel overlaps using the ‘‘persistent random
PRC1 (bulk protein in overlap) PRC1 (bulk protein in overlap)
walk’’ model, yielding persistence time Tp = 2.31 ± 0.68 s
and mean velocity v = 0.52 ± 0.06 mm/s (mean ± 95% CI,
30 Xklp1 in overlap
B 100 C Xklp1 on single MT
see Experimental Procedures). Error bars are SEM.
Xklp1 in overlap 25 PRC1 in overlap (D) Calculated probability distribution of the average
Xklp1 on single MT exploratory distance (black dots, see Extended Experi-
PRC1 in overlap 20
MSD [µm2]

mental Procedures) of Xklp1–GFP molecules in antiparallel


1−CDF

10-1
15 overlaps and the predicted probability curve (dashed red)
calculated from the persistent random walk model using
10
the parameter values as obtained from the fits in (B) and
5 (C). For comparison, calculated average exploratory
0 distance distributions are plotted for Xklp1–GFP on single
10-2
0 5 10 15 20 25 30 0 2 4 6 8 10 12 14 16 stabilized microtubules and PRC1–Alexa 647 in overlaps
dwell time [s] time [s] (as in B and C). Inset: semi-logarithmic plot of average
D 1.0
100 exploratory distances.
See also Figure S5, Figure S6, and Movie S8.
calculated probability

0.8 10-1

0.6
10-2
0.4
0 2 4 6 8 10 Xenopus kinesin-4 Xklp1 binds only weakly to
Xklp1 in overlap microtubules. However, by interacting with
Xklp1 on single MT
0.2 PRC1 in overlap PRC1, Xklp1 is recruited to antiparallel microtu-
0
bule overlaps, reminiscent of selective recruit-
0 2 4 6 8 10 ment of sliding motors of the kinesin-5 and kine-
avg. exploratory distance [µm]
sin-6 families by Ase1 in budding yeast and
fission yeast, respectively (Fu et al., 2009; Khme-
et al., 2007). Autonomous bundling of antiparallel microtubules linskii et al., 2009). This recruitment results in the local inhibition of
places PRC1 at the core of the activities essential for midzone microtubule plus-end growth in antiparallel overlaps by Xklp1,
formation and explains why PRC1 is absolutely required for the leading to a defined steady-state overlap length. This explains
bundling and stabilization of antiparallel microtubule overlaps, why the midzone unnaturally extends but half-spindles remain
as indicated by the complete loss of central anaphasic microtu- connected upon loss of kinesin-4 in vivo, resulting in long, distorted
bule bundles in the absence of PRC1 in vivo (Kurasawa et al., anaphase spindles (Kurasawa et al., 2004; Zhu and Jiang, 2005).
2004; Mollinari et al., 2002; Zhu and Jiang, 2005).
Vertebrate PRC1 has previously been shown to interact with Processive Overlap Exploration by Kinesin-4 Ensures
kinesin-4 during anaphase, an interaction necessary for proper Adaptive Overlap Length Control
midzone organization (Kurasawa et al., 2004; Zhu and Jiang, Xklp1 reduces the dynamicity of microtubules by lowering the
2005). Here, we establish a hierarchy of interactions: full-length turnover of tubulin at the growing microtubule end. On individual

428 Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc.


Xklp1 at growing microtubule ends (Figure 5A) is likely to be
MT seed growth
responsible for the dose-dependent inhibition of microtubule
Recognition
- + MT pair
growth (Figure 4). The targeting of Xklp1 to growing microtubule
+ -
PRC1 single MT ends is reminiscent of the manner by which the microtubule de-
+ - polymerizing kinesins Kip3 (kinesin-8) (Varga et al., 2006, 2009)
and MCAK (kinesin-13) (Helenius et al., 2006) reach the ends of
artificially stabilized microtubules. Both proteins are ‘‘collected’’
Recruitment - +
+
- from an extended region near the end of stabilized microtubules
Xklp1
in vitro either by directional (Kip3) or diffusive (MCAK) motility.
+ - Xklp1 (kinesin-4) is also collected from an extended region,
however, in this case from the microtubule overlap by processive
bidirectional motility (Figure 6A, Figure S5A). For such an
Processive - +
- ‘‘antenna mechanism’’ to be effective under conditions where
Exploration +
microtubules grow, it is important that the velocity of Xklp1 in
+ - the overlap (0.5 mm/s [Figure 6C, Figure S5B]) is considerably
faster than the velocity with which the microtubules elongate
(35 nm/s in our in vitro experiments and 200 nm/s in vivo
STOP
Growth - [Rusan et al., 2001; Tournebize et al., 1997]). This might explain
Inhibition STOP - why Xklp1 is a fast motor in comparison to other mitotic kinesins.
The lowest Xklp1 concentration (5 nM) required to form antipar-
+ -
allel microtubule overlaps with stable steady-state lengths
resulted in overlaps of around 5 mm (Figure 4B). This length is in
Figure 7. A Two-Component System Controls Antiparallel Microtu- remarkably good agreement with the maximal exploratory
bule Overlaps distance of individual Xklp1 molecules in PRC1-enriched antipar-
Model illustrating schematically the activities that lead in their combination to allel overlaps (Figure 6D). This suggests that the threshold amount
antiparallel microtubule overlap formation and length control (see Discussion). of Xklp1 required to induce a complete stop of microtubule growth
is reached at this concentration by exploiting the entire explor-
microtubules this activity can be observed in vitro only at highly atory range of Xklp1 molecules in the overlap. At higher concen-
elevated Xklp1 concentrations (several hundred nanomolar) well trations, the threshold amount is already reached by ‘‘collecting’’
above physiological levels (Bieling et al., 2010a). In contrast, from shorter overlaps, leading to characteristic overlap lengths as
recruitment of Xklp1 to antiparallel microtubule overlaps by a function of the Xklp1 concentration. In Xenopus egg extract, the
PRC1 results in complete inhibition of microtubule growth concentration of Xklp1 is approximately 100 nM (Bieling et al.,
already at lower, close-to physiological concentrations at which 2010a). Our experiments predict that such concentrations lead
individual microtubules remain unaffected in their growth. As to overlap lengths of 2 mm (Figure 4B), which is in agreement
a consequence, specific recruitment of Xklp1 ensures the local with a midzone length of 2–3 mm as measured in vivo (Kurasawa
inhibition of growth of only those microtubules engaged in anti- et al., 2004; Mastronarde et al., 1993; Zhu and Jiang, 2005).
parallel, PRC1-decorated microtubule overlaps. Activation of The PRC1/kinesin-4 system offers adaptive, self-regulatory
the PRC1/kinesin-4 system, through dephosphorylation at control of overlap size by introducing a negative feedback
anaphase onset, therefore results in tight bundling and growth between overlap length and microtubule growth. Shortening
inhibition of antiparallel microtubule overlaps in the central the microtubule overlaps as a consequence of motor-driven
spindle (Kurasawa et al., 2004; Mollinari et al., 2002; Zhu and microtubule sliding would result in lowered amounts of kinesin-
Jiang, 2005), whereas microtubules residing outside of the mid- 4 at the microtubule ends because the motor could be collected
zone are likely not affected by the PRC1/kinesin-4 system. now only from shorter distances. Similar to our reversibility
Xklp1 motors move processively with velocities of around experiment where we reduced the global concentration of
0.5 mm/s within the PRC1-decorated antiparallel microtubule Xklp1 (Figures 4C–4E), the system would be predicted to dynam-
overlap (Figure 6C, Figure S5B). The drag force exerted by ically respond to antiparallel sliding resulting in shortening
PRC1 on Xklp1 can be estimated to be low (Extended Experi- overlaps by initiating microtubule elongation. Local, selective
mental Procedures) compared to typical stall forces of kinesins inhibition of microtubule growth by kinesin-4 in antiparallel
(Carter and Cross, 2005; Svoboda and Block, 1994), explaining overlaps at conditions that globally favor microtubule elongation
why Xklp1 is slowed down only mildly by interacting with therefore allows for a dynamic control of overlap length.
PRC1. This explains why Xklp1 efficiently explores the antipar-
allel overlap instead of generating substantial microtubule sliding Antiparallel Microtubule Overlaps in Yeasts
forces. This contrasts Xklp1 to antiparallel microtubule sliding and Metazoans
motors like kinesin-5, which reside relatively static between Maintenance of stable antiparallel microtubule overlaps in the
the sliding microtubules (Kapitein et al., 2005; Kapoor and spindle center requires coordination between microtubule
Mitchison, 2001; Miyamoto et al., 2004; Uteng et al., 2008). (de)polymerization and microtubule sliding. In particular, spindle
How does processive movement of Xklp1 within the antipar- elongation during anaphase B must not compromise spindle
allel overlap region result in length control? Accumulation of stability by potentially reducing the length of the antiparallel

Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc. 429


overlap region. Several proposals have been made for mecha- defined length in vitro, an essential process for proper midzone
nisms to locally regulate microtubule dynamics in the anaphase formation in metazoan anaphase spindles. The in vitro reconsti-
spindle (Cheerambathur et al., 2007; Gardner et al., 2008; Pear- tution allowed us to dissect the mechanism of autonomous
son et al., 2006). Interestingly, recent work in fission yeast has antiparallel microtubule recognition by PRC1 and of local micro-
linked the conserved microtubule-binding protein CLASP to tubule growth inhibition and overlap length control by kinesin-4.
the stabilization of antiparallel microtubule bundles (Bratman In the future, the extension of our reconstitution approach by
and Chang, 2007). Ase1-dependent recruitment of CLASP to including other midzone proteins promises to yield further mech-
antiparallel microtubule overlaps has been suggested to prevent anistic understanding of aspects of the organization and function
microtubule depolymerization past the overlap by inducing of the late mitotic spindle.
microtubule rescue (Bratman and Chang, 2007). A similar
PRC1-dependent stabilizing function of CLASP for antiparallel EXPERIMENTAL PROCEDURES
microtubule overlaps during anaphase appears also to exist in
cultured human cells (Liu et al., 2009). Dynamic Microtubule Overlap Assay
A flow chamber assembled from a biotin-PEG functionalized glass coverslip
Although local induction of microtubule rescue might explain
and a PLL-PEG passivated glass slide separated by two stripes of double-
how overlap stability is ensured, it does not readily explain how sided tape (Bieling et al., 2010b) was filled with a series of solutions: (1)
microtubule overgrowth is prevented and thus how a defined Pluronic F-127 and k-casein, (2) NeutrAvidin and k-casein, (3) buffer, (4) short
overlap size is established. This is especially the case for meta- GMP–CPP stabilized, biotinylated brightly labeled microtubule seeds (contain-
zoan systems in which microtubule growth velocities are high ing 26% Alexa Fluor 568-labeled tubulin), (5) buffer. Microtubule growth was
(Kinoshita et al., 2001). Our in vitro experiments have demon- initiated by flowing in dimly labeled tubulin (containing 10% Alexa Fluor
strated how the PRC1/kinesin-4 system can control antiparallel 568-labeled tubulin) together with midzone proteins at the indicated concen-
trations. The final assay buffer was 80 mM K-PIPES, pH 6.8, 85 mM KCl,
overlap length at conditions that globally favor microtubule elon-
85 mM K-acetate, 4 mM MgCl2, 1 mM GTP, 2 mM MgATP, 1 mM EGTA,
gation. It might be the combination of inhibition of microtubule 10 mM b-mercaptoethanol, 0.25% [wt/wt] Brij-35, 0.1% methyl cellulose,
growth by kinesin-4 and the promotion of rescues by CLASP 100 mg/ml b-casein, and oxygen scavengers. If not stated otherwise, the
that ensures the stabilization and length control of antiparallel concentrations of proteins were 5 nM PRC1–Alexa 647, 15 nM Xklp1–GFP
microtubule overlaps in metazoans. The adaptive nature of this or Xklp1506–GFP, and 17.5 mM dimly labeled tubulin.
mechanism would allow for independent regulation of antiparallel To test the reversibility of antiparallel microtubule overlap formation, over-
laps were allowed to form in the presence of 5 nM PRC1–Alexa 647, 100 nM
microtubule sliding activities at different stages of anaphase.
Xklp1–GFP (PRC1:Xklp1 ratio of 1:20), and 17.5 mM dimly labeled tubulin for
In contrast to metazoan anaphase spindles, the length of 7–10 min at 28 C ± 1 C. Then the PRC1:Xklp1 ratio was changed to 1:1 by
antiparallel microtubule overlaps does not appear to be tightly flowing in a solution with only 5 nM Xklp1–GFP, leaving all other concentrations
controlled in yeast. Microtubule plus ends are dynamic in fission constant.
yeast anaphase spindles as well as in bundled interphase arrays Multicolor time-lapse imaging using TIRF microscopy was always
(Sagolla et al., 2003). The slower microtubule growth velocities performed at 28 C ± 1 C.
in yeast might be one reason for less stringent control of the
Single-Molecule Imaging
overlap size in this organism. In addition, yeast uses the proces-
Single full-length Xklp1–GFP molecules were imaged at a concentration of
sive motor kinesin-8 (Klp5/6 in fission yeast and Kip3 in budding
2.5 nM on individual paclitaxel-stabilized, biotinylated, and fluorescently
yeast) as the major microtubule plus-end-destabilizing factor labeled microtubules (5% Alexa 568-labeled tubulin) in low ionic strength
(Cottingham et al., 1999; Gupta et al., 2006; West et al., 2001). buffer (80 mM K-PIPES, pH 6.8, 85 mM K-acetate, 4 mM MgCl2, 2 mM MgATP,
Length-dependent destabilization of microtubules by kinesin-8 1 mM EGTA, 10 mM b-mercaptoethanol, 10 mM paclitaxel, 0.25% [wt/wt]
(Tischer et al., 2009; Varga et al., 2006) might be sufficient for Brij-35, 100 mg/ml b-casein, and oxygen scavengers). Xklp1506–GFP at 20
a less stringent control of overlap stability. This might explain nM, or Kin401–GFP at 0.1 nM as a control, was imaged in very low ionic strength
buffer (12 mM K-PIPES, pH 6.8, 2 mM MgCl2, 2 mM MgATP, 1 mM EGTA,
the absence of kinesin-4 from both fission and budding yeast.
10 mM b-mercaptoethanol, 10 mM paclitaxel, 100 mg/ml b-casein, and oxygen
During interphase, fission yeast also uses antiparallel microtu- scavengers).
bule overlaps as means to organize the microtubule cytoskeleton. For imaging individual PRC1–Alexa 647 molecules in dynamic antiparallel
Recently, a model for the formation of such overlaps was microtubule overlaps, the PRC1–Alexa 647 concentration was lowered to 50
proposed based on the combined action of the sliding motor pM or to 5 pM in the presence of 5 nM PRC1–Alexa 488. For imaging individual
Klp2 (a kinesin-14) and Ase1 (Janson et al., 2007). In this model, Xklp1–GFP molecules in dynamic, PRC1-containing antiparallel microtubule
overlaps, the Xklp1–GFP concentration was lowered to 500 pM.
antiparallel microtubules slide with respect to each other driven
by minus-end-directed motors targeted selectively to microtu-
Data Analysis
bule plus ends. Sliding stops either as a consequence of plus The movements of single fluorescent molecules were analyzed using auto-
ends growing beyond the overlap region or possibly by friction ex- mated particle tracking (Kalaimoscope, TransInsight, Dresden, Germany)
erted by overlap-enriched Ase1 (Janson et al., 2007). This leads to (Telley et al., 2009). For dwell time distributions, the cumulative distribution
the formation of antiparallel microtubule overlaps with inverted function (CDF) of data was calculated using the ‘‘ecdf’’ function in Matlab
polarity and dynamically varying sizes, a mechanism that differs (MathWorks), and either a monoexponential or a biexponential function was
from that of tight overlap size control during metazoan anaphase. fitted to (1–CDF) data using a ‘‘least-squares’’ approach. Histograms of run
lengths were fitted with a monoexponential function as described (Telley et al.,
2009). MSD curves were calculated and fitted using the function v2 Dt2 + 2D
Conclusion Dt + offset, with the mean velocity v and the diffusion coefficient D. For the
We have reconstituted a two-component system necessary and case of Xklp1 in the overlap the MSD curve was fitted to a ‘‘persistent
sufficient to form antiparallel microtubule overlaps with precisely random walk’’ model (Othmer et al., 1988; Tranquillo and Lauffenburger,

430 Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc.


1987): MSD = 2v2Tp [Dt – Tp (1 – exp(–Dt/Tp)] with the persistence time Tp and Fu, C., Ward, J.J., Loiodice, I., Velve-Casquillas, G., Nedelec, F.J., and Tran,
mean velocity v, and hence v2Tp the effective diffusion coefficient. The P.T. (2009). Phospho-regulated interaction between kinesin-6 Klp9p and
‘‘average exploratory distance’’ distribution in microtubule overlaps was microtubule bundler Ase1p promotes spindle elongation. Dev. Cell 17,
calculated by combining information from the dwell time distribution and the 257–267.
MSD curve in antiparallel microtubule overlaps, thereby eliminating time. Gardner, M.K., Bouck, D.C., Paliulis, L.V., Meehl, J.B., O’Toole, E.T., Haase,
Microtubule dynamics was determined by kymograph analysis using ImageJ. J., Soubry, A., Joglekar, A.P., Winey, M., Salmon, E.D., et al. (2008).
The growth versus tubulin concentration relationship in the presence or Chromosome congression by Kinesin-5 motor-mediated disassembly of
absence of Xklp1 was analyzed using a multiple linear regression model with longer kinetochore microtubules. Cell 135, 894–906.
a categorical factor g (+Xklp1: g = 0, –Xklp1: g = 1) and y = b1 + m1 x + g
Glotzer, M. (2009). The 3Ms of central spindle assembly: microtubules, motors
(b2 + m2 x). Generally, this analysis provides a significance test for differences
and MAPs. Nat. Rev. Mol. Cell Biol. 10, 9–20.
in the slope m and abscissa b of two linear relationships. Here, the slope and
abscissa represent the kinetic parameters kon and koff, respectively, of the Gupta, M.L., Jr., Carvalho, P., Roof, D.M., and Pellman, D. (2006). Plus
microtubule dynamics. end-specific depolymerase activity of Kip3, a kinesin-8 protein, explains its
role in positioning the yeast mitotic spindle. Nat. Cell Biol. 8, 913–923.

SUPPLEMENTAL INFORMATION Helenius, J., Brouhard, G., Kalaidzidis, Y., Diez, S., and Howard, J. (2006).
The depolymerizing kinesin MCAK uses lattice diffusion to rapidly target
Supplemental Information includes Extended Experimental Procedures, six microtubule ends. Nature 441, 115–119.
figures, and eight movies and can be found with this article online at doi:10. Howard, J., and Hyman, A.A. (2007). Microtubule polymerases and depoly-
1016/j.cell.2010.06.033. merases. Curr. Opin. Cell Biol. 19, 31–35.
Janson, M.E., de Dood, M.E., and Dogterom, M. (2003). Dynamic instability of
ACKNOWLEDGMENTS microtubules is regulated by force. J. Cell Biol. 161, 1029–1034.
Janson, M.E., Loughlin, R., Loiodice, I., Fu, C., Brunner, D., Nedelec, F.J., and
We thank Vladimir Rybin for analytical ultracentrifugation, Jan Ellenberg,
Tran, P.T. (2007). Crosslinkers and motors organize dynamic microtubules to
Elmar Schiebel, Johanna Roostalu, and Scott Hansen for critically reading
form stable bipolar arrays in fission yeast. Cell 128, 357–368.
the manuscript, and the DFG, HFSPO, the European Commission (STREP
‘‘Active Biomics’’), and the Swiss National Science Foundation for financial Jiang, W., Jimenez, G., Wells, N.J., Hope, T.J., Wahl, G.M., Hunter, T., and
support. Fukunaga, R. (1998). PRC1: A human mitotic spindle-associated CDK
substrate protein required for cytokinesis. Mol. Cell 2, 877–885.
Received: January 4, 2010 Kapitein, L.C., Peterman, E.J., Kwok, B.H., Kim, J.H., Kapoor, T.M., and
Revised: April 19, 2010 Schmidt, C.F. (2005). The bipolar mitotic kinesin Eg5 moves on both microtu-
Accepted: June 7, 2010 bules that it crosslinks. Nature 435, 114–118.
Published: August 5, 2010 Kapitein, L.C., Janson, M.E., van den Wildenberg, S.M., Hoogenraad, C.C.,
Schmidt, C.F., and Peterman, E.J. (2008). Microtubule-driven multimerization
REFERENCES recruits ase1p onto overlapping microtubules. Curr. Biol. 18, 1713–1717.
Kapoor, T.M., and Mitchison, T.J. (2001). Eg5 is static in bipolar spindles
Arnal, I., Heichette, C., Diamantopoulos, G.S., and Chretien, D. (2004).
relative to tubulin: evidence for a static spindle matrix. J. Cell Biol. 154,
CLIP-170/tubulin-curved oligomers coassemble at microtubule ends and
1125–1133.
promote rescues. Curr. Biol. 14, 2086–2095.
Khmelinskii, A., and Schiebel, E. (2008). Assembling the spindle midzone in the
Bieling, P., Kronja, I., and Surrey, T. (2010a). Microtubule motility on reconsti-
right place at the right time. Cell Cycle 7, 283–286.
tuted meiotic chromatin. Curr. Biol. 20, 763–769.
Khmelinskii, A., Lawrence, C., Roostalu, J., and Schiebel, E. (2007).
Bieling, P., Telley, I.A., Hentrich, C., Piehler, J., and Surrey, T. (2010b).
Cdc14-regulated midzone assembly controls anaphase B. J. Cell Biol. 177,
Fluorescence microscopy assays on chemically functionalized surfaces for
981–993.
quantitative imaging of microtubule, motor, and +TIP dynamics. Methods
Cell Biol. 95, 555–580. Khmelinskii, A., Roostalu, J., Roque, H., Antony, C., and Schiebel, E. (2009).
Bratman, S.V., and Chang, F. (2007). Stabilization of overlapping microtubules Phosphorylation-dependent protein interactions at the spindle midzone
by fission yeast CLASP. Dev. Cell 13, 812–827. mediate cell cycle regulation of spindle elongation. Dev. Cell 17, 244–256.

Bringmann, H., Skiniotis, G., Spilker, A., Kandels-Lewis, S., Vernos, I., and Kinoshita, K., Arnal, I., Desai, A., Drechsel, D.N., and Hyman, A.A. (2001).
Surrey, T. (2004). A kinesin-like motor inhibits microtubule dynamic instability. Reconstitution of physiological microtubule dynamics using purified compo-
Science 303, 1519–1522. nents. Science 294, 1340–1343.

Brouhard, G.J., Stear, J.H., Noetzel, T.L., Al-Bassam, J., Kinoshita, K., Kurasawa, Y., Earnshaw, W.C., Mochizuki, Y., Dohmae, N., and Todokoro, K.
Harrison, S.C., Howard, J., and Hyman, A.A. (2008). XMAP215 is a processive (2004). Essential roles of KIF4 and its binding partner PRC1 in organized
microtubule polymerase. Cell 132, 79–88. central spindle midzone formation. EMBO J. 23, 3237–3248.
Carter, N.J., and Cross, R.A. (2005). Mechanics of the kinesin step. Nature 435, Kwon, M., Morales-Mulia, S., Brust-Mascher, I., Rogers, G.C., Sharp, D.J., and
308–312. Scholey, J.M. (2004). The chromokinesin, KLP3A, dives mitotic spindle pole
separation during prometaphase and anaphase and facilitates chromatid
Cheerambathur, D.K., Civelekoglu-Scholey, G., Brust-Mascher, I., Sommi, P.,
motility. Mol. Biol. Cell 15, 219–233.
Mogilner, A., and Scholey, J.M. (2007). Quantitative analysis of an anaphase B
switch: predicted role for a microtubule catastrophe gradient. J. Cell Biol. 177, Lee, Y.M., and Kim, W. (2004). Kinesin superfamily protein member 4 (KIF4) is
995–1004. localized to midzone and midbody in dividing cells. Exp. Mol. Med. 36, 93–97.
Cottingham, F.R., Gheber, L., Miller, D.L., and Hoyt, M.A. (1999). Novel roles Liu, J., Wang, Z., Jiang, K., Zhang, L., Zhao, L., Hua, S., Yan, F., Yang, Y.,
for saccharomyces cerevisiae mitotic spindle motors. J. Cell Biol. 147, Wang, D., Fu, C., et al. (2009). PRC1 cooperates with CLASP1 to organize
335–350. central spindle plasticity in mitosis. J. Biol. Chem. 284, 23059–23071.
Drechsel, D.N., Hyman, A.A., Cobb, M.H., and Kirschner, M.W. (1992). Loiodice, I., Staub, J., Setty, T.G., Nguyen, N.P., Paoletti, A., and Tran, P.T.
Modulation of the dynamic instability of tubulin assembly by the (2005). Ase1p organizes antiparallel microtubule arrays during interphase
microtubule-associated protein tau. Mol. Biol. Cell 3, 1141–1154. and mitosis in fission yeast. Mol. Biol. Cell 16, 1756–1768.

Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc. 431


Mastronarde, D.N., McDonald, K.L., Ding, R., and McIntosh, J.R. (1993). Tranquillo, R.T., and Lauffenburger, D.A. (1987). Stochastic model of leukocyte
Interpolar spindle microtubules in PTK cells. J. Cell Biol. 123, 1475–1489. chemosensory movement. J. Math. Biol. 25, 229–262.
Miyamoto, D.T., Perlman, Z.E., Burbank, K.S., Groen, A.C., and Mitchison, T.J. Uteng, M., Hentrich, C., Miura, K., Bieling, P., and Surrey, T. (2008). Poleward
(2004). The kinesin Eg5 drives poleward microtubule flux in Xenopus laevis egg transport of Eg5 by dynein-dynactin in Xenopus laevis egg extract spindles.
extract spindles. J. Cell Biol. 167, 813–818. J. Cell Biol. 182, 715–726.
Mollinari, C., Kleman, J.P., Jiang, W., Schoehn, G., Hunter, T., and Margolis, Varga, V., Helenius, J., Tanaka, K., Hyman, A.A., Tanaka, T.U., and Howard, J.
R.L. (2002). PRC1 is a microtubule binding and bundling protein essential to (2006). Yeast kinesin-8 depolymerizes microtubules in a length-dependent
maintain the mitotic spindle midzone. J. Cell Biol. 157, 1175–1186. manner. Nat. Cell Biol. 8, 957–962.
Muller, S., Smertenko, A., Wagner, V., Heinrich, M., Hussey, P.J., and Hauser, Varga, V., Leduc, C., Bormuth, V., Diez, S., and Howard, J. (2009). Kinesin-8
M.T. (2004). The plant microtubule-associated protein AtMAP65-3/PLE is motors act cooperatively to mediate length-dependent microtubule depoly-
essential for cytokinetic phragmoplast function. Curr. Biol. 14, 412–417. merization. Cell 138, 1174–1183.

Othmer, H.G., Dunbar, S.R., and Alt, W. (1988). Models of dispersal in Verbrugghe, K.J., and White, J.G. (2004). SPD-1 is required for the formation of
biological systems. J. Math. Biol. 26, 263–298. the spindle midzone but is not essential for the completion of cytokinesis in C.
elegans embryos. Curr. Biol. 14, 1755–1760.
Pearson, C.G., Gardner, M.K., Paliulis, L.V., Salmon, E.D., Odde, D.J., and
Bloom, K. (2006). Measuring nanometer scale gradients in spindle microtubule Verni, F., Somma, M.P., Gunsalus, K.C., Bonaccorsi, S., Belloni, G., Goldberg,
dynamics using model convolution microscopy. Mol. Biol. Cell 17, 4069–4079. M.L., and Gatti, M. (2004). Feo, the Drosophila homolog of PRC1, is required
for central-spindle formation and cytokinesis. Curr. Biol. 14, 1569–1575.
Powers, J., Rose, D.J., Saunders, A., Dunkelbarger, S., Strome, S., and
Vernos, I., Raats, J., Hirano, T., Heasman, J., Karsenti, E., and Wylie, C. (1995).
Saxton, W.M. (2004). Loss of KLP-19 polar ejection force causes misorienta-
Xklp1, a chromosomal Xenopus kinesin-like protein essential for spindle
tion and missegregation of holocentric chromosomes. J. Cell Biol. 166,
organization and chromosome positioning. Cell 81, 117–127.
991–1001.
Walczak, C.E., and Heald, R. (2008). Mechanisms of mitotic spindle assembly
Rusan, N.M., Fagerstrom, C.J., Yvon, A.M., and Wadsworth, P. (2001). Cell
and function. Int. Rev. Cytol. 265, 111–158.
cycle-dependent changes in microtubule dynamics in living cells expressing
green fluorescent protein-alpha tubulin. Mol. Biol. Cell 12, 971–980. Walker, R.A., O’Brien, E.T., Pryer, N.K., Soboeiro, M.F., Voter, W.A., Erickson,
H.P., and Salmon, E.D. (1988). Dynamic instability of individual microtubules
Sagolla, M.J., Uzawa, S., and Cande, W.Z. (2003). Individual microtubule
analyzed by video light microscopy: rate constants and transition frequencies.
dynamics contribute to the function of mitotic and cytoplasmic arrays in fission
J. Cell Biol. 107, 1437–1448.
yeast. J. Cell Sci. 116, 4891–4903.
West, R.R., Malmstrom, T., Troxell, C.L., and McIntosh, J.R. (2001). Two
Schuyler, S.C., Liu, J.Y., and Pellman, D. (2003). The molecular function of
related kinesins, klp5+ and klp6+, foster microtubule disassembly and are
Ase1p: evidence for a MAP-dependent midzone-specific spindle matrix.
required for meiosis in fission yeast. Mol. Biol. Cell 12, 3919–3932.
Microtubule-associated proteins. J. Cell Biol. 160, 517–528.
Zhu, C., and Jiang, W. (2005). Cell cycle-dependent translocation of PRC1 on
Svoboda, K., and Block, S.M. (1994). Force and velocity measured for single the spindle by Kif4 is essential for midzone formation and cytokinesis. Proc.
kinesin molecules. Cell 77, 773–784. Natl. Acad. Sci. USA 102, 343–348.
Telley, I.A., Bieling, P., and Surrey, T. (2009). Obstacles on the microtubule Zhu, C., Zhao, J., Bibikova, M., Leverson, J.D., Bossy-Wetzel, E., Fan, J.B.,
reduce the processivity of Kinesin-1 in a minimal in vitro system and in cell Abraham, R.T., and Jiang, W. (2005). Functional analysis of human microtu-
extract. Biophys. J. 96, 3341–3353. bule-based motor proteins, the kinesins and dyneins, in mitosis/cytokinesis
Tischer, C., Brunner, D., and Dogterom, M. (2009). Force- and kinesin-8- using RNA interference. Mol. Biol. Cell 16, 3187–3199.
dependent effects in the spatial regulation of fission yeast microtubule
dynamics. Mol. Syst. Biol. 5, 250. Note Added in Proof
Tournebize, R., Andersen, S.S., Verde, F., Doree, M., Karsenti, E., and Hyman, In a manuscript by Subramanian et al., which appears in this issue of Cell, the
A.A. (1997). Distinct roles of PP1 and PP2A-like phosphatases in control of authors arrive at similar conclusions regarding the nature of microtubule cross-
microtubule dynamics during mitosis. EMBO J. 16, 5537–5549. links formed by PRC1.

432 Cell 142, 420–432, August 6, 2010 ª2010 Elsevier Inc.


Insights into Antiparallel Microtubule
Crosslinking by PRC1, a Conserved
Nonmotor Microtubule Binding Protein
Radhika Subramanian,1 Elizabeth M. Wilson-Kubalek,2 Christopher P. Arthur,2 Matthew J. Bick,3 Elizabeth A. Campbell,3
Seth A. Darst,3 Ronald A. Milligan,2 and Tarun M. Kapoor1,*
1Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY 10065, USA
2Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
3Laboratory of Molecular Biophysics, The Rockefeller University, New York, NY 10065, USA

*Correspondence: kapoor@mail.rockefeller.edu
DOI 10.1016/j.cell.2010.07.012

SUMMARY important roles in dividing and nondividing cells (Ribbeck et al.,


2006; Sasabe and Machida, 2006; Schuyler et al., 2003; Thadani
Formation of microtubule architectures, required for et al., 2009; Zeng, 2000). Current models for the functions of
cell shape maintenance in yeast, directional cell these proteins are based on cellular localizations and loss-of-
expansion in plants and cytokinesis in eukaryotes, function studies. However, we lack any structural data to explain
depends on antiparallel microtubule crosslinking how microtubule crosslinking is achieved by these MAPs.
by the conserved MAP65 protein family. Here, we Recently, there have been several advances in our under-
standing of the structure of nonmotor MAPs. Among the best
combine structural and single molecule fluorescence
characterized class of MAPs are the +TIP proteins (e.g.,
methods to examine how PRC1, the human MAP65, XMAP215, EB1 and CLIP170), which can dynamically track the
crosslinks antiparallel microtubules. We find that growing end of a microtubule. Microtubule binding in these
PRC1’s microtubule binding is mediated by a struc- proteins is mediated by calponin-homology, CAP/Gly or TOG
tured domain with a spectrin-fold and an unstruc- domains (Slep and Vale, 2007). Similarly, structural work on
tured Lys/Arg-rich domain. These two domains, at Ndc80, a conserved mitotic MAP, has revealed how a calpolin-
each end of a homodimer, are connected by a linkage homology domain may be used to establish kinetochore-micro-
that is flexible on single microtubules, but forms well- tubule associations during cell division (Ciferri et al., 2008; Wei
defined crossbridges between antiparallel filaments. et al., 2007; Wilson-Kubalek et al., 2008). However, due to lack
Further, we show that PRC1 crosslinks are compliant of similarity in primary sequence, it is not very likely that these
and do not substantially resist filament sliding by structural models will shed light on nonmotor MAPs that can
crosslink two microtubules.
motor proteins in vitro. Together, our data show
As a step toward developing structural models for how non-
how MAP65s, by combining structural flexibility and motor MAPs may crosslink microtubules, we focused on the
rigidity, tune microtubule associations to establish conserved MAP65 family, which plays key roles in microtubule
crosslinks that selectively ‘‘mark’’ antiparallel over- organization in eukaryotes. Since their initial discovery in bud-
lap in dynamic cytoskeletal networks. ding yeast, microtubule crosslinking functions of the MAP65
proteins have been shown to be required for cell shape mainte-
INTRODUCTION nance in yeast cells, directional cell expansion in plants and
formation of the central spindle in eukaryotes (Chan et al.,
The dynamic reorganization of microtubule networks plays a crit- 1999; Jiang et al., 1998; Loiodice et al., 2005; Yamashita et al.,
ical role in diverse biological processes, including cell migration, 2005). Currently, at least three activities have been ascribed to
neuronal transport and cell division. It is now clear that different these proteins. First, MAP65s can selectively crosslink microtu-
cytoskeletal architectures arise from the interplay between bules in an antiparallel orientation (Gaillard et al., 2008; Loiodice
motor proteins, which can crosslink and move microtubules rela- et al., 2005). Second, these nonmotor crosslinkers can oppose
tive to one another, and nonmotor microtubule associated filament movements driven by motor proteins. For example,
proteins (MAPs), which can crosslink microtubules to stabilize Ase1, the fungal MAP65, is proposed to antagonize kinesin-14
specific orientations (Glotzer, 2009; Manning and Compton, driven filament sliding required for organizing microtubules
2008). While we have good biophysical and structural models during interphase (Janson et al., 2007). Third, these crosslinking
for motor proteins that crosslink microtubules, much less is proteins can recruit signaling proteins or kinesins to the microtu-
known about nonmotor microtubule crosslinking proteins. bule structures they stabilize. For example, the recruitment
Several nonmotor MAPs that crosslink microtubules (e.g., of Polo-like kinase to the central spindle during cytokinesis is
MAP65, NuMA, NuSAP and Mia1p) are now known to play mediated via interactions with PRC1, the human MAP65 (Neef

Cell 142, 433–443, August 6, 2010 ª2010 Elsevier Inc. 433


Figure 1. Single-Molecule Analysis of Microtubule Binding by PRC1
(A) Schematic of PRC10 s domain organization and a guide for constructs used
in the fluorescence microscopy assays (purple: coiled-coil domain; green:
microtubule binding domain; black: C-terminal domain).
(B) Fluorescence intensity analysis of two PRC1 constructs, GFP-PRC1-FL
(aa: 1–620; intensity = 2.5 3 104 ± 0.9 3 104, N = 469) and GFP-PRC1-NS
(aa: 1–466; intensity = 2.0 3 104 ± 0.8 3 104, N = 156). Dimeric-Eg5-GFP
(Intensity = 2.5x104 ± 1.0x104, N = 377) and tetrameric-Eg5-GFP (Intensity =
4.2x104 ± 2.2 3 104, N = 290) were used as references. Intensities are reported
as mean ± SD.
(C–H) Single molecule TIRF assay was used to examine the association of
PRC1 constructs (green) with microtubules (orange) immobilized on a glass
surface. (C) Schematic for assay showing the two constructs, GFP-PRC1-FL
and GFP-PRC1-NSDC (aa 1–486). Single frames showing two-color overlays
(top) and associated kymographs (below) of GFP-PRC1-FL (D and E) or
GFP-PRC1-NSDC (F and G). (H) Distribution of microtubule association life-
times for GFP-PRC1-FL (blue) and GFP-PRC1-NSDC (red).
(I–K) Microtubule association of GFP-PRC1-NSDC under different ionic
strength conditions. Representative kymographs from assays at 0.75x motility
buffer (I), motility buffer (J), motility buffer+20 mM KCl. (K) The scale bar repre-
sents 1.5 mm, 10 s. See also Figure S1 and Figure S3.

et al., 2007) and kinesin-5 driven microtubule sliding during


anaphase depends on Ase1 (Khmelinskii et al., 2009). Currently,
we do not have a structural framework to explain how these
MAPs specifically crosslink antiparallel microtubules. Moreover,
the activities of MAP65s have not been reconstituted in the pres-
ence of motor proteins to test if MAP65s resist filament sliding by
motor proteins or if their main function is to act as ‘‘marks’’ that
recruit other proteins to regions of antiparallel microtubule
overlap in dynamic networks.
Here we show, using single molecule fluorescence microscopy
assays, X-ray crystallography and electron microscopy that
PRC1 uses structured and unstructured domains to bind micro-
tubules. These domains at each end of a PRC1 homodimer are
connected by a linker that adopts a rigid conformation only when
crosslinking microtubules. We also show, in assays combining
TIRF and fluorescent speckle microscopy (FSM), that PRC1
does not substantially resist filament sliding by kinesin-5. Based
on these results, we propose a model for how a crosslinking MAP
can achieve specific and compliant crosslinking of microtubules
by balancing structural rigidity and flexibility.

RESULTS

Structured and Unstructured Domains Mediate


Microtubule Binding in PRC1
PRC1, like other Map65 family proteins, has a modular architec-
ture with an N-terminal coiled-coil domain, a central region that
can mediate microtubule binding, and a C-terminal regulatory
domain (Figure 1A). While the central domain is thought to
be required for microtubule binding, the contributions of the
C-terminal domain remain poorly characterized. To address
this, we analyzed the microtubule binding activity of PRC1 using
two approaches, a TIRF microscopy assay to examine the prop-
erties of single molecules and a microtubule cosedimentation
assay to analyze equilibrium binding.
For visualizing PRC1 molecules by fluorescence microscopy
we expressed and purified recombinant GFP-tagged full-length
PRC1 in bacteria. We found that C-terminal tags on PRC1

434 Cell 142, 433–443, August 6, 2010 ª2010 Elsevier Inc.


resulted in constructs that were highly unstable and therefore
used a construct with GFP fused to the N-terminus of PRC1
(GFP-PRC1-FL). We first examined the oligomerization state of
single GFP-PRC1-FL molecules. Analysis of the intensities of
single fluorescent spots of GFP-PRC1-FL immobilized on a glass
coverslip, when compared to intensities of known dimeric and
tetrameric reference constructs, indicated that GFP-PRC1-FL
is a dimer (Figure 1B), similar to the yeast homolog, Ase1 (Kapi-
tein et al., 2008a; Schuyler et al., 2003). We also confirmed that
untagged PRC1 is a homo-dimer and has an extended confor-
mation (see below, and data not shown). Previous studies have
suggested that PRC1 exists as a homo-tetramer, but gel filtration
chromatography alone can sometimes be unreliable for such
analyses of proteins with elongated structures (Zhu et al.,
2006). To examine the interaction of single PRC1 molecules
with a microtubule, we used streptavidin to attach X-rhoda-
mine-labeled, biotinylated microtubules to a glass surface. After
blocking the glass surface, we added low concentrations of
GFP-PRC1-FL (20 pM) (Figure 1C). Similar to what is observed
for Ase1, we found that single GFP-PRC1-FL molecules diffuse
in 1-D along the microtubule surface (Figure S1D and 1E; (Kapi-
tein et al., 2008a)). Analysis of time-lapse sequences using
kymographs showed that individual GFP-PRC1-FL molecules
can maintain associations with the microtubule lattice that can
last several seconds (t1/2 = 7 ± 0.4 s; Figure 1H).
To examine the contribution of the C terminus of PRC1 to
microtubule binding, we first generated a construct comprised
of just the N-terminal dimerization and the microtubule-binding
domain (aa 1–466, hereafter GFP-PRC1-NS). Intensity distribu-
tion of GFP-PRC1-NS indicated that this construct, like the
full-length protein, is a dimer (Figure 1B). We were unable to
detect microtubule associations of single molecules of GFP-
PRC1-NS at the concentrations needed to resolve single
molecules using TIRF microscopy (<10 nM; data not shown), Figure 2. Microtubule Cosedimentation Assays to Determine Equi-
suggesting that residues in the C terminus domain contribute librium Dissociation Constants for PRC10 s Microtubule-Binding
Domains
to microtubule binding. Secondary structure algorithms predict
(A) Schematic for constructs used in this assay: PRC1-S (aa 341–466) and
that the C-terminal domain in PRC1 is likely to be unstructured. PRC1-SC (aa 341–620). SDS-PAGE analysis of cosedimentation assays for
Consistent with this prediction, we find that PRC1’s C terminus PRC1-SC (B) and PRC1-S (C). Arrows in (B) indicate C terminus proteolysis
is prone to proteolysis. A highly susceptible cleavage site resides products in PRC1-SC. Bands marked with red boxes in (B) indicate the relative
between two Lys/Arg-rich basic clusters in this domain (aa. Asn tubulin concentration at which 50% of the different PRC1-SC truncation prod-
500; Figure S1 available online). As basic amino acids are often ucts cosediment with microtubules. (D) Band intensities from the gels in (C)
were used to determine fraction protein bound, and plotted against microtu-
implicated in mediating interactions with the negatively charged
bule concentration (n = 3, mean ± SD). The data were fit to a modified Hill equa-
microtubules surface, we generated a GFP-fused PRC1 con- tion to determine Kd’s (PRC1-S: 3.3 ± 1.8 mM; PRC1-SC: 0.6 ± 0.3 mM).
struct with one of the two basic-residue clusters (aa 1–486,
hereafter GFP-PRC1-NSDC). Under TIRF microscopy condi-
tions we could readily observe microtubule binding by single domain to PRC1’s microtubule interaction. For these experi-
GFP-PRC1-NSDC molecules. Diffusion on the microtubule was ments we generated PRC1 constructs lacking the dimerization
apparent for the subset of binding events that lasted several domain so that we could exclude the potentially complex effects
seconds (Figures 1F and 1G). Analysis of the life-times of micro- of filament crosslinking/bundling on this analysis (Figure 2A).
tubule association indicated that this construct has a higher These PRC1 constructs were confirmed to be monomeric by
microtubule unbinding rate than the full-length protein (Fig- size exclusion chromatography (data not shown). The microtu-
ure 1H, lifetime is less than the lower limit of reliable event bule binding affinity (Kd) of a construct comprising of the central
detection, < 3 s). Increasing salt concentrations reduced the domain and the C terminus (aa 341–620, named PRC1-SC) was
GFP-PRC1-NSDC binding lifetimes, consistent with a charge- 0.6 ± 0.3 mM (Figures 2B and 2D). A construct comprising of the
dependent microtubule-binding interaction mediated by central domain alone (aa 341–466, hereafter named PRC1-S)
PRC1’s C terminus (Figures 1I–1K). had a 5-fold weaker binding (Kd: 3.3 ± 1.8 mM; Figures 2C
We next carried out microtubule cosedimentation assays for and 2D). Interestingly, we observe cooperative microtubule
an independent analysis of the contribution of the C-terminal binding by PRC1-SC (Hill coefficient of 3 ± 1). This suggests

Cell 142, 433–443, August 6, 2010 ª2010 Elsevier Inc. 435


that the C terminus may be responsible for interactions between tubule cosedimentation assays to compare microtubule binding
PRC1 molecules on the microtubule lattice analogous to what of mutant and wild-type PRC1-S constructs (Figure 3G). We
has been previously reported for Ase1 (Kapitein et al., 2008a). found that mutation of each of the basic residues in the most
Together, these data indicate that PRC1’s microtubule binding conserved region reduced microtubule binding 2- to 4-fold,
is mediated by two regions, one that is predicted to be structured though none of the individual mutations abolished microtubule
and another, unstructured region, rich in Lys/Arg residues. binding (Figure 3H). To test that the observed microtubule
binding is not simply a nonspecific electrostatic interaction, we
PRC1’s Microtubule-Binding Domain Has a Spectrin Fold tested two other constructs. First, we made a C terminus trunca-
To determine the structural basis of microtubule binding by tion in PRC1-SC (labeled del453–466; aa 341–452) to remove
PRC1, we focused on its central microtubule-binding domain. three surface exposed lysines on helix-3. Second, we generated
As this domain has no obvious homology to any known microtu- a construct in which a lysine residue on helix-2 (K407), distal to
bule binding motif, we used X-ray crystallography to determine the potential microtubule binding site, was mutated to alanine.
its structure. Nonisomorphous crystals of the PRC1-S construct Both of these constructs show less than 50% reduction in
were obtained from both the native protein, which diffracted to microtubule binding. From this analysis, we propose that the
1.75 Å resolution, and the selenomethionyl-substituted protein, spectrin domain in PRC1 uses a basic surface comprising of
which diffracted to 2.0 Å resolution. The structure of the seleno- conserved residues at one end of the triple helix bundle to
methionyl-substituted protein was solved by single-wavelength mediate microtubule binding (Figure 3F, circle).
anomalous dispersion, refined, and, subsequently used as
a model to solve the structure of the native protein using molec- Spectrin Domain Fits with an Optimal Orientation
ular replacement (Table S1). We found that the microtubule- into the Cryo-EM Density Map of the PRC1-Microtubule
binding domain of PRC1 is an 70 Å long 3-helix bundle (labeled Complex
helix-1, -2, and -3), with connecting loops (labeled loop1-2 and To investigate the interaction of PRC1 with the microtubule
loop2-3) and N- and C- termini at opposite ends (Figure 3A lattice we examined the structure of dimeric PRC1 bound to
and Figures S2A and S2B). Residues from all three helices single microtubules by cryo-electron microscopy (Cryo-EM)
contribute to a hydrophobic core which dominates the interface and helical image analysis. As PRC1-FL extensively bundles
between the three helices (Figure 3B). This core is flanked on microtubules at concentrations needed for this analysis, we
both ends of the helix bundle by two salt bridges mediated by used our findings from single molecule experiments to engineer
conserved residues (Figure 3C). a construct PRC1-NSDC (aa 1–486; Figure 4A), which was
Using DALI, we found that PRC1-S has high structural similarity more suitable for this analysis (Figure 1A). This construct was
to spectrin domains (Figure 3D), motifs commonly found in confirmed to be dimeric by HPLC Size Exclusion Chromatog-
proteins associated with the actin cytoskeleton (e.g., alpha- raphy/Laser Light Scattering Analysis (Figure S3), and found to
actinin, spectrin and dystrophin; (Djinovic-Carugo et al., 2002)). retain sufficient binding to fully decorate single microtubules
However, a spectrin fold has not thus far been found in any other without extensive bundling.
known MAP. Interestingly, in actin-binding proteins, the elon- Diffraction of individual cryo-EM images of PRC1-NSDC
gated and rigid triple-helix of a spectrin domain does not directly bound to microtubules showed an 80 Å layer line, indicating
mediate actin binding, but acts as a spacer between canonical that one PRC1-NSDC molecule binds a a/b-tubulin heterodimer.
actin-binding motifs, such as calponin homology domains. The 3D reconstruction of microtubule bound PRC1-NSDC
Therefore, it appears that the spectrin fold has been evolutionarily showed a single rod-shaped density corresponding to PRC1-
‘recycled’ and re-engineered to act as a microtubule interacting NSDC, protruding approximately perpendicular to the microtu-
domain in the Map65 protein family. These findings establish bule lattice (microtubule side view, Figure 4B; top view,
a new role for the spectrin motif as a microtubule-binding domain. Figure 4C). Interestingly, the PRC1 density in the reconstruction
is of the same size as PRC1’s spectrin domain. The optimal fit of
A Conserved Basic Region Forms the Microtubule- the spectrin domain crystal structure to the reconstructions
Binding Surface in the Spectrin Domain of PRC1 oriented the conserved, basic residues at the junction of helix-1
As it was unclear how the spectrin fold would bind microtubules, and helix-2 toward the microtubule lattice (Figure 4D and 4E).
we analyzed the electrostatic surface potential and the conser- This binding model is also consistent with our site-directed
vation of surface residues for this domain. A map of the electro- mutagenesis analysis (Figure 3H). Comparison of the 3D recon-
static surface potential shows a positively charged region struction of this complex with cryo-EM structures of motor
comprising of loop1-2 and residues from all three helices that proteins (e.g kinesin and dynein’s microtubule-binding domain)
are proximal to this loop in the three dimensional structure bound to microtubules suggests that the binding site for PRC1
(Figure 3E). Interestingly, a cluster of highly conserved residues partially overlaps with, but is not identical to, the microtubule-
is also located within this region (Figure 3F), suggesting that binding surface used by motor proteins (Carter et al., 2008;
the junction of helix-1 and helix-2 could form the microtubule Figure S4B). Surprisingly, we see no extra density distal to the
binding region in the spectrin fold. To test this hypothesis we microtubule surface that would correspond to the N-terminus
generated constructs of PRC1’s spectrin domain in which basic coiled-coil dimerization domain in PRC1-NSDC. This indicates
residues in this region were mutated to alanine (Figure S2C). that the coiled-coil domain is likely to be flexible and can
Each of these constructs was characterized to be monomeric have more than one conformation when bound to a single
by size-exclusion chromatography (Figure S2D). We used micro- microtubule.

436 Cell 142, 433–443, August 6, 2010 ª2010 Elsevier Inc.


Figure 3. PRC10 s Conserved Microtubule-Binding Domain Adopts a Spectrin Fold
(A) Ribbon diagram shows the overall structure. Five disordered residues in the loop between helix-1 and 2 are indicated by dots.
(B) Hydrophobic residues from helix-1 (cyan), helix-2 (yellow) and helix-3 (green), which form the core of the triple helix bundle, are highlighted (sticks).
(C) Salt bridges between charged residues from helix-1 (cyan), helix-2 (yellow) and helix-3 (green) are indicated (spheres).
(D) Overlay of PRC10 s spectrin domain (orange) with its closest structural homolog, which is a spectrin repeat in Plectin (blue, PDB = 2odu-A, Z-score = 7.7,
rmsd = 3.4 Å calculated using DALI).
(E) Surface representation showing electrostatic potential of the spectrin domain in PRC1 (Red to blue is 10 kbT to +10 kbT, as calculated using APBS).
(F) Residues with low (cyan), intermediate (white), and high (magenta) conservation on the surface of the PRC10 s spectrin domain. The labeled residues were
selected for mutagenesis studies.
(G) SDS-PAGE analysis of microtubule cosedimentation assays of PRC1-S mutants at 27 mM tubulin.
(H) Band intensities from (G) were quantified to determine the fraction of PRC1 bound to microtubules (n = 3, error bars indicate SE). See also Figure S2.

To further understand the PRC1-microtubule interaction, we PRC1-NSDC. It is possible that this density corresponds to
obtained a second EM reconstruction with a monomeric PRC1’s C terminus folding back to interact with the spectrin
construct that lacked most of the oligomerization domain but domain or to the amino acids at PRC1’s N-terminus that may
contained the spectrin domain and the entire C terminus be ordered when this construct binds microtubules. As we do
(aa 303–620). The observed density for this construct overlaps not observe any additional density past the C terminus of the
with the PRC1-NSDC density close to the microtubule lattice spectrin domain that is close to the microtubule lattice, we favor
(Figures S4C–S4E). We also observe some additional density the possibility that the Lys/Arg-rich domain is disordered even
distal to the microtubule surface which is not observed in when PRC1 is bound to a microtubule. Together, these data

Cell 142, 433–443, August 6, 2010 ª2010 Elsevier Inc. 437


Figure 5. The Dimerization Domain in PRC1 Is Ordered When Cross-
linking Two Microtubules
Figure 4. The Spectrin Domain Fits into the Cryo-EM Density Map of (A) Schematic highlights the section viewed in the cryotomographic recon-
the PRC1-Microtubule Complex with an Optimal Orientation structions shown in (B) and (C), which encompasses the two microtubules
(A) Schematic comparing full-length PRC1 to the construct PRC1-NSDC and the PRC1 crosslinks.
(aa 1–486) used for Cryo-EM analysis. (B and C) Slices through cryotomographic 3D reconstructions of microtubules
(B) Surface rendered side-view of 3D EM density map of the microtubule- crosslinked by PRC1-NSDC. Slices represent the central area of the microtu-
PRC1-NSDC complex. bules and the bound PRC1-NSDC. The top and bottom of the microtubules are
(C) Top view of cryo-EM density map of undecorated tubulin (gold) superim- excluded in these views. Examples of microtubule pairs with dense (B) and
posed with the microtubule-PRC1-NSDC complex (purple). sparse (C) PRC1 occupancy. Insets show 4-fold enlargements of the cross-
(D and E) (D) Side- and (E) top-view of the crystal structure (cyan ribbon links between microtubules. Small red arrows in the inset highlight individual
diagram) docked into the PRC1 density (purple mesh) protruding from an cross-bridges. Blue arrow in (B) indicates a PRC1 molecule that does not
a/b tubulin dimer (gold). Residues R377 and K387, which are involved in micro- form crosslinks with another microtubule. Long red arrows in (B) indicate the
tubule binding, are indicated in red and black respectively. Microtubule polarity of microtubules, portions of which are highlighted in blue for clarity.
polarity indicated in (B) was determined by comparison with a 3D EM structure The top two microtubules in (B) share PRC1 crossbridges as do the bottom
of a dynein-microtubule complex, which has a well defined polarity (Carter two microtubules, however, the middle two microtubules are separated by
et al., 2008). See also Figure S4. another microtubule (dotted blue line) that is positioned below the plane and
only the top of its tubulin lattice can be observed.
show that PRC1’s spectrin domain is highly ordered and less
flexible than the other domains when associated with a single However, the conformational differences between crosslinking
microtubule. and noncrosslinking molecules could not be visualized in this
study. It also remains unknown whether establishing ordered
Cryo-Electron Tomography Reveals Ordered PRC1 crossbridges is a property of single MAP65 molecules or results
Crosslinks between Two Antiparallel Microtubules from protein-protein interactions that may be involved in PRC1’s
Helical reconstructions suggest that PRC1 may not be an inher- co-operative microtubule binding (Figure 2D). To address these
ently rigid molecule when bound to one microtubule. It is difficult questions, we employed cryo-electron tomography to obtain
to explain how a flexible protein with two microtubule interaction a higher resolution structure of two microtubules crosslinked
surfaces at opposite ends of dimer can achieve the crosslinking by PRC1 (Figure 5A). For this analysis we used the PRC1
specificity reported for MAP65 proteins (Gaillard et al., 2008; construct we had used for helical reconstruction (PRC1-NSDC;
Janson et al., 2007). The only other structural analysis of cross- Figure 4A). 3D reconstructions from cryo-electron tomography
linking by the MAP65 family is a cryo-EM study of the plant of microtubules densely crosslinked by PRC1-NSDC shows
homolog of PRC1, which show that the protein can form dense rod-like striations connecting two microtubules (Figure 5B
crossbridges between two microtubules (Gaillard et al., 2008). and inset). From analyzing the overall direction of crosslinking

438 Cell 142, 433–443, August 6, 2010 ª2010 Elsevier Inc.


protein density, we estimated that 90% (n = 20) of the cross- PRC1-FL shows a 10 ± 4 -fold preference for regions where
linked microtubules were antiparallel. This shows that the C two microtubule overlap relative to regions of single microtu-
terminus truncation construct has antiparallel crosslinking spec- bules (N = 15; Figures S5A–S5C; (Kapitein et al., 2008a)). To drive
ificity similar to full-length MAP65 and Ase1 (Gaillard et al., 2008; the relative sliding of microtubules in this ‘sandwich’, we added
Schuyler et al., 2003). We find that the average distance between kinesin-5, a well characterized motor protein needed for cell
two crosslinked filaments was 35 nm (±2 nm; n = 20) and the division.
crossbridge angle of PRC1 linkages was 70 (±5; n = 15) rela- Under these conditions we observed two types of events.
tive to the microtubule surface. As helical reconstruction shows First, kinesin-5 could move a shorter microtubule relative to
that the spectrin domain in PRC1 protrudes almost perpendic- a longer filament in the ‘sandwich’ such that the amount of
ular to the microtubule surface, the crossbridge orientation likely overlap tracked by GFP-PRC1, remained unchanged (Figures
results from 20 hinges between the oligomerization and the 6B–6D). Second, we observed events in which the microtubule
spectrin domains. overlap in the ‘sandwich’ reduced at the rate of filament motion
Consistent with what is seen from the helical reconstruction (Figure 6E and Figures S5D and S5E) and GFP-PRC1 dynami-
analysis, the tomograms show that the PRC1 densities observed cally tracked the microtubule overlap zone (Figure 6F). Remark-
on the microtubule surface distal to crosslinked microtubule (i.e., ably, in this set of events, the velocity of relative filament sliding
the noncrosslinking PRC1 molecules, Figure 5B, blue arrow) by kinesin-5 did not increase as the extent of microtubule over-
were less ordered than those between two filaments. This indi- lap reduced (Figures 6G and 6H). Fluorescence intensity analysis
cates that noncrosslinking and crosslinking PRC1 molecules indicated that the number of PRC1-crosslinks in the overlap
have distinct conformations. region decreased proportionately (data not shown). Analysis of
We next examined if the regular crossbridge geometry and the all data show that kinesin-5’s sliding velocity reduced only
intermicrotubule distances observed are retained in regions of 2.4 fold over an 18-fold change in PRC1 concentration
sparse PRC1 decoration between crosslinked microtubules. (Figure 6I). At higher PRC1 concentrations in this assay extensive
We find that single PRC1 molecules still form crossbridges microtubule bundling was observed and it was very difficult to
with orientations similar to those observed in regions of dense reliably detect pairs of crosslinked microtubules. The few events
crosslinks (Figure 5C and inset). Interestingly, we found that that could be analyzed under these conditions indicated that
PRC1 crosslinks were not observed in regions where filament a further reduction in kinesin-5 driven filament sliding was not
spacing was significantly greater than 35 nm, indicating that observed as PRC1 concentration was increased (Figure S5F-I).
PRC1 molecules exhibit specificity for a narrow range of inter- We estimated the ratio of PRC1 and kinesin-5 at microtubule
microtubule distances. These findings agree with our observa- overlap zones in our assays. Using GFP-kinesin-5 at concentra-
tions in fluorescence microscopy assays in which GFP-PRC1- tions similar to the un-tagged kinesin-5 used in the above
FL accumulation is seen only at a narrow range of microtubule assays, we found that microtubule sliding was driven by a few
crosslinking angles (unpublished data). Together, our data indi- motor protein molecules, and unlike PRC1, kinesin-5 did not
cate that although the dimerization domain is not entirely rigid show a preference for microtubule overlap zones (data not
when PRC1 is bound to a single microtubule, this domain in indi- shown and (Hentrich and Surrey, 2010)). The average fluores-
vidual PRC1 molecules can adopt a specific conformation upon cence intensity of single GFP-PRC1-FL molecules was then
crosslinking two microtubules. The rigidity in the linker, together used to determine the number of GFP-PRC1-FL molecules at
with oriented binding by the spectrin domain, can allow PRC1 to microtubule overlap regions. These data indicate that the ratio
be a selective crosslinker of antiparallel microtubules aligned of GFP-PRC1-FL to kinesin-5 molecules at microtubule overlap
with a narrow range of interfilament spacing. zones was 25 (total solution concentration: GFP-PRC1-FL =
0.54 nM, kinesin-5 = 1.8 nM). This shows that crosslinks formed
PRC1 Is a Compliant Crosslinker of Antiparallel by multiple PRC1 molecules do not substantially resist the rela-
Microtubules tive microtubule movement driven by fewer kinesin-5 molecules.
Our analysis of microtubule binding by the structured and
unstructured domains in PRC1 shows that these interactions Discussion
have moderate affinity. Together with the observed 1-D diffusion Members of the MAP65 family organize microtubules by prefer-
of single PRC1 molecules on microtubules, these data predict entially crosslinking filaments with an antiparallel orientation. In
that PRC1 crosslinks may not substantially resist filament sliding this study, we provide a structural framework for how three
by motor proteins. To test this hypothesis, we devised an in vitro structurally distinct domains in human MAP65 (PRC1) are
assay to visualize near-simultaneously PRC1 localization and combined to achieve selective and efficient crosslinking of anti-
motor-protein driven sliding of pairs of crosslinked microtubules. parallel microtubules, as would be needed during the self-orga-
We attached biotinylated microtubules to a glass surface via nization of dynamic cytoskeletal networks.
streptavidin. These microtubules also incorporated low levels The conserved microtubule-binding domain in PRC1 has
of fluorescent tubulin for analysis of relative microtubule motion a spectrin fold. First identified in spectrin, a constituent of the
using fluorescent speckle microscopy (FSM). GFP-PRC1-FL membrane skeleton, this fold has since been seen in several
was added to the immobilized microtubules, followed by nonbio- actin crosslinking proteins such as alpha-actinin and dystrophin
tinylated microtubules to generate a ‘sandwich’ comprised of (Djinovic-Carugo et al., 2002). In these proteins, spectrin-
two microtubules crosslinked by PRC1 (Figure 6A). As would domains appear as repeated units that connect other actin-
be expected, based on studies of Ase1, we find that GFP- binding domains and regulate properties of the cytoskeletal

Cell 142, 433–443, August 6, 2010 ª2010 Elsevier Inc. 439


structures. In PRC1, this domain appears to have evolved to
mediate microtubule binding, an entirely different role for this
protein fold. Further, the spectrin fold is unrelated to the other
protein folds (e.g Cap/Gly motifs and calponin-homology
domains) that are known to mediate interactions with the micro-
tubule lattice. It has been proposed that some protein surfaces
are evolutionarily selected to act as a versatile protein interaction
platform. This is best exemplified in the Fc domain of IgG, which
is structurally adapted to interact with several different protein
scaffolds (DeLano et al., 2000). Similarly, it appears that the
microtubule surface has evolved to bind a large variety of unre-
lated structural motifs and thereby accommodate diverse
MAPs that can carry out a wide range of functions (Amos and
Schlieper, 2005).
Microtubule binding by the spectrin domain is augmented by
a Lys/Arg rich unstructured domain in PRC1. Synergistic microtu-
bule binding by structured and unstructured domains have also
been seen in other MAPs such as Ndc80 (Guimaraes et al.,
2008; Miller et al., 2008), suggesting that this feature may be
a frequent adaptation in MAPs. There are also at least two other
functions ascribed to these unstructured microtubule-binding
domains. First, these unstructured domains are often sites of
phospho-regulation (Holt et al., 2009). Interestingly, Cdk1 phos-
phorylation sites in PRC1 map to the unstructured Lys/Arg
domain (Zhu et al., 2006). Our data suggest that these phosphor-
ylations would directly attenuate microtubule affinity by reducing
the net positive charge of the domain. Dephosphorylation of these
residues would activate PRC1’s microtubule binding at
anaphase, as would be needed for PRC1’s functions during the
final stages of cell division. Second, as has been suggested for
Ndc80, these domains are proposed to allow a mode of attach-
ment which does not significantly resist microtubule movement.
We find that PRC1 does not strongly oppose microtubule sliding
by kinesin-5, indicating that moderate binding affinities and diffu-
sive microtubule interactions of PRC1 can permit microtubule
movement while maintaining attachment. This result is also
consistent with the reported diffusion constant of Ase1 which indi-
cates that > 100 crosslinker molecules would be needed to
generate a resistive force greater than 1.5 pN, which is in the range
of the force required to inhibit kinesin-5 movement (Bormuth et al.,
2009; Kapitein et al., 2008a; Korneev et al., 2007; Valentine and
Block, 2009). This would be relevant in vivo, when microtubule
bundling by Ase1 and sliding by kinesin-5 are both required for
spindle elongation in anaphase B (Khmelinskii et al., 2009).
Crossbridges between two PRC1-crosslinked microtubules
are formed by PRC1’s dimerization domain. These are seen to
Figure 6. PRC1 Crosslinks Do Not Substantially Resist the Relative project at an angle of 70 relative to the microtubule lattice.
Sliding of Two Microtubules by Kinesin-5
Binding at a fixed angle relative to the microtubule lattice has
(A) Schematic illustrating the assay used to examine the effect of PRC1 (green)
on kinesin-5 (cyan)-mediated relative sliding of two microtubules (orange),
when extent of overlap between microtubules is unchanged. Near-simulta- (E) Schematic illustrating the assay when overlap between microtubules
neous dual-mode microscopy was used to image microtubules (via wide-field decreases during relative microtubule sliding.
fluorescent speckle microscopy) and GFP-PRC1-FL (via Total Internal Reflec- (F–H) Frames from a time-lapse sequence (2 min interval) (F) and corresponding
tion Fluorescence (TIRF) microscopy). kymographs showing microtubule movement (7 nm/s) (G) and GFP-PRC1-FL
(B) Frames from a time-lapse sequence (1 min interval) show GFP-PRC1-FL localization to overlap region that reduces due to relative sliding of filaments (H).
(green) enriched at regions where two crosslinked microtubules (red) overlap. (I) Velocity distributions for kinesin-5 driven microtubule sliding at 1.8 nM
(C) Corresponding kymograph shows the surface-attached static microtubule kinesin-5 and 0.034 (V = 23.1 ± 6.3 nm/s, N = 43), 0.14 nM (V = 17.7 ±
(vertical streaks) and a moving microtubule (diagonal streaks, 16.5 nm/s). 3.5 nm/s, N = 39), and 0.54 nM GFP-PRC1-FL (V = 9.7 ± 3.1 nm/s, N = 41).
(D) Kymograph shows that the GFP-PRC1-FL decorated region moves at the Velocities are reported as mean ± SD. The scale bars represent 1.5 mm,
velocity of the moving microtubule. 100 s. See also Figure S5.

440 Cell 142, 433–443, August 6, 2010 ª2010 Elsevier Inc.


bound to one microtubule. Our results show that the spectrin
domain is the most ordered region in a PRC1-microtubule com-
plex and uses a well-defined surface for microtubule binding.
This suggests that this domain makes contacts with the microtu-
bule lattice that decode filament orientation. Second, the micro-
tubule polarity needs to be transmitted across the linker to
the second spectrin domain in the PRC1 homodimer. Our data
show that the dimerization domain in PRC1 has a single confor-
mation when crosslinking two microtubules. The structural
rigidity of this domain is likely to be responsible for PRC1’s selec-
tivity for antiparallel microtubules. Though specificity can be
achieved by oriented binding of the spectrin domain and rigidity
in the linker domain, the weak microtubule binding affinity of the
spectrin domain alone does not explain the extensive filament
bundling induced by PRC1. To increase binding and conse-
quently crosslinking efficiency, PRC1 uses an unstructured posi-
tively charged domain for maintaining long-lived associations
with microtubules. Additionally, the flexibility in the linker domain
of PRC1, which appears to have more than one conformation
when bound to one microtubule, may allow for an initial contact
with a second microtubule that could have a wide-range of
orientations. Relative to a highly rigid structure, such flexibility
Figure 7. PRC1 Is a Compliant, Microtubule-Overlap Tracking could increase the crosslinking efficiency of PRC1 molecules.
Protein that Tunes Structural Rigidity to Specifically Crosslink Two These features would enable PRC1 to stay associated with the
Antiparallel Microtubules
first microtubule encountered, explore its length by 1-D diffu-
(A) A model for how PRC1 can align microtubules into antiparallel arrays. The
spectrin domain in PRC1 can make oriented contacts with the microtubule to sion, and thereby increase the probability of capturing a second
decode filament polarity. The unstructured domain acts to enhance the filament for establishing antiparallel linkages between two
binding affinity, while allowing diffusion along microtubules. The dimerization microtubules.
domain is not entirely rigid on a single microtubule but adopts a specific Many cellular processes require recognition of a ‘‘mark’’ at
conformation when crosslinking two microtubules. precise locations. Post-translational modifications such as ubiq-
(B) The rigidity and flexibility of the different domains in PRC1 can facilitate
uitination and methylation are some of the common marking
sorting of randomly oriented microtubules into antiparallel arrays and allow
PRC1 to function as a selective ‘mark’ for microtubule overlap regions.
mechanisms for proteins and DNA in cells. In the microtubule
cytoskeleton, the +TIP-proteins track and identify microtubule
growing microtubule plus-ends (Akhmanova and Steinmetz,
also been reported for Ndc80 (Wilson-Kubalek et al., 2008) but 2008). Similarly, by recognizing specific microtubule geometries
the implications of defined projection angles for any microtu- and forming compliant crosslinks, the MAP65 proteins can mark
bule-binding protein is thus far unknown. The crossbridge also antiparallel microtubule overlap during the self-organization of
determines the intermicrotubule spacing between two microtu- dynamic cytoskeletal networks.
bules. This intermicrotubule distance could also affect the ability
of motor proteins to bind and slide PRC1 crosslinked microtu- EXPERIMENTAL PROCEDURES
bules, providing an additional mechanism for activating or
deactivating specific motors at these structures. For example, Crystallization of PRC1-S
the reported length of kinesin-5 motor is approximately 95 nm, PRC1-S (341-466) was concentrated to 50 mg/ml in motility buffer (80 mM
which is 2-fold greater than the 37 nm intermicrotubule spacing PIPES (pH 6.8), 1 mM MgCl2, 1 mM EGTA) and 150 mM KCl and crystallized
by hanging drop vapor diffusion at 4 C using an equal volume of protein
of PRC1 crosslinked microtubules (Kashina et al., 1996). Hence,
sample and crystallization solution consisting of 0.1 M CHES (pH 9.5) and
the reduction in the velocity of microtubule sliding by kinesin-5 30%–35% PEG 3350. Crystals (needles of dimension 25 X 25 X 300 mm)
seen in our experiments at high PRC1 concentrations could appeared between 1 - 3 weeks. Microseeding was used to obtain crystals of
result from the inability of kinesin-5 to bind properly and the selenomethionyl derivative of the construct. Crystals were frozen in liquid
efficiently walk along both microtubules with dense PRC1 cross- nitrogen after a 5 min soak in a solution comprising of the crystallization solu-
links. Such modulation of motor activity through control of tion with 5%–10% v/v glycerol. Both native and SAD datasets were collected
along single needles at NE-CAT beamline 24ID-E at APS at the peak selenium
interfilament spacing has been proposed to affect the magnitude
wavelength 0.97918 Å and processed as described in the supplement.
of active forces generated in striated muscles during muscle
contraction (Millman, 1998).
Based on our results, we propose a structural model for how Cryo-EM and Image Analysis
Microtubules were polymerized as previously described (Wilson-Kubalek
polarity-specific crosslinking is mediated by PRC1 (Figures 7A
et al., 2008). Microtubules (0.166 mg/ml in motility buffer) were applied to
and 7B). The ability of a microtubule associated protein to distin- plasma cleaned C-flat grids and incubated with PRC1-NSDC (0.53 mg/ml) or
guish parallel and antiparallel filaments relies on two factors. PRC1-SC’ (0.9 mg/ml). The sample was then vitrified in liquid ethane, using
First, PRC1 molecules must decode filament polarity when a manual plunger. The data sets were collected using a GATAN cryoholder

Cell 142, 433–443, August 6, 2010 ª2010 Elsevier Inc. 441


and a FEI Tecnai F20 transmission electron microscope equipped with a Gatan ACKNOWLEDGMENTS
Ultrascan 4000 3 4000 pixel CCD camera. Images of the decorated microtu-
bules were recorded in low-dose conditions (<10 e/Å) at a magnification of We thank K. Rajashankar (Argonne Advanced Photon Source) and Deena Oren
29,000 and a nominal defocus range from 1.5 to 2.0 mm. For image analysis (Rockefeller University Structural Biology Resource Center (SBRC)) for tech-
of the PRC1-microtubule complexes, PRC1-decorated 15-protofilament nical assistance. T.M.K. is grateful to the NIH (GM65933 and GM65933-S1)
microtubules were selected from CCD images. 3D reconstructions were calcu- for support. R.S. is a recipient of the Rockefeller Women and Science postdoc-
lated using Phoelix, essentially as described elsewhere (Whittaker et al., 1995). toral fellowship. R.A.M. acknowledges support from the NIH (GM52468). We
also acknowledge the National Resource for Automated Molecular Micros-
3D EM Reconstructions copy (NIH RR17573), the SBRC (NIH 1S10RR022321-01), and the Keck
Surface representations of side- and top- views of the cryo-EM reconstruc- Facility at Yale University (NIH 1S10RR023748-01) for instrument use.
tions (Figure 4 and S4) were produced using the Chimera software package
(Pettersen et al., 2004). To interpret the PRC1 densities protruding from the Received: May 10, 2010
microtubule density, we manually docked the crystal structure of the spectrin Revised: June 21, 2010
domain into the EM density reconstructions using Chimera. Accepted: July 6, 2010
Published: August 5, 2010
Cryotomography
Tilt-series images were acquired using the Serial EM software package (Mas- REFERENCES
tronarde, 2005) on a Tecnai F20 microscope through a range of +/ 50 at 1.5
increments. The specimen was subjected to a total dose of 100 electrons. Akhmanova, A., and Steinmetz, M.O. (2008). Tracking the ends: a dynamic
The tomographic image reconstruction was performed using the IMOD soft- protein network controls the fate of microtubule tips. Nat. Rev. Mol. Cell
ware package (Kremer et al., 1996). Biol. 9, 309–322.
Amos, L.A., and Schlieper, D. (2005). Microtubules and maps. Adv. Protein
Fluorescence Microscopy
Chem. 71, 257–298.
All experiments were performed on an instrument described previously (Kapi-
tein et al., 2008b). GFP was imaged with TIRF illumination from a 491-nm laser Bormuth, V., Varga, V., Howard, J., and Schaffer, E. (2009). Protein friction
source (Cobalt Calypso 50; Solamere Technology) and X-rhodamine was limits diffusive and directed movements of kinesin motors on microtubules.
imaged with wide-field illumination. For all experiments, oxygen scavenging Science 325, 870–873.
mix (OS) comprising of 25 mM glucose, 40 mg/ml glucose oxidase, 35 mg/ml Carter, A.P., Garbarino, J.E., Wilson-Kubalek, E.M., Shipley, W.E., Cho, C.,
catalase and 0.5% beta-mercaptoethanol was included in the final buffer. Milligan, R.A., Vale, R.D., and Gibbons, I.R. (2008). Structure and functional
Kinesin-5/PRC1 sliding experiments were performed as described previ- role of dynein’s microtubule-binding domain. Science 322, 1691–1695.
ously with some modifications (Kapitein et al., 2008b). Briefly, X-rhodamine-
Chan, J., Jensen, C.G., Jensen, L.C., Bush, M., and Lloyd, C.W. (1999). The
labeled biotinylated taxol-stabilized microtubules were immobilized on the
65-kDa carrot microtubule-associated protein forms regularly arranged fila-
coverslip by first coating the surface with biotinylated BSA followed by strepta-
mentous cross-bridges between microtubules. Proc. Natl. Acad. Sci. USA
vidin. After a brief incubation with casein to block nonspecific binding to
96, 14931–14936.
the surface, GFP-PRC1-FL was added and allowed to bind the immobilized
microtubules. This was followed by addition of X-rhodamine-labeled microtu- Ciferri, C., Pasqualato, S., Screpanti, E., Varetti, G., Santaguida, S., Dos Reis,
bules which generated a ‘sandwich’ with two microtubules crosslinked by G., Maiolica, A., Polka, J., De Luca, J.G., De Wulf, P., et al. (2008). Implications
PRC1. After washing the solution microtubules, the final assay mix comprising for kinetochore-microtubule attachment from the structure of an engineered
of GFP-PRC1-FL and/or kinesin-5 in motility buffer supplemented with Ndc80 complex. Cell 133, 427–439.
80 mM KCl, 2 mM MgCl2, 1 mg/mL k-casein, 1 mM MgATP, 20 mM taxol and DeLano, W.L., Ultsch, M.H., de Vos, A.M., and Wells, J.A. (2000). Convergent
OS mix was added to the flow chamber. TIRF and wide-field images were solutions to binding at a protein-protein interface. Science 287, 1279–1283.
acquired near simultaneously with 0.2 s exposure, 0.1 s-1 frame rate and EM
Djinovic-Carugo, K., Gautel, M., Ylanne, J., and Young, P. (2002). The spectrin
gain set to 100 and 200 for TIRF and wide-field respectively. Each pair of moving
repeat: a structural platform for cytoskeletal protein assemblies. FEBS Lett.
microtubule was analyzed by kymographs using MetaMorph (MDS Analytical
513, 119–123.
Technologies). Sliding velocities were calculated from the slopes of the diag-
onal streaks generated by the movement of one speckled microtubule over Gaillard, J., Neumann, E., Van Damme, D., Stoppin-Mellet, V., Ebel, C.,
an immobilized microtubule. In a few rare events (<5%), we observed an abrupt Barbier, E., Geelen, D., and Vantard, M. (2008). Two microtubule-associated
2-fold change in microtubule movement velocity in the kymographs. Based proteins of Arabidopsis MAP65s promote antiparallel microtubule bundling.
on our analyses of kinesin-5 sliding alone (without PRC1), we believe this is Mol. Biol. Cell 19, 4534–4544.
due to kinesin-5 switching between walking on both microtubules it crosslinks Glotzer, M. (2009). The 3Ms of central spindle assembly: microtubules, motors
to walking on only one of the two microtubules. This change in motility mode and MAPs. Nat. Rev. Mol. Cell Biol. 10, 9–20.
leads to a 2-fold reduction in relative filament sliding. In these cases, both Guimaraes, G.J., Dong, Y., McEwen, B.F., and Deluca, J.G. (2008). Kineto-
velocities were measured and are included in the histograms shown. chore-microtubule attachment relies on the disordered N-terminal tail domain
of Hec1. Curr. Biol. 18, 1778–1784.
ACCESSION NUMBERS Hentrich, C., and Surrey, T. (2010). Microtubule organization by the antago-
nistic mitotic motors kinesin-5 and kinesin-14. J. Cell Biol. 189, 465–480.
Coordinates have been deposited with PDB IDs 3NRX (native crystal structure)
and 3NRY (SelenoMet crystal structure) and EMDB accession codes Holt, L.J., Tuch, B.B., Villen, J., Johnson, A.D., Gygi, S.P., and Morgan, D.O.
EMD-5205 (EM map of PRC1 [aa 1-486]) and EMD-5212 (EM map of PRC1 (2009). Global analysis of Cdk1 substrate phosphorylation sites provides
[aa 303-620]). insights into evolution. Science 325, 1682–1686.
Janson, M.E., Loughlin, R., Loiodice, I., Fu, C., Brunner, D., Nedelec, F.J., and
SUPPLEMENTAL INFORMATION Tran, P.T. (2007). Crosslinkers and motors organize dynamic microtubules to
form stable bipolar arrays in fission yeast. Cell 128, 357–368.
Supplemental Information includes Extended Experimental Procedures, five Jiang, W., Jimenez, G., Wells, N.J., Hope, T.J., Wahl, G.M., Hunter, T., and
figures, and one table and can be found with this article online at doi: Fukunaga, R. (1998). PRC1: a human mitotic spindle-associated CDK
10.1016/j.cell.2010.07.012. substrate protein required for cytokinesis. Mol. Cell 2, 877–885.

442 Cell 142, 433–443, August 6, 2010 ª2010 Elsevier Inc.


Kapitein, L.C., Janson, M.E., van den Wildenberg, S.M., Hoogenraad, C.C., NuSAP, a mitotic RanGTP target that stabilizes and cross-links microtubules.
Schmidt, C.F., and Peterman, E.J. (2008a). Microtubule-driven multimerization Mol. Biol. Cell 17, 2646–2660.
recruits ase1p onto overlapping microtubules. Curr. Biol. 18, 1713–1717. Sasabe, M., and Machida, Y. (2006). MAP65: a bridge linking a MAP kinase to
Kapitein, L.C., Kwok, B.H., Weinger, J.S., Schmidt, C.F., Kapoor, T.M., and microtubule turnover. Curr. Opin. Plant Biol. 9, 563–570.
Peterman, E.J. (2008b). Microtubule cross-linking triggers the directional Schuyler, S.C., Liu, J.Y., and Pellman, D. (2003). The molecular function of
motility of kinesin-5. J. Cell Biol. 182, 421–428. Ase1p: evidence for a MAP-dependent midzone-specific spindle matrix.
Kashina, A.S., Baskin, R.J., Cole, D.G., Wedaman, K.P., Saxton, W.M., and Microtubule-associated proteins. J. Cell Biol. 160, 517–528.
Scholey, J.M. (1996). A bipolar kinesin. Nature 379, 270–272.
Slep, K.C., and Vale, R.D. (2007). Structural basis of microtubule plus end
Khmelinskii, A., Roostalu, J., Roque, H., Antony, C., and Schiebel, E. (2009). tracking by XMAP215, CLIP-170, and EB1. Mol. Cell 27, 976–991.
Phosphorylation-dependent protein interactions at the spindle midzone
Thadani, R., Ling, Y.C., and Oliferenko, S. (2009). The fission yeast TACC
mediate cell cycle regulation of spindle elongation. Dev. Cell 17, 244–256.
protein Mia1p stabilizes microtubule arrays by length-independent crosslink-
Korneev, M.J., Lakamper, S., and Schmidt, C.F. (2007). Load-dependent ing. Curr. Biol. 19, 1861–1868.
release limits the processive stepping of the tetrameric Eg5 motor. Eur.
Valentine, M.T., and Block, S.M. (2009). Force and premature binding of ADP
Biophys. J. 36, 675–681.
can regulate the processivity of individual Eg5 dimers. Biophys. J. 97,
Kremer, J.R., Mastronarde, D.N., and McIntosh, J.R. (1996). Computer visual- 1671–1677.
ization of three-dimensional image data using IMOD. J. Struct. Biol. 116,
71–76. Wei, R.R., Al-Bassam, J., and Harrison, S.C. (2007). The Ndc80/HEC1
complex is a contact point for kinetochore-microtubule attachment. Nat.
Loiodice, I., Staub, J., Setty, T.G., Nguyen, N.P., Paoletti, A., and Tran, P.T.
Struct. Mol. Biol. 14, 54–59.
(2005). Ase1p organizes antiparallel microtubule arrays during interphase
and mitosis in fission yeast. Mol. Biol. Cell 16, 1756–1768. Whittaker, M., Carragher, B.O., and Milligan, R.A. (1995). PHOELIX: a package
for semi-automated helical reconstruction. Ultramicroscopy 58, 245–259.
Manning, A.L., and Compton, D.A. (2008). Structural and regulatory roles of
nonmotor spindle proteins. Curr. Opin. Cell Biol. 20, 101–106. Wilson-Kubalek, E.M., Cheeseman, I.M., Yoshioka, C., Desai, A., and Milligan,
R.A. (2008). Orientation and structure of the Ndc80 complex on the microtu-
Mastronarde, D.N. (2005). Automated electron microscope tomography using
bule lattice. J. Cell Biol. 182, 1055–1061.
robust prediction of specimen movements. J. Struct. Biol. 152, 36–51.
Miller, S.A., Johnson, M.L., and Stukenberg, P.T. (2008). Kinetochore attach- Yamashita, A., Sato, M., Fujita, A., Yamamoto, M., and Toda, T. (2005). The
ments require an interaction between unstructured tails on microtubules and roles of fission yeast ase1 in mitotic cell division, meiotic nuclear oscillation,
Ndc80(Hec1). Curr. Biol. 18, 1785–1791. and cytokinesis checkpoint signaling. Mol. Biol. Cell 16, 1378–1395.

Millman, B.M. (1998). The filament lattice of striated muscle. Physiol. Rev. 78, Zeng, C. (2000). NuMA: a nuclear protein involved in mitotic centrosome
359–391. function. Microsc. Res. Tech. 49, 467–477.

Neef, R., Gruneberg, U., Kopajtich, R., Li, X., Nigg, E.A., Sillje, H., and Barr, F.A. Zhu, C., Lau, E., Schwarzenbacher, R., Bossy-Wetzel, E., and Jiang, W. (2006).
(2007). Choice of Plk1 docking partners during mitosis and cytokinesis is Spatiotemporal control of spindle midzone formation by PRC1 in human cells.
controlled by the activation state of Cdk1. Nat. Cell Biol. 9, 436–444. Proc. Natl. Acad. Sci. USA 103, 6196–6201.
Pettersen, E.F., Goddard, T.D., Huang, C.C., Couch, G.S., Greenblatt, D.M.,
Meng, E.C., and Ferrin, T.E. (2004). UCSF Chimera–a visualization system Note Added in Proof
for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612. In a manuscript by Bieling et al., which appears in this issue of Cell, the authors
Ribbeck, K., Groen, A.C., Santarella, R., Bohnsack, M.T., Raemaekers, T., arrive at similar conclusions regarding the nature of microtubule crosslinks
Kocher, T., Gentzel, M., Gorlich, D., Wilm, M., Carmeliet, G., et al. (2006). formed by PRC1.

Cell 142, 433–443, August 6, 2010 ª2010 Elsevier Inc. 443


Aurora Kinases and Protein Phosphatase 1
Mediate Chromosome Congression
through Regulation of CENP-E
Yumi Kim,1,2,4 Andrew J. Holland,1,4 Weijie Lan,1 and Don W. Cleveland1,3,*
1Ludwig Institute for Cancer Research
2Department of Molecular and Cellular Biology, University of California, Berkeley, CA 94720, USA
3Department of Cellular and Molecular Medicine

University of California, San Diego, La Jolla, CA 92093, USA


4These authors contributed equally to this work

*Correspondence: dcleveland@ucsd.edu
DOI 10.1016/j.cell.2010.06.039

SUMMARY ward motion (Rieder and Alexander, 1990). Although some


chromosomes achieve biorientation without being transported
Opposing roles of Aurora kinases and protein phos- to the spindle pole, dynein-mediated transport is an important
phatase 1 (PP1) during mitosis have long been sug- mechanism to collect chromosomes to a common microtu-
gested. Here, we demonstrate that Aurora kinases bule-dense region, where kinetochores have a greater chance
A and B phosphorylate a conserved residue on the of promoting efficient chromosome alignment.
kinetochore motor CENP-E. PP1 binds CENP-E via Congression of polar-localized chromosomes to a metaphase
position is powered by a processive, plus end-directed kineto-
a motif overlapping this phosphorylation site and
chore motor CENP-E (Kinesin-7) (Kapoor et al., 2006; Kim
binding is disrupted by Aurora phosphorylation. et al., 2008). In various cell types and organisms, removal or
Phosphorylation of CENP-E by the Auroras is en- inhibition of CENP-E leads to a failure in complete metaphase
riched at spindle poles, disrupting binding of PP1 chromosome alignment, with a few unattached chromosomes
and reducing CENP-E’s affinity for individual micro- found close to the spindle poles (Putkey et al., 2002; Wood
tubules. This phosphorylation is required for CENP- et al., 1997; Yao et al., 2000; Yucel et al., 2000). Even the kinet-
E-mediated towing of initially polar chromosomes ochores that do become bioriented and fully aligned in the
toward the cell center. Kinetochores on such chro- absence of CENP-E stably bind only half as many microtubules
mosomes cannot make subsequent stable attach- (Putkey et al., 2002). Our finding that CENP-E possesses a highly
ment to spindle microtubules when dephosphoryla- flexible and very long coiled-coil (Kim et al., 2008) raises the
tion of CENP-E or rebinding of PP1 to CENP-E is possibility that, while it can work advantageously for initial
capture, CENP-E may also contribute, in part, to the inappro-
blocked. Thus, an Aurora/PP1 phosphorylation
priate attachments of kinetochores. Indeed, the process of
switch modulates CENP-E motor activity as an capturing spindle microtubules by kinetochores is prone to
essential feature of chromosome congression from errors. Undesirable attachment frequently occurs in early prom-
poles and localized PP1 delivery by CENP-E to the etaphase, with a single kinetochore capturing microtubules from
outer kinetochore is necessary for stable microtu- both spindle poles (merotelic attachment), or both sister kineto-
bule capture by those chromosomes. chores attached to the same pole (syntelic attachment) (Cimini
and Degrassi, 2005). These improper kinetochore attachments,
INTRODUCTION if not resolved, can lead to chromosome missegregation and
aneuploidy (Holland and Cleveland, 2009).
Accurate chromosome segregation during mitosis requires the Correction of aberrant kinetochore attachment requires
bipolar attachment of duplicated chromosomes to spindle a conserved Ser/Thr kinase Aurora/Ipl1 (Lampson et al., 2004;
microtubules emanating from opposite poles. Each time a cell Tanaka et al., 2002). While budding yeast has a single Aurora
divides, a specialized proteinaceous structure called the kineto- kinase Ipl1, metazoans express at least two Aurora kinases,
chore assembles on the surface of each centromere, and it is Aurora A and B. Like Ipl1, Aurora B is a component of the chromo-
the kinetochore that binds to spindle microtubules and directs some passenger complex (together with INCENP, Survivin, and
chromosome motion during mitosis (Cleveland et al., 2003). Borealin/Dasra) and is targeted to the inner centromere from
Microtubule capture by the kinetochore is a stochastic process. prophase to metaphase (Ruchaud et al., 2007). Aurora B is
Initial kinetochore attachment is often mediated via an interac- thought to aid chromosome biorientation by destabilizing the
tion with the lateral surface of a microtubule, and kinetochores kinetochore-microtubule interaction of improperly attached
attached in this manner undergo rapid, dynein-mediated pole- chromosomes (Cimini et al., 2006). Several proteins directly

444 Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc.


involved in microtubule capture at the kinetochore, including conserved stretch of basic residues downstream of the CENP-E
Dam1 in budding yeast and the core kinetochore microtubule coiled-coil neck (Figure 1A). Consisting of four or more consec-
binding components in metazoans (Ndc80 and KNL1), are known utive arginines or lysines, this basic stretch and the following
Aurora B substrates (Cheeseman et al., 2002, 2006; Gestaut threonine (422 in human and 424 in Xenopus laevis) are
et al., 2008; Liu et al., 2010), and phosphorylation by Aurora B conserved in almost all the eukaryotes that possess a clear
has been shown to decrease the affinity of these proteins for CENP-E homolog. Interestingly, the conserved threonine resides
microtubules (Cheeseman et al., 2006; Gestaut et al., 2008). in a consensus motif for phosphorylation by Aurora kinase
Despite the high sequence similarity with Aurora B, Aurora A (Cheeseman et al., 2002) and has been previously mapped as
plays distinct roles during mitosis. Localized to the centrosomes a phosphorylation site in a mass spectrometry-based proteomic
during interphase and at the spindle poles during mitosis, Aurora screen of mitotic spindles (Nousiainen et al., 2006).
A has been implicated in promoting mitotic entry and is required To test whether CENP-E T422 (424 in Xenopus laevis) is phos-
for centrosome maturation and separation (Marumoto et al., phorylated by Aurora kinases, we performed in vitro kinase
2005). Inhibition of Aurora A has also been reported to cause assays using purified Aurora kinases and portions of Xenopus
chromosome congression defects (Hoar et al., 2007; Kunitoku CENP-E as a substrate. Xenopus Aurora B, together with its
et al., 2003; Marumoto et al., 2003); however, how Aurora A activator INCENP, phosphorylated both full-length and a motor
acts to promote chromosome alignment is unknown. fragment of CENP-E (CENP-E1-473). However, Aurora B failed
Genetic evidence in yeast (Francisco et al., 1994) and in verte- to phosphorylate CENP-E1-473 in which threonine 424 had
brates (Emanuele et al., 2008; Liu et al., 2010) suggest that the been converted to alanine (Figure 1B). Xenopus CENP-E
Aurora kinase activity is opposed by the ubiquitous Ser/Thr phos- T424 was also readily phosphorylated by Aurora A (Figures
phatase, protein phosphatase 1 (PP1/Glc7). In vertebrates, PP1 S1A and S1B available online), confirming that the conserved
isoforms a and g can be detected at outer kinetochores (Trinkle- threonine located close to the CENP-E motor domain is phos-
Mulcahy et al., 2006; Trinkle-Mulcahy et al., 2003), and PP1 has phorylated by both Aurora A and B in vitro. The stoichiometry
been shown to stabilize kinetochore-microtubule attachment by of CENP-E1-473 phosphorylation by Aurora A saturated at two
counteracting Aurora B kinase activity (Liu et al., 2010; Sassoon moles of PO4 per mole of CENP-E1-473, most likely with an
et al., 1999). Recently, the non-essential yeast protein Fin1 and additional phosphorylation site located C-terminal to T424, as
conserved kinetochore protein KNL1 have been identified to target a shorter CENP-E1-415 fragment was not phosphorylated by
some PP1 to yeast and vertebrate kinetochores, respectively either Aurora kinase (Figure S1B).
(Akiyoshi et al., 2009; Liu et al., 2010). However, whether the kinet-
ochore possesses multiple docking modules for PP1 is not known. Aurora A and B Contribute to Phosphorylation
Phosphorylation of the C-terminal tail of CENP-E by Cdk1, of CENP-E T422 in Cells
MAPK, or Mps1 has been previously proposed either to regulate To examine the phosphorylation of CENP-E T422 in vivo, a rabbit
CENP-E motor activity prior to its binding to kinetochores (Espeut polyclonal antibody was generated against a phosphopeptide of
et al., 2008) or inhibit a microtubule binding site in the tail that may human CENP-E surrounding T422 (Figure S1C). The affinity
link anti-parallel, midzone microtubules in anaphase (Liao et al., purified anti-pT422 antibody recognized recombinant human
1994; Zecevic et al., 1998). Additionally, among a proteomic CENP-E1-429 only in the presence of active kinase (Figure 1C)
screen of 260 mitotic phosphoproteins, CENP-E was identified and recognition of phosphorylated Xenopus CENP-E1-428 by
to be multiply phosphorylated during mitosis (Nousiainen et al., the anti-pT422 antibody was abolished by the mutation T424A
2006). However, the significance of the phosphorylations of (Figure S1D). The anti-pT422 antibody also recognized wild-
CENP-E has not been established. Using purified components, type (WT) CENP-E immunoprecipitated from nocodazole-
selective inhibitors and a phosphospecific antibody, here we arrested human (DLD-1) cells, but not CENP-E containing a
demonstrate that Aurora kinases, both A and B, phosphorylate T422A mutation or WT CENP-E that had been incubated with
a single conserved residue close to the CENP-E motor domain. l-phosphatase (Figure 1D). Together, these results demonstrate
We also identify a docking motif for PP1 that overlaps the site that the anti-pT422 antibody specifically recognizes CENP-E
of phosphorylation and demonstrate that PP1 binding to CENP-E phosphorylated at T422.
is disrupted by Aurora-mediated phosphorylation. Our findings To establish whether Aurora A or B phosphorylates CENP-E
establish an Aurora/PP1 phosphorylation switch that is required T422 in cells, we took advantage of the anti-pT422 antibody
not only for congression of polar chromosomes through modula- and a series of small molecule inhibitors that specifically inhibit
tion of the intrinsic motor properties of CENP-E, but also for either one or both of the Aurora kinases. As expected, treatment
subsequent stable biorientation of those chromosomes by with the dual Aurora kinase inhibitor VX-680 (Harrington et al.,
CENP-E’s delivery of PP1 to the outer kinetochore. 2004) abolished phosphorylation of the Aurora A substrate
Transforming acidic coiled-coil 3 (p-TACC3) and the Aurora B
RESULTS substrate histone H3 (p-Histone H3) (Figure 1E). VX-680 treat-
ment abolished phosphorylation of CENP-E at T422, whereas
A Conserved Phosphorylation Site Near the Neck treatments with an Aurora A specific inhibitor (MLN8054) or an
Domain of CENP-E Is Phosphorylated by Aurora Aurora B specific inhibitor (ZM447439) (Ditchfield et al., 2003;
A and B In Vitro Manfredi et al., 2007) resulted in only a partial reduction in
In searching for the origin of the one-dimensional diffusion found T422 phosphorylation, indicating that inhibition of either Aurora
in CENP-E motility (Kim et al., 2008), we identified a highly kinase alone is not sufficient to eliminate the phosphorylation

Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc. 445


A 1 324 Figure 1. A Conserved Threonine Close to
CENP-E motor core neck the Motor Domain of CENP-E Is Phosphory-
335 345 355 365 375 385 395 405 415 425
lated by Aurora A and B on the Kinetochores
Human VNE VS TDE AL L KR YRKE I MD L KKQL EE VS - - L E TR AQAMEKDQL AQL L EEKD L L QK VQNEK I EN L TRML VTSSS L T L QQE L K AKRKRR VTWC L G
of Unaligned Chromosomes
Mouse VNE VSNDE AL L KR YRRE I AD L RKQL EE VN - - TK TR AQEMEKDQL AQL L DEKD L L QK VQDEK I NN L KRML VTSSS I AL QQE L E I KRKRR VTWC YG (A) Alignment of CENP-E protein sequences using
Chicken VNE VL DDD AL L KR YRKE I L D L KKQL EE VS - - MK TQ I H AMEKHQL AH L L EEKNS L QKMQEDR I RN L TEML VTS AS FSSKQN AK ARRRRR VTWAPG
Xenopus VNE VL DDE AL L KR YRKE I L D L KKQL EN L ESSSE TK AQAMAKEEH TQL L AE I KQL HKEREDR I WH L TN I VVASSQES - QQDQR VKRKRR VTWAPG ClustalW algorithm. a-helical coiled-coil in the
Drosophila VNEMVSD ATMMKR L ERE I K VL KDK L AEEE - - - - - - - - - - - - - - - - - RKNENQQK VEH L ERQ I KHDMHK I I CGHS L SDKGQQ - - - - KRRR TWCP T
a d a d a d a d a d a d Paircoil score CENP-E neck were predicted for human CENP-E
0
Neck linker α-helical coiled-coil using Paircoil (Berger et al., 1995). Heptad repeat
0.5 Phosphothreonine
positions (a and d) in the coiled-coil are indicated.
B Aurora B kinase assay C Aurora B kinase assay E (B) In vitro kinase assays using Aurora B/INCENP
to phosphorylate Xenopus full-length CENP-E
4A

4A

WT
KD
Aurora B: -
2

2
T4

T4

or CENP-E1-473 with or without T424A mutation.


E

E
73

73

3
-47

-47
- NP-

- NP-

HCENP-E1-429
E 1-4

E 1-4

Coomassie
Coomassie staining of purified proteins and auto-
E1

E1

(49 kDa)
E

E
XC

XC
XC

XC

XC
XC

kDa kDa
radiogram showing incorporation of g-32P ATP.
pT422 Input
175 175 (C) Coomassie staining and immunoblot for pT422
HCENP-E
83 83
using human CENP-E1-429 incubated with wild-
A

type (WT) or kinase dead (KD) Aurora B.


22

62
WT

WT

62
D
T4

MycLAP-CENP-E:
MycLAP-CENP-E

λ-PPase: - - + IP
(D) CENP-E immunoprecipitates from nocoda-
(340 kDa)

47.5 47.5
Myc zole-treated DLD-1 cells expressing either WT or
IP
pT422
T422A MycGFP-CENP-E were blotted with the
Inhibit
32
P Coomassie pT422 antibody. Half the WT immunoprecipitate
was treated with l-phosphatase.
DNA Tubulin ACA pT422 Merge
F (E) Nocodazole-arrested DLD-1 cells expressing
MycGFP-CENP-E were treated with Aurora kinase
prometa
Early

inhibitors and MG132 and blotted for P-TACC3


(Aurora A substrate), P-Histone H3 (Aurora B
substrate), and tubulin (loading control). CENP-E
immunoprecipitates using a Myc antibody were
prometa
Late

blotted with the pT422 antibody.


(F) PtK2 cells were stained for DNA (Blue), Tubulin
(Green), ACA, and pT422 (Red).
(G) Nocodazole-arrested HeLa cells were treated
prometa

with VX-680 and MG132 and stained for CENP-E


Late

(Green), pT422 (Red) and DNA (Blue).


(H) pT422 fluorescence intensity was normalized
to the total CENP-E fluorescence. Plots show the
G CENP-E pT422 Merge H mean of > 15 cells per condition from two indepen-
Normalized to pT422 to

dent experiments. ***p < 0.0001 by t test. Error


total CENP-E ratio

1.2 n>15
***
DMSO
Noc +

1.0 bars represent SEM. See also Figure S1 and


0.8 Figure S2.
0.6

0.4
VX-680
Noc +

0.2

0
DMSO VX680

of CENP-E T422. However, when cells were treated with the outer kinetochore protein Bub1 (Figure S2A). Kinetochore-
MLN8054 and ZM447439 together to inhibit both Aurora A localized pT422 disappeared following depletion of CENP-E by
and B, phosphorylation of T422 was completely inhibited (Fig- siRNA (Figure S2B), confirming the specificity of the pT422 stain-
ure 1E). Thus, we conclude that both Aurora A and B contribute ing at kinetochores. Inhibition of Aurora kinases with VX-680
to the phosphorylation of CENP-E at T422 in vivo. sharply reduced kinetochore-localized pT422 signal (Figure 1G).
When normalized to the total level of CENP-E at the kinetochore
Phosphorylation of CENP-E T422 Is Enriched (which is also reduced in VX-680 treated cells (Ditchfield et al.,
on Kinetochores Close to the Spindle Poles 2003)), a > 90% reduction in T422 phosphorylation was seen
In unperturbed PtK2 cells, pT422 staining was uniformly detect- following VX-680 treatment (Figure 1H), demonstrating that
able at individual kinetochores in early prometaphase, which kinetochore-localized CENP-E is a substrate for Aurora kinases
colocalized with the centromere components recognized by in vivo.
autoantisera containing centromere antibodies (ACA) (Figure 1F).
The kinetochore-localized pT422 signal was reduced on chro- Aurora-Mediated Phosphorylation of CENP-E T422
mosomes congressed to the equator of the cells, but remained Reduces Its Affinity for Microtubules
enriched at the kinetochores of unaligned chromosomes that To determine if phosphorylation of T422 affects the motor
are close to the spindle poles (Figure 1F). In nocodazole-treated properties of CENP-E, we phosphorylated T424 of Xenopus
HeLa cells, the pT422 antibody recognized a large crescent CENP-E motor (CENP-E1-473) and measured CENP-E’s microtu-
around kinetochore pairs, which colocalized with CENP-E and bule-stimulated ATPase activity in the presence of an increasing

446 Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc.


A ATPase assays B Microtubule pelleting assays (2 mM MgADP) Figure 2. Phosphorylation of T424 of Xeno-
pus CENP-E by Aurora Kinase Reduces
15 n=3 Microtubule (μM): 0 0.125 0.25 0.5 1 2 5 CENP-E’s Affinity for Microtubules and
ATPase rate (s-1)

S P S P S P S P S P S P S P Reduces Its Run Length In Vitro


10 XCE1-473 MT (A) ATPase rates of Xenopus CENP-E1-473 and
XCE1-473 phosphorylated CENP-E1-473 measured with
MT
5 Aurora A increasing concentrations of microtubules. Plots
T424A XCE1-473
show the mean of three independent experiments
0
MT and error bars represent SEM. kcat and KmMT
0 1 2 3 4 5 T424A XCE1-473 MT values are represented ± SE.
Microtubules (μM) Aurora A
(B) Equilibrium binding of WT or T424A CENP-E1-473

CENP-E bound (%)


70
(incubated with or without Aurora A) to microtu-
60
50 WT bules in the ADP state. S, supernatant; P, pellet.
40 WT + Aurora A Percent of CENP-E1-473 bound is shown below
30
T424A (Xenopus) (n = 3; Error bars represent SD).
20
10
T424A + Aurora A (C) Kymographs showing diffusive motion of
0 CENP-E1-473-RFP preincubated with or without
Aurora A in 3 mM MgADP at 33 C.
0 1 2 3 4 5
Microtubule (μM)
(D) Duration of binding (t) was distributed exponen-
tially, and the mean binding time (tmean) was
C 3 mM MgADP, 33 ˚C D XCE1-473 F XCE1-473 determined by fitting the data into a cumulative
XCE1-473 + Aurora A XCE1-473 + Aurora A
XCE1-473 XCE1-473 + Aurora A
distribution function (exp [-t/tmean]). Inset shows
1-cumulative probability

17 sec 1-cumulative probability 1.6 μm 1-cumulative probability of CENP-E1-473-RFP


time

1 12 sec 1 1.2 μm
Frequency of events (%)

Frequency of events (%)

binding time plotted on a log scale. The tmean is


0.1
17 ± 0.13 s for CENP-E1-473-RFP, n = 231 and
0.1
60
70
50
60
12 ± 0.07 s for CENP-E1-473-RFP plus Aurora A,
0.01 0.01

50 40
40
0.001
0 25 50 75 100
0.001
0 2 4 6 8 10 n = 240. Values represented ± SE of the curve fit.
30
30 Duration of binding (sec) Run length (μm)
20
Probability < 0.0001 that the duration of binding
20
10 10 for CENP-E1-473-RFP plus Aurora A was distrib-
112 sec 0 0 uted the same as the duration of binding for
0 20 40 60 80 100 0 1 2 3 4 5 6 7 8
2 μm
Duration of binding (sec) Run length (μm) CENP-E1-473-RFP (by Kolmogorov-Smirnov Test).
(E) Kymographs showing processive motion of
E 3 mM MgATP, 33 ˚C G CENP-E1-473-RFP in the presence of 3 mM MgATP
XCE1-473 XCE1-473 + Aurora A XCE1-473 at 33 C. See Movie S1.
time

(F) Run length of CENP-E1-473-RFP was determined


5 μm

by fitting the data into a cumulative distribution func-


Microtubules

tion. Inset shows 1-cumulative probability of CENP-


E1-473-RFP run length plotted on a log scale. Mean
run length is 1.6 ± 0.02 mm for CENP-E1-473-RFP,
n = 337 and 1.2 ± 0.02 mm for CENP-E1-473-RFP
plus Aurora A, n = 294. See Movie S1. Probability <
112 sec
0.05 that the run lengths for CENP-E1-473-RFP
2 μm
plus Aurora A were distributed the same as the
CENP-E1-473-RFP run lengths (by Kolmogorov-
Smirnov Test). Values represent ± SE of the curve fit.
(G) Accumulated CENP-E (green) at the end of the
microtubules (red). See also Figure S3.

concentration of microtubules (Figure 2A). The maximal ATP and the gliding speed (data not shown), it is likely that the phos-
turnover rate (kcat) was not affected by Aurora A phosphorylation phorylation of T424 reduces CENP-E’s microtubule affinity
(13 ± 0.6 s-1 for CENP-E1-473, n = 3; 14 ± 1.3 s-1 for CENP-E1-473 primarily in its ADP bound state without affecting the rate-limiting
plus Aurora A, n = 3) (Figure 2A). However, the concentration of step in CENP-E enzymatic cycle (Woehlke et al., 1997). To test
microtubules required to reach the half maximal ATPase rate this hypothesis, the extent of Xenopus CENP-E1-473 binding to
(KmMT) was increased by > 3 fold following phosphorylation microtubules was determined with or without prior phosphoryla-
(0.17 ± 0.03 mM for CENP-E1-473, n = 3; 0.64 ± 0.15 mM tion by Aurora kinase (Figure 2B). Phosphorylation of WT
CENP-E1-473 plus Aurora A, n = 3) (Figure 2A). CENP-E1-473 by Aurora A reduced the amount of CENP-E that
KmMT reflects CENP-E’s affinity for microtubules. In the cosedimented with microtubules by 50% with a corresponding
absence of microtubules, kinesins are tightly bound to ADP in 50% increase in apparent Kd(MT) (2 mM for CENP-E1-473; 3 mM for
solution and the rate of ADP release is extremely low (Hackney, CENP-E1-473 plus Aurora A). By contrast, Aurora A did not affect
1988). However, binding of ADP-bound kinesin to microtubules microtubule binding of T424A CENP-E1-473 (apparent Kd(MT) of
greatly accelerates the rate of ADP release, and the kinesin 3.5 mM T424A CENP-E1-473; 3.4 mM for T424A CENP-E1-473
proceeds to complete its enzymatic cycle. Since phosphorylation plus Aurora A), confirming that phosphorylation at T424 reduces
of CENP-E increased KmMT without significantly affecting kcat the affinity of CENP-E for microtubules in the ADP state.

Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc. 447


A Figure 3. Phosphorylation of CENP-E at
T422 Is Required for Chromosome Align-
MycGFP Motor Discontinuous coild-coil KT MT ment
tag domain targeting binding
(A) Diagram of MycLAP-CENP-E transgene with
Myc GFP
previously identified phosphorylation sites. Splice
T422 S454 S611 S1211 T1268 S2509 S2555 variations exist within the CENP-E coiled-coil
S2512 S2558
S2543 domain and phosphorylation sites are numbered
with respect to their position in the CENP-E clone
B CENP-E siRNA targeted siRNA resistant used in this study.
against the 3'UTR of CENP-E transgene (B) Schematic of replacing endogenous CENP-E
endogenous CENP-E induced with Tet with a siRNA resistant transgene.
(C) Immunoblot showing depletion of CENP-E by
Time (h): 0 12 24 36 48 60 72 80 siRNA.
(D) Immunoblot showing knockdown with control
Plate cells Transfect Replate Add Tet (GAPDH) or CENP-E siRNA in cells expressing
siRNA cells Begin filming/ various MycLAP-CENP-E transgenes.
harvest protein (E) Box and whisker plots showing the time spent in
for immunoblot mitosis for cells expressing MycLAP-CENP-E
following transfection of control (GAPDH, green) or
CENP-E (red) siRNA. >90 cells per condition are
C D W tal MycGFP-CENP-E
plotted from at least three independent experiments.
(F) Immunofluorescence images of cells in which

A
n

or

22
re

Control siRNA C-E

A
T

ig
endogenous CENP-E has been replaced with WT
Pa

9A

T4
10
R
siRNA
Control siRNA: + - + - + - + - + - + - or T422A MycGFP-CENP-E. GFP (green); Tubulin
Dilution (%): 100 50 30 20 10 100 kDa CENP-E siRNA: - + - + - + - + - + - + kDa (red). See also Figure S4, Figure S5, Figure S6, Movie
CENP-E Myc S2, and Movie S3.
275 275
Tubulin 55 CENP-E
275
Tubulin 55
require ATP hydrolysis (Kim et al., 2008).
E F WT T422A Following phosphorylation, the duration
of CENP-E1-473-RFP binding to microtu-
n>90 bules was shortened by 30% in the
Time in mitosis (min)

DNA

presence of ADP (t = 17 ± 0.13 s for


CENP-E1-473-RFP, n = 231; t = 12 ± 0.07 s
for CENP-E1-473-RFP plus Aurora A,
n = 240) (Figure 2D), consistent with the
GFP/Tubulin

observation that phosphorylation of


T424 reduces CENP-E’s affinity to micro-
Parental WT Rigor 10A 9A T422A tubules in the ADP bound state.
MycGFP-CENP-E
Processivity of CENP-E in the presence
of ATP was reduced after phosphoryla-
Phosphorylation Reduces CENP-E Processivity tion on T424, with run lengths of phosphorylated CENP-E1-473-
Total Internal Reflection Fluorescence (TIRF) microscopy was RFP on individual microtubules 25% shorter than those of the
used to determine how Aurora phosphorylation affects properties unphosphorylated motor (1.6 ± 0.02 mm for CENP-E1-473-RFP,
of individual CENP-E molecules. Xenopus CENP-E1-473 was n = 337; 1.2 ± 0.02 mm for CENP-E1-473-RFP plus Aurora A,
tagged with the monomeric, photostable red fluorescent protein n = 294) (Figures 2E and 2F). Importantly, once reaching a micro-
TagRFP-T (CENP-E1-473-RFP) (Shaner et al., 2008). Oregon tubule end using its plus end-directed motility, individual
Green 488-labeled GMPCPP microtubules were tethered to CENP-E dimers did not immediately dissociate (Figure 2G), but
a coverslip in a flow chamber (Figure S3A) and CENP-E1-473- remained bound there for 5.8 s (±0.8 SEM, n = 26), a feature
RFP was added in the presence of apyrase to induce rigor previously observed for several other processive kinesins
binding. As expected, CENP-E1-473-RFP was stably bound in (Case et al., 1997; Okada and Hirokawa, 1999; Varga et al.,
the absence of nucleotides, and fluorescence signals were 2006).
photobleached in one or two steps 89% of the time (75 double
steps and 68 single step; n = 160), consistent with a dimeric state Phosphorylation of CENP-E T422 Is Required
for the CENP-E1-473 motor (Figure S3B). When CENP-E1-473-RFP for Chromosome Congression
was introduced into the flow chamber in a buffer containing ADP, CENP-E is phosphorylated during mitosis on at least ten sites
both phosphorylated and unphosphorylated CENP-E1-473-RFP (Figure 3A) (Nousiainen et al., 2006; Zecevic et al., 1998), albeit
remained loosely bound to microtubules without displaying the significance of these phosphorylations has not been tested.
directional motility (Figure 2C), supporting our previous observa- To determine the consequence of preventing CENP-E phos-
tion that CENP-E motility contains a diffusive mode that does not phorylation in human cells, we developed a strategy to replace

448 Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc.


endogenous CENP-E with phosphorylation defective transgenes CENP-E Phosphorylation by Aurora Is Required
(Figure 3B). Full-length CENP-E fused at the N-terminus to for Congression of Polar Chromosomes
a MycGFP epitope tag (Figure 3A) was integrated at a predefined CENP-E has been implicated in powering chromosome congres-
genomic locus in DLD-1 cells using FRT/Flp-mediated recombi- sion by transporting mono-oriented chromosomes to the spindle
nation and expression was induced by addition of tetracycline equator along mature kinetochore fibers of already bioriented
(Figure S4A). Time-lapse microcopy revealed that the subcellular chromosomes (Kapoor et al., 2006). To test whether phosphory-
distribution of WT MycGFP-CENP-E closely mirrored that of lation of T422 is required for this process, we adopted a method
endogenous CENP-E, localizing to kinetochores after nuclear to enrich mono-oriented, polar chromosomes (Kapoor et al.,
envelope breakdown and relocating to the spindle midzone in 2006; Lampson et al., 2004) in cells in which endogenous
anaphase and to the midbody during cytokinesis (Figure S5A). CENP-E was replaced with the WT or T422A MycLAP-CENP-E
Transfection of siRNA targeting the 30 untranslated region of (Figures 4A and 4B). Cells were first treated with monastrol
CENP-E mRNA depleted endogenous CENP-E by 90% across to generate monopolar spindles with a high frequency of synteli-
the population, yielding it undetectable at the kinetochores of cally-attached chromosomes and released from monastrol in
most mitotic cells (Figure 3C and data not shown). As expected, the presence of an Aurora kinase inhibitor (ZM) to allow bipolar
depletion of CENP-E extended the average duration of mitosis spindles to form while preserving improper kinetochore attach-
(to 221 min) compared to control transfected cells (71 min) ments. Following the removal of ZM, congression of mal-
(Figures 3D and 3E). Importantly, this delay was largely rescued oriented chromosomes was assessed (Figures 4A and 4B). As
by the expression of MycGFP-CENP-E (producing an average of a control, cells were treated in parallel with DMSO to determine
100 min in mitosis). Replacing endogenous CENP-E with a rigor the extent of chromosome misalignment in an unperturbed
mutant (T93N) (Figure 3D) strongly exacerbated the mitotic delay mitosis. The enrichment of improper kinetochore attachments
(to 506 min on average) with a few chromosomes chronically significantly increased the number of polar chromosomes in cells
misaligned near the spindle poles (Figure 3E), confirming our defective in T422 phosphorylation, but not in cells expressing
previous finding that the motor activity of CENP-E is essential WT CENP-E (Figures 4C–4F). Live cell imaging demonstrated
for metaphase chromosome alignment (Kim et al., 2008). that, following reactivation of the Aurora kinases, improperly
Replacement of endogenous CENP-E with a variant (10A) with attached chromosomes were frequently moved to either spindle
all 10 phosphorylation sites abolished (by substitution to pole in cells expressing WT or T422A CENP-E (Figure S7A–S7C).
alanines) produced a robust mitotic delay (505 min on average). However, these chromosomes remained closely associated with
On the other hand, abolishing phosphorylation of the nine sites those poles in cells expressing T422A CENP-E (Figure S7C),
other than T422 (9A) (Figure 3D) had little effect on mitotic establishing that phosphorylation of CENP-E on T422 by Aurora
progression (114 min on average in mitosis) (Figure 3E). Surpris- kinases is required for the congression of polar chromosomes.
ingly, preventing phosphorylation of T422 alone was sufficient to
produce a substantial mitotic delay (425 min on average), Aurora-Mediated Phosphorylation of CENP-E T422
demonstrating that of these 10 CENP-E phosphorylation sites, Opposes Direct Binding of CENP-E to the Catalytic
phosphorylation at T422 makes the largest contribution to timely Subunit of PP1
mitotic progression. Following CENP-E T422 is a highly conserved tryptophan, thereby
Replacing endogenous CENP-E with the T422A mutant producing a RRVTW sequence that conforms to the docking motif
prevented complete metaphase chromosome alignment, with for protein phosphatase 1 (PP1) (Figure 5A) (Hendrickx et al., 2009).
a few chromosomes remaining close to the spindle poles in Indeed, our mass spectrometry analysis of tandem affinity purified
> 85% of cells (Figure 3F, Figures S4B–S4D, and Figure S5B), CENP-E from mitotic human cells identified the catalytic subunit of
a phenotype highly reminiscent of that observed with diminished PP1 to be associated with CENP-E (data not shown) and PP1 was
levels of CENP-E (Weaver et al., 2003). Phosphorylation of T422 also present in CENP-E immunoprecipitates from nocodazole-
was not required for the kinetochore recruitment of CENP-E arrested DLD-1 cells (Figure 5D). The interaction between CENP-E
(Figures S6A and S6B). To eliminate the possibility that mutation and PP1 is direct, as recombinant CENP-E motor (CENP-E1-473)
of T422 caused defects other than simply preventing phosphor- was recovered together with PP1g in a pulldown experiment using
ylation, we created an additional CENP-E phosphodeficient Microcystin-beads (Figure 5B). Recovery of a stoichiometric (1:1)
mutant, in which two arginines in the Aurora consensus motif complex of CENP-E and PP1 required addition of > 5 molar excess
were converted to lysines (RR:KK) (Figure S6C). Mutation of of CENP-E over PP1, indicating a weak affinity between CENP-E
RR:KK did not abolish the epitope of the pT422 antibody (data and PP1. Further, CENP-E with a W425A substitution had mark-
not shown). However, recombinant Xenopus CENP-E1-428 edly reduced binding to PP1 (Figure 5B), demonstrating that the
carrying the RR:KK mutation could not be efficiently phosphory- interaction between CENP-E and PP1 is mediated through the
lated by Aurora A and B in vitro (Figure S6D and data not shown) PP1 docking motif. To test whether phosphorylated T422 is
and the RR:KK mutant was not phosphorylated on T422 in a substrate for PP1, phosphorylated CENP-E1-473 was incubated
human cells (Figure S6E). Indeed, replacing endogenous with either PP1g or PP1g preinactivated with the inhibitor Micro-
CENP-E with the RR:KK mutant caused a mitotic delay (Fig- cystin (Figure 5C). Monitoring of CENP-E’s phosphorylation status
ure S6F) similar to that observed with the T422A mutant with with the pT422 antibody revealed that PP1g rapidly dephosphory-
a few chromosomes remaining close to the spindle poles, con- lated CENP-E T422 (Figure 5C).
firming that phosphorylation of CENP-E at T422 is required for Previous reports have shown that phosphorylation of serine or
chromosome congression. threonine overlapping the PP1 docking motif impairs the binding

Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc. 449


A Figure 4. CENP-E Phosphorylation by
DMSO MG
DMSO MG1 3 2 Aurora Is Required for Congression of Polar
3 hr 1 .5 hr Chromosomes
(A) Drug treatment scheme to enrich mono-
Mon ZM+MG MG oriented chromosomes near spindle poles.
Monast rol ZM + MG1 3 2 MG1 3 2
(B) Diagram illustrating attachment status of
2 hr 1 hr 1 .5 hr
Fix for IF chromosomes following treatment with drug
regime in (A).
(C and D) Immunofluorescence images of drug-
Monast rol ZM + MG1 3 2 MG1 3 2
B treated cells in which endogenous CENP-E was
replaced with either WT or T422A MycLAP-
CENP-E. KNL-1 (green); DNA (red).
? (E) Percentage of cells from (C) and (D) with polar
chromosomes.
? (F) Number of polar chromosomes in cells from (C)
and (D) displaying chromosome misalignment.
Bars represent the mean of four independent
experiments. ***p < 0.0001 by t test. Error bars
DMSO MG Mon ZM+MG MG represent SEM. See also Figure S7 and Movie S4.
C D
WT T422A WT T422A

in vivo significance of the dephosphoryla-


tion of CENP-E T422 by PP1, we microin-
DNA DNA DNA DNA
KNL1 KNL1 KNL1 KNL1 jected rhodamine-labeled pT422 anti-
bodies into HeLa cells stably expressing
histone H2B-YFP. Consistent with our
E DMS O MG F ***
Cells with polar chromosomes (%)

100 Mon ZM+MG MG


immunofluorescence analysis (Figure 1F),
Number of polar chromosomes

DMS O MG the microinjected rhodamine-labeled


CENP-E siRNA Mon ZM+MG MG
80 pT422 antibody was virtually absent
CENP-E siRNA
from aligned kinetochores, but accumu-
60 lated to high levels at the kinetochores of
chromosomes positioned close to the
40 spindle poles (Figure 6B). Microinjection
of the pT422 antibody substantially de-
20
layed the duration of mitosis compared
0 to control injected cells (average 125 min
WT T422A WT T422A for rhodamine-labeled rabbit IgG injected
MycGFP-CENP-E MycGFP-CENP-E cells; 619 min for rhodamine-labeled
pT422 injected cells) (Figures 6C–6E).
Interestingly, antibody-mediated preser-
to PP1 (Hubbard and Cohen, 1989). Given that CENP-E T422 is vation of phosphorylation on CENP-E T422 promoted dynamic
overlapped by a consensus motif for Aurora kinases and chromosome movements distinct from the chromosome
a conserved motif for PP1 binding, we tested whether Aurora behaviors observed when T422 phosphorylation is abolished
phosphorylation at T422 disrupts PP1’s binding to CENP-E. (Figure S5B). Polar chromosomes congressed to the equator of
Following in vivo inhibition of T422 phosphorylation with the the cell, but most failed to make stable microtubule attachments
pan Aurora inhibitor VX-680, the amount of PP1 associated and fell back out of the spindle equator or continued to move
with CENP-E was dramatically increased (Figure 5D). Moreover, forward to the other pole (Figure 6C). Consistently, the microin-
phosphorylation of CENP-E1-473 by Aurora A resulted in a jected pT422 antibody remained enriched on the kinetochores
10-fold reduction in the binding of CENP-E to the catalytically of chromosomes juxtaposed to the metaphase plate that did
inactive (D64N) PP1g in vitro (Figure 5E), demonstrating that not form stable microtubule attachments (Figure 6B). Thus,
Aurora-mediated phosphorylation of CENP-E T422 opposes despite CENP-E-mediated congression of chromosomes to the
direct binding of CENP-E to PP1. proximity of the spindle equator, stable kinetochore attachment
(and subsequent biorientation) does not occur when dephos-
Dephosphorylation of CENP-E T422 by PP1 Is Required phorylation of CENP-E by PP1 is blocked.
for Stable Biorientation of the Chromosomes
Congressed by CENP-E DISCUSSION
The pT422 antibody inhibited PP1-mediated dephosphorylation
of Xenopus CENP-E1-473 at T424 (the position homologous to Here, we demonstrate that phosphorylation by Aurora kinases
T422 in human CENP-E) in vitro (Figure 6A). Thus, to test the of a conserved residue (T422) close to the CENP-E motor

450 Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc.


A 420 425 B Input (1/10) PP1-pulldown Figure 5. Aurora Phosphorylation of CENP-E
Xenopus K RKRR VTWAP T422 Opposes Direct Binding of CENP-E to
WT W425A WT W425A
Human K RKRR VTWC L PP1
Mouse KRKRR VTWC Y CENP-E1-473 1x 2x 5x 1x 2x 5x - 1x 2x 5x 5x 1x 2x 5x 5x
(A) CENP-E T422 overlaps the conserved PP1
Chicken RRRRR VTWAP PP1γ - - - - - - + + + + - + + + - kDa
Drosophila - - KRRR TWCP
docking motif (R/K-R/K-V-X-F/W).
CENP-E1-473 57 (B) Coomassie staining of the PP1-bound
PP1 docking motif
complexes purified with Microcystin-agarose. 1,
PP1γ 40
Aurora 2, or 5 molar excess of WT or W425A Xenopus
phosphorylation CENP-E1-473 was incubated with Xenopus PP1g.
(C) In vitro phosphatase assay using PP1g (with or
without Microcystin) and phosphorylated Xenopus
C Aurora A D MycGFP-CENP-E
1-473 1-473 CENP-E1-473 as substrate. Immunoblot shows
CENP-E p-CENP-E IP: IgG Myc phosphorylation of T422 and coomassie stain
t = 0 min
30 min t = 30 min

0
shows purified proteins.

SO
-68
PP1 PP1 + Microcystin (D) Nocodazole-arrested DLD-1 cells expressing

DM
VX
kDa
MycGFP-CENP-E were treated with VX-680 and
Time (min): 0 30 35 40 45 60 35 40 45 60 Myc MG132 before harvesting. CENP-E immunoprecip-
275
57 kDa
itates were blotted for pT422 and PP1.
pT422
pT422 (E) Coomassie staining of the PP1-bound
1-473 275
CENP-E complexes purified with Microcystin-agarose. 1, 2
Coomassie
PP1 36 or 5 molar excess of WT CENP-E1-473 was preincu-
Aurora A
bated with either WT or kinase-dead Aurora A
before incubating with catalytically inactive
E (D64N) PP1g.
Input (1/10) PP1-pulldown
+ KD AurA + AurA + KD AurA + AurA
1x 2x 5x 1x 2x 5x - 1x 2x 5x 5x 1x 2x 5x 5x CENP-E1-473
- - - - - - + + + + - + + + - D64N PP1γ

CENP-E
1-473 Thus, Aurora-mediated destabilization of
Aurora A
KD Aurora A
CENP-E tethering to individual spindle
D64N PP1γ microtubules yields a variant of kinetic
proofreading (Hopfield, 1974), with local,
destabilized attachment as a means to
domain is essential to promote the congression of polar chro- eliminate incorrect initial attachments, while allowing productive
mosomes and dephosphorylation of this site is required for CENP-E-powered movement along a kinetochore microtubule
the stable biorientation of these kinetochores. Aurora-medi- bundle.
ated phosphorylation of this site regulates the intrinsic motor A requirement for Aurora A in modulating CENP-E offers
properties of CENP-E and disrupts the binding of the a mechanistic explanation for prior reports that Aurora A inhibi-
opposing phosphatase PP1 to CENP-E, thereby establishing tion causes chromosome misalignment with a few chromo-
a bistable phosphoswitch for regulation of CENP-E (see sche- somes found close to the spindle poles (Hoar et al., 2007;
matic in Figure 7). Kunitoku et al., 2003; Marumoto et al., 2003). Although Aurora
The Aurora phosphorylation site on CENP-E is adjacent to its A-mediated phosphorylation of the centromere-specific histone
coiled-coil neck, next to several conserved positively charged H3 variant CENP-A has previously been proposed to promote
amino acids. Phosphorylation at T422 diminishes the basic chromosome congression (Kunitoku et al., 2003), we conclude
charge of what we propose to be an electrostatic tether directly that CENP-E is the kinetochore substrate whose Aurora
involved in microtubule binding (analogous to similar domains A-dependent phosphorylation is directly required for chromo-
identified in other kinesin family members [Okada and Hirokawa, some congression. For Aurora B, the absence of tension exerted
1999; Ovechkina et al., 2002; Thorn et al., 2000]). Consistently, on mono-oriented polar kinetochores and the juxtaposed posi-
phosphorylation at T422 reduces CENP-E’s affinity for microtu- tion of sister kinetochores on syntelically attached chromo-
bules and allows the motor to dissociate more readily during somes would bring it in close proximity to the highly elongated
processive runs. Phosphorylation of CENP-E 422 is highest on and flexible CENP-E, allowing Aurora B phosphorylation to
the kinetochores close to the spindle poles. Since Aurora A is modulate processivity of CENP-E attached to kinetochores
concentrated at the poles, it is likely to be responsible for phos- with reduced tension (Figures 7A and 7B). Further, Aurora
phorylation of T422 on such polar-oriented chromosomes. B-dependent phosphorylation in and around the inner centro-
Aurora phosphorylation reduces the proportion of time that meres of sister kinetochores would also be expected to prefer-
each motor molecule is bound unproductively to the many entially destabilize any incorrect attachments made by the
dynamic astral microtubules nucleated near the pole (Figure 7B). 230 nm long CENP-E to microtubules that reach across the
Phosphorylation-dependent reduction in CENP-E residence inter-kinetochore space.
time on an individual microtubule of a kinetochore fiber, on the Recent evidence has demonstrated that KNL1, one of the core
other hand, will be of little consequence, as rapid rebinding to microtubule binding components thought to be responsible for
an adjacent microtubule is likely, given the high local concentra- end-on attachment at metazoan kinetochores (Cheeseman
tion of parallel microtubules that comprise the fiber (Figure 7C). et al., 2006), binds PP1 on chromosomes aligned at metaphase.

Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc. 451


A B Figure 6. Dephosphorylation of CENP-E
pT422-Rhod Ab injected cells
Aurora A T422 by PP1 Is Required for the Stable Bio-
1-473 1-473
Histone H2B-YFP pT422-Rhod Ab rientation of Chromosomes Congressed
CENP-E p-CENP-E
from a Spindle Pole by CENP-E

Projection
t = 0 min t = 30 min

+pT422 +Rabbit IgG (A) Phosphorylated Xenopus CENP-E1-473 (1 mM)


antibody was preincubated with rabbit IgG or the pT422
1-473 1-473 antibody (3 mM) and subsequently mixed with
t = 60 min p-CENP-E p-CENP-E
PP1g (0.2 mM). Phosphorylation of T422 was
PP1 PP1 determined using rhodamine-labeled pT422 anti-

45° rotation
Boil and probe with pT422-Rhod body and visualized using the rhodamine fluoro-
Time (min): 0 30 60 65 70 75 90 60 65 70 75 90 phore. Anti-Myc immunoblot shows the CENP-
E1-473 loading.
pT422-Rhod 49 kDa
(B) Reconstructed Z-sections of rhodamine-
Myc labeled pT422 antibody-injected live HeLa cells
expressing Histone H2B-YFP. Histone H2B-YFP
C Histone H2B-YFP pT422-Rhod Ab (purple); pT422-Rhod antibody (green).
Rabbit IgG-Rhod

0:00 0:05 0:10 1:00 1:15 1:20 2:25 (C) Time-lapse images of antibody-microinjected
(green) HeLa cells stably expressing Histone
Inject:

H2B-YFP (purple). Numbered arrows track the


movement of four individual chromosomes which
congress to the equator, but fail to stably biorient.
Stills are taken from Movie S5 and Movie S6.
(D) Box and whisker plots showing the time spent
0:00 3:20 3:50 4:00 4:50 5:05
3 3
in mitosis for microinjected cells. Uninjected cells
1 1 1 in the same field of view were also examined. For
pT422-Rhod Ab

each antibody, >120 microinjected cells were


2 2 2
Inject:

observed from two independent experiments. (E)


5:20 7:25 7:35 8:15 8:45 8:50 Bar graphs showing the percentage of abnormal
mitosis for the cells observed in (D).
3 4 4
4

D evidence implicates dephosphorylation


E
of the core microtubule-binding proteins 100
Time in mitosis (min)

Abnormal mitosis (%)

1000
80 (e.g., KNL1 and the four member Ndc80
750
60
complex) by CENP-E-bound PP1 as an
500 essential step in reversing their prior inac-
40
250
tivation by Aurora-dependent phosphor-
20
ylation.
0
Non-Inj Inj Non-inj
o Inj
0
Non-Inj Inj Non-Inj Inj Finally, the spatial regulation of CENP-
Ab injected: Rabbit IgG-Rhod pT422-Rhod Ab injected: Rabbit IgG-Rhod pT422-Rhod E by Aurora kinases and PP1 may provide
an insight into the classic observation that
Binding is through a motif for PP1 docking with an overlapping phosphorylation controls the directionality of two opposing
Aurora phosphorylation site (Liu et al., 2010), a situation similar kinetochore motors on isolated chromosomes (Hyman and
to what we now report for CENP-E. Thus, the vertebrate kineto- Mitchison, 1991). To coordinate prometaphase chromosome
chore has evolved multiple modules for recruiting PP1, with movement, this phosphorylation-dependent switch must turn
recruitment by KNL1 and CENP-E each providing different func- off the minus end-directed motor and turn on the plus end-
tions. Blocking KNL1 recruitment of PP1 increased the number directed motor at the spindle poles. Here, we have shown that
of kinetochores without cold stable microtubules and decreased the plus-end directed motor properties of CENP-E (and binding
the level of PP1 recruited to kinetochores. Nevertheless, it did of PP1) are altered by a gradient of Aurora kinase activity
not affect congression or chromosome alignment, but did lead emanating from the spindle poles. This provides spatial informa-
to an unexplained inhibition of cell growth (Liu et al., 2010). In tion within the mitotic spindle to regulate CENP-E activity
contrast, we have now shown that once CENP-E tows initially according to the position of chromosome.
misoriented chromosomes to the cell center, its subsequent
dephosphorylation and rebinding of PP1 is essential for stable EXPERIMENTAL PROCEDURES
microtubule attachment to the kinetochores on these chromo-
somes (Figure 7D). Thus, we propose a model in which CENP-E Constructs
powers chromosome movement away from the high Aurora The full-length human CENP-E open reading frame was cloned into a pcDNA5/
FRT/TO based vector (Invitrogen) modified to contain an amino-terminus
activity at poles and then exploits its flexible coiled-coil and
Myc-LAP epitope tag. The LAP tag consists of GFP-TEV-S-peptide as previ-
plus end-directed motility to deliver PP1 phosphatase activity ously described (Cheeseman et al., 2004). TagRFP-T (a kind gift from Roger
within its 230 nm reach at the outer kinetochore (Figures 7E Tsien, UCSD) was cloned into pET23d vector (Novagen) containing Xenopus
and 7F). For the kinetochores on these chromosomes, our CENP-E (aa1-473). This cloning strategy generates a 16 amino acid-long linker

452 Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc.


Phosphorylation by Aurora B
Centromere protein E
+
A P P P D P
P P
P P
Cytoplasmic dynein P P
P P PP1
Aurora B PP1 +
activity
PP1
P
+
P
Protein phosphatase 1
+
- + - - + - K-fiber
PP1

Dynein-dependent poleward movement


-
KMN network PP1-mediated dephosphorylation of
Aurora A along a laterally attached microtubule;
+ + CENP-E away from polar Aurora A
Ndc80 complex activity microtubule binding by KMN network
Mis12 is inhibited by phosphorylation
complex
KNL1
ATP, Aurora A
Phosphate B P P E
P PP1 PP1
P

Kinetochore
P P

Centrioles
P P
+ -
K-fiber + P

P - - P
P PP1
Microtubules + PP1 - + - + P
P
Aurora A phosphorylation of CENP-E PP1 bound to CENP-E dephosphorylates
+ at pole releases PP1 + Ndc80 and KNL1, enabling their end-on
microtubule attachment

PP1
C P
P F
P P
PP1 + -

Kinetochore
P PP1
P
P
P
P
+ PP1

K-fiber + PP1
PP1

+ -
- - - +
Processive tracking of p-CENP-E only Stable K-fiber attachment by PP1-bound
+ along a K-fiber
+ KNL1/ Mis12 and Ndc80 complex

Figure 7. A Model of CENP-E Regulation by Aurora Kinases and PP1


(A) Unattached chromosomes are translocated poleward along a single astral microtubule in a dynein-dependent manner (Rieder and Alexander, 1990).
(B) Aurora A phosphorylates CENP-E at T422 near the spindle poles, releasing PP1 from CENP-E. Phosphorylation of CENP-E provides selectivity toward
bundles of kinetochore microtubules (K-fiber) for CENP-E to glide along.
(C) Our evidence here demonstrates that CENP-E powers the congression of polar chromosomes to the spindle equator along the K-fiber of an already bioriented
chromosome, as earlier proposed (Kapoor et al., 2006).
(D) As chromosomes congress, kinetochores move away from the Aurora A gradient concentrated at the spindle poles and CENP-E is dephosphorylated.
(E) Dephosphorylated CENP-E recruits a high local concentration of PP1 to the outer kinetochores of chromosomes it has translocated away from a pole. CENP-E
delivered PP1 is essential for stable, microtubule capture by the kinetochores of these towed chromosomes through its dephosphorylation of key components at
the kinetochore-microtubule interface (e.g., Ndc80 and KNL1), thereby increasing their affinity for microtubules.
(F) Upon biorientation, kinetochore substrates become separated from the inner centromeric Aurora B. Kinetochore-recruited PP1 through its direct binding to
KNL1 and CENP-E stabilizes kinetochore-microtubule interactions.

(MASMTGGGGMGRLR) between CENP-E and TagRFP-T. All human and 0.5 mM and MLN8054 (a kind gift of Patrick Eyers), 0.25 mM. All small molecules
Xenopus CENP-E mutants were generated by site-directed mutagenesis were from Sigma-Aldrich unless otherwise specified.
(QuickChange, Stratagene).
Immunofluorescence Microscopy
Cells were pre-extracted for 90 s in MTSB (100 mM PIPES [pH 6.8], 0.1%
Cell Culture and siRNA Treatment
Triton X-100, 0.1 mM CaCl2, 1 mM MgCl2) and fixed in 2% formaldehyde in
Cells were maintained at 37 C in a 5% CO2 atmosphere in Dulbecco’s
MTSB. Cells were blocked in 2.5% FBS, 0.2 M glycine, 0.1% Triton X-100 in
modified eagle medium (DMEM) containing 10% tetracycline free fetal bovine
PBS for 1 hr. For the pT422 staining, cells were extracted and fixed in the pres-
serum (Clontech), 100 U/ml penicillin, 100 U/ml streptomycin and 2 mM L-
ence of 500 nM Microcystin-LR (EMD). Antibody incubations were conducted
glutamine. For siRNA treatment, 1.5 3 105 cells were plated in a 6-well plate
in blocking solution for 1 hr. DNA was detected using DAPI and cells were
and duplexed siRNAs were introduced using Oligofectamine (Invitrogen).
mounted in ProLong (Invitrogen). Images were collected using a DeltaVision
siRNAs directed against CENP-E (50 -CCACUAGAGUUGAAAGAUA-30 ) and
Core system (Applied Precision) controlling an interline charge-coupled device
GAPDH (50 -UGGUUUACAUGAUCCAAUA-30 ) were purchased from Dharma-
camera (Cooldsnap, Raper). Kinetochore signal intensity was determined
con. Cells were processed for immunofluorescence microscopy or live cell
using MetaMorph (Molecular Devices), by measuring integrated fluorescence
imaging 48 hr after transfection.
intensity with a 10 3 10 pixel square. Background signal was subtracted
from an area adjacent to the kinetochore. The mean integrated fluorescence
Generation of Stable Cell Lines intensity of at least 10 kinetochore pairs per cell was calculated. Antibodies
Stable DLD-1, H2B-RFP cell lines expressing CENP-E were generated using used are specified in the Extended Experimental Procedures.
the FRT/Flp-mediated recombination as described previously (Holland et al.,
2010). Small molecules were used at the following final concentrations: noco- Single Molecule Assays
dazole, 0.2 mg/ml; taxol, 10 mM; monastrol, 20 mM; S-Trityl-L-cysteine, 5 mM; CENP-E single molecule assays were performed as previously described (Kim
MG132, 20 mM; ZM447439 (Tocris Bioscience), 3 mM; VX-680 (Selleck) et al., 2008) with the following modifications. Slides and 22 3 22-mm square

Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc. 453


coverslips were silanized as described (Helenius et al., 2006). A flow chamber Cleveland, D.W., Mao, Y., and Sullivan, K.F. (2003). Centromeres and
was incubated with 50 mg/ml of a rat monoclonal anti-tubulin antibody (YL1/2, Kinetochores: From Epigenetics to Mitotic Checkpoint Signaling. Cell 112,
Serotec) for 5 min, followed by 1% Pluronic F-127 (Invitrogen) in BRB80 for 407–421.
15 min and Oregon Green 488-labeled GMPCPP microtubules for 10 min. Ditchfield, C., Johnson, V.L., Tighe, A., Ellston, R., Haworth, C., Johnson, T.,
0.2 mg/ml of Xenopus CENP-E1-473-RFP was incubated with 50 mg/ml of Mortlock, A., Keen, N., and Taylor, S.S. (2003). Aurora B couples chromosome
Aurora A in 20 mM Tris (pH 7.5), 25 mM KCl, 1 mM MgCl2, 1mM DTT, alignment with anaphase by targeting BubR1, Mad2, and Cenp-E to kineto-
0.1 mM MgATP for 15 min at room temperature and diluted to 0.5 nM before chores. J. Cell Biol. 161, 267–280.
imaging in motility buffer (25 mM K-PIPES, [pH 6.9], 5 mM MgCl2, 1 mM EGTA,
Emanuele, M.J., Lan, W., Jwa, M., Miller, S.A., Chan, C.S.M., and Stukenberg,
1 mM DTT, 0.25% Brij35, 0.5 mg/ml casein, 4.5 mg/ml glucose, 0.2 mg/ml
P.T. (2008). Aurora B kinase and protein phosphatase 1 have opposing roles in
glucose oxidase, 0.35 mg/ml catalase, 0.5% bME) containing either 3 mM
modulating kinetochore assembly. J. Cell Biol. 181, 241–254.
MgATP or 3 mM MgADP. Frames were captured every 500 ms with 200 ms
exposure, and the typical duration of imaging was 2–3 min. Note, that since Espeut, J., Gaussen, A., Bieling, P., Morin, V., Prieto, S., Fesquet, D., Surrey,
imaging was performed at an elevated temperature (33 C) and in higher T., and Abrieu, A. (2008). Phosphorylation Relieves Autoinhibition of the
MgCl2, the speed of CENP-E movement was faster than that measured at Kinetochore Motor Cenp-E. Mol. Cell 29, 637–643.
room temperature in our previous study (Kim et al., 2008). Francisco, L., Wang, W., and Chan, C.S. (1994). Type 1 protein phosphatase
acts in opposition to IpL1 protein kinase in regulating yeast chromosome
segregation. Mol. Cell. Biol. 14, 4731–4740.
SUPPLEMENTAL INFORMATION
Gestaut, D.R., Graczyk, B., Cooper, J., Widlund, P.O., Zelter, A., Wordeman,
Supplemental Information includes Extended Experimental Procedures, L., Asbury, C.L., and Davis, T.N. (2008). Phosphoregulation and depolymeriza-
Supplemental References, seven figures, and six movies and can be found tion-driven movement of the Dam1 complex do not require ring formation. Nat.
with this article online at doi:10.1016/j.cell.2010.06.039. Cell Biol. 10, 407–414.
Hackney, D.D. (1988). Kinesin ATPase: rate-limiting ADP release. Proc. Natl.
Acad. Sci. USA 85, 6314–6318.
ACKNOWLEDGMENTS
Harrington, E.A., Bebbington, D., Moore, J., Rasmussen, R.K., Ajose-
Adeogun, A.O., Nakayama, T., Graham, J.A., Demur, C., Hercend, T., Diu-
The authors would like to thank Stephen Taylor, Patrick Eyers, Todd Stuken-
Hercend, A., et al. (2004). VX-680, a potent and selective small-molecule
berg, and Arshad Desai for providing reagents. This work was supported by
inhibitor of the Aurora kinases, suppresses tumor growth in vivo. Nat. Med.
a grant (GM29513) from the National Institutes of Health to D.W.C., who
10, 262–267.
receives salary support from the Ludwig Institute for Cancer Research.
A.J.H. is supported by a European Molecular Biology Organization (EMBO) Helenius, J., Brouhard, G., Kalaidzidis, Y., Diez, S., and Howard, J. (2006).
Long-Term Fellowship. The depolymerizing kinesin MCAK uses lattice diffusion to rapidly target
microtubule ends. Nature 441, 115–119.
Received: October 27, 2009 Hendrickx, A., Beullens, M., Ceulemans, H., Den Abt, T., Van Eynde, A.,
Revised: April 26, 2010 Nicolaescu, E., Lesage, B., and Bollen, M. (2009). Docking Motif-Guided
Accepted: June 14, 2010 Mapping of the Interactome of Protein Phosphatase-1. Chem. Biol. 16,
Published: August 5, 2010 365–371.
Hoar, K., Chakravarty, A., Rabino, C., Wysong, D., Bowman, D., Roy, N., and
REFERENCES Ecsedy, J.A. (2007). MLN8054, a Small-Molecule Inhibitor of Aurora A, Causes
Spindle Pole and Chromosome Congression Defects Leading to Aneuploidy.
Akiyoshi, B., Nelson, C.R., Ranish, J.A., and Biggins, S. (2009). Quantitative Mol. Cell. Biol. 27, 4513–4525.
proteomic analysis of purified yeast kinetochores identifies a PP1 regulatory Holland, A.J., and Cleveland, D.W. (2009). Boveri revisited: chromosomal
subunit. Genes Dev. 23, 2887–2899. instability, aneuploidy and tumorigenesis. Nat. Rev. Mol. Cell Biol. 10,
Berger, B., Wilson, D.B., Wolf, E., Tonchev, T., Milla, M., and Kim, P.S. (1995). 478–487.
Predicting coiled coils by use of pairwise residue correlations. Proc. Natl. Holland, A.J., Lan, W., Niessen, S., Hoover, H., and Cleveland, D.W. (2010).
Acad. Sci. USA 92, 8259–8263. Polo-like kinase 4 kinase activity limits centrosome overduplication by
Case, R.B., Pierce, D.W., Hom-Booher, N., Hart, C.L., and Vale, R.D. (1997). autoregulating its own stability. J. Cell Biol. 188, 191–198.
The Directional Preference of Kinesin Motors Is Specified by an Element Hopfield, J.J. (1974). Kinetic proofreading: A new mechanism for reducing
outside of the Motor Catalytic Domain. Cell 90, 959–966. errors in biosynthetic processes requiring high specificity. Proc. Natl. Acad.
Cheeseman, I.M., Anderson, S., Jwa, M., Green, E.M., Kang, J.-s., Yates, J.R., Sci. USA 71, 4135–4139.
Chan, C.S.M., Drubin, D.G., and Barnes, G. (2002). Phosphoregulation of Hubbard, M.J., and Cohen, P. (1989). Regulation of protein phosphatase-1G
Kinetochore-Microtubule Attachments by the Aurora Kinase Ipl1p. Cell 111, from rabbit skeletal muscle. 1. Phosphorylation by cAMP-dependent protein
163–172. kinase at site 2 releases catalytic subunit from the glycogen-bound
Cheeseman, I.M., Niessen, S., Anderson, S., Hyndman, F., Yates, J.R., holoenzyme. FEBS 186, 701.
Oegema, K., and Desai, A. (2004). A conserved protein network controls Hyman, A.A., and Mitchison, T.J. (1991). Two different microtubule-based
assembly of the outer kinetochore and its ability to sustain tension. Genes motor activities with opposite polarities in kinetochores. Nature 351, 206–211.
Dev. 18, 2255–2268. Kapoor, T.M., Lampson, M.A., Hergert, P., Cameron, L., Cimini, D., Salmon,
Cheeseman, I.M., Chappie, J.S., Wilson-Kubalek, E.M., and Desai, A. (2006). E.D., McEwen, B.F., and Khodjakov, A. (2006). Chromosomes Can Congress
The Conserved KMN Network Constitutes the Core Microtubule-Binding to the Metaphase Plate Before Biorientation. Science 311, 388–391.
Site of the Kinetochore. Cell 127, 983–997. Kim, Y., Heuser, J.E., Waterman, C.M., and Cleveland, D.W. (2008). CENP-E
Cimini, D., and Degrassi, F. (2005). Aneuploidy: a matter of bad connections. combines a slow, processive motor and a flexible coiled coil to produce an
Trends Cell Biol. 15, 442–451. essential motile kinetochore tether. J. Cell Biol. 181, 411–419.
Cimini, D., Wan, X., Hirel, C.B., and Salmon, E.D. (2006). Aurora Kinase Kunitoku, N., Sasayama, T., Marumoto, T., Zhang, D., Honda, S., Kobayashi,
Promotes Turnover of Kinetochore Microtubules to Reduce Chromosome O., Hatakeyama, K., Ushio, Y., Saya, H., and Hirota, T. (2003). CENP-A
Segregation Errors. Curr. Biol. 16, 1711–1718. Phosphorylation by Aurora-A in Prophase Is Required for Enrichment of

454 Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc.


Aurora-B at Inner Centromeres and for Kinetochore Function. Dev. Cell 5, Shaner, N.C., Lin, M.Z., McKeown, M.R., Steinbach, P.A., Hazelwood, K.L.,
853–864. Davidson, M.W., and Tsien, R.Y. (2008). Improving the photostability of bright
Lampson, M.A., Renduchitala, K., Khodjakov, A., and Kapoor, T.M. (2004). monomeric orange and red fluorescent proteins. Nat. Methods 5, 545–551.
Correcting improper chromosome-spindle attachments during cell division. Tanaka, T.U., Rachidi, N., Janke, C., Pereira, G., Galova, M., Schiebel, E.,
Nat. Cell Biol. 6, 232–237. Stark, M.J.R., and Nasmyth, K. (2002). Evidence that the Ipl1-Sli15 (Aurora
Liao, H., Li, G., and Yen, T.J. (1994). Mitotic regulation of microtubule Kinase-INCENP) complex promotes chromosome biorientation by altering
cross-linking activity of CENP-E kinetochore protein. Science 265, 394–398. kinetochore-spindle pole connections. Cell 108, 317–329.
Liu, D., Vleugel, M., Backer, C.B., Hori, T., Fukagawa, T., Cheeseman, I.M., Thorn, K.S., Ubersax, J.A., and Vale, R.D. (2000). Engineering the Processive
and Lampson, M.A. (2010). Regulated targeting of protein phosphatase 1 to Run Length of the Kinesin Motor. J. Cell Biol. 151, 1093–1100.
the outer kinetochore by KNL1 opposes Aurora B kinase. J. Cell Biol. 188,
809–820. Trinkle-Mulcahy, L., Andersen, J., Lam, Y.W., Moorhead, G., Mann, M., and
Lamond, A.I. (2006). Repo-Man recruits PP1g to chromatin and is essential
Manfredi, M.G., Ecsedy, J.A., Meetze, K.A., Balani, S.K., Burenkova, O., Chen,
for cell viability. J. Cell Biol. 172, 679–692.
W., Galvin, K.M., Hoar, K.M., Huck, J.J., LeRoy, P.J., et al. (2007). Antitumor
activity of MLN8054, an orally active small-molecule inhibitor of Aurora A Trinkle-Mulcahy, L., Andrews, P.D., Wickramasinghe, S., Sleeman, J.,
kinase. Proc. Natl. Acad. Sci. USA 104, 4106–4111. Prescott, A., Lam, Y.W., Lyon, C., Swedlow, J.R., and Lamond, A.I. (2003).
Marumoto, T., Honda, S., Hara, T., Nitta, M., Hirota, T., Kohmura, E., and Saya, Time-lapse imaging reveals dynamic relocalization of PP1gamma throughout
H. (2003). Aurora-A kinase maintains the fidelity of early and late mitotic events the mammalian cell cycle. Mol. Biol. Cell 14, 107.
in HeLa cells. J. Biol. Chem. 278, 51786–51795. Varga, V., Helenius, J., Tanaka, K., Hyman, A.A., Tanaka, T.U., and Howard, J.
Marumoto, T., Zhang, D., and Saya, H. (2005). Aurora-A - A guardian of poles. (2006). Yeast kinesin-8 depolymerizes microtubules in a length-dependent
Nat. Rev. Cancer 5, 42–50. manner. Nat. Cell Biol. 8, 957–962.
Nousiainen, M., Sillje, H.H., Sauer, G., Nigg, E.A., and Korner, R. (2006). Weaver, B.A.A., Bonday, Z.Q., Putkey, F.R., Kops, G.J.P.L., Silk, A.D., and
Phosphoproteome analysis of the human mitotic spindle. Proc. Natl. Acad. Cleveland, D.W. (2003). Centromere-associated protein-E is essential for the
Sci. USA 103, 5391–5396. mammalian mitotic checkpoint to prevent aneuploidy due to single chromo-
Okada, Y., and Hirokawa, N. (1999). A Processive Single-Headed Motor: some loss. J. Cell Biol. 162, 551–563.
Kinesin Superfamily Protein KIF1A. Science 283, 1152–1157. Woehlke, G., Ruby, A.K., Hart, C.L., Ly, B., Hom-Booher, N., and Vale, R.D.
Ovechkina, Y., Wagenbach, M., and Wordeman, L. (2002). K-loop insertion (1997). Microtubule Interaction Site of the Kinesin Motor. Cell 90, 207–216.
restores microtubule depolymerizing activity of a ‘‘neckless’’ MCAK mutant.
Wood, K.W., Sakowicz, R., Goldstein, L.S.B., and Cleveland, D.W. (1997).
J. Cell Biol. 159, 557–562.
CENP-E Is a Plus End-Directed Kinetochore Motor Required for Metaphase
Putkey, F.R., Cramer, T., Morphew, M.K., Silk, A.D., Johnson, R.S., McIntosh, Chromosome Alignment. Cell 91, 357–366.
J.R., and Cleveland, D.W. (2002). Unstable Kinetochore-Microtubule Capture
and Chromosomal Instability Following Deletion of CENP-E. Dev. Cell 3, Yao, X., Abrieu, A., Zheng, Y., Sullivan, K.F., and Cleveland, D.W. (2000).
351–365. CENP-E forms a link between attachment of spindle microtubules to kineto-
chores and the mitotic checkpoint. Nat. Cell Biol. 2, 484–491.
Rieder, C.L., and Alexander, S.P. (1990). Kinetochores are transported
poleward along a single astral microtubule during chromosome attachment Yucel, J.K., Marszalek, J.D., McIntosh, J.R., Goldstein, L.S.B., Cleveland,
to the spindle in newt lung cells. J. Cell Biol. 110, 81–95. D.W., and Philp, A.V. (2000). CENP-meta, an Essential Kinetochore Kinesin
Ruchaud, S., Carmena, M., and Earnshaw, W.C. (2007). Chromosomal Required for the Maintenance of Metaphase Chromosome Alignment in
passengers: conducting cell division. Nat. Rev. Mol. Cell Biol. 8, 798–812. Drosophila. J. Cell Biol. 150, 1–12.
Sassoon, I., Severin, F.F., Andrews, P.D., Taba, M.-R., Kaplan, K.B., Ashford, Zecevic, M., Catling, A.D., Eblen, S.T., Renzi, L., Hittle, J.C., Yen, T.J.,
A.J., Stark, M.J.R., Sorger, P.K., and Hyman, A.A. (1999). Regulation of Gorbsky, G.J., and Weber, M.J. (1998). Active MAP Kinase in Mitosis:
Saccharomyces cerevisiae kinetochores by the type 1 phosphatase Glc7p. Localization at Kinetochores and Association with the Motor Protein
Genes Dev. 13, 545–555. CENP-E. J. Cell Biol. 142, 1547–1558.

Cell 142, 444–455, August 6, 2010 ª2010 Elsevier Inc. 455


PNPASE Regulates RNA Import
into Mitochondria
Geng Wang,1 Hsiao-Wen Chen,4 Yavuz Oktay,1 Jin Zhang,5 Eric L. Allen,5 Geoffrey M. Smith,5 Kelly C. Fan,5
Jason S. Hong,5 Samuel W. French,5 J. Michael McCaffery,6 Robert N. Lightowlers,7 Herbert C. Morse III,8
Carla M. Koehler,1,2,* and Michael A. Teitell2,3,5,*
1Department of Chemistry and Biochemistry
2Molecular Biology Institute
3Jonsson Comprehensive Cancer Center, Broad Stem Cell Research Center, California NanoSystems Institute, and Center for Cell Control

University of California at Los Angeles, Los Angeles, CA 90095, USA


4Center for Molecular and Mitochondrial Medicine and Genetics, University of California at Irvine, Irvine, CA 92697, USA
5Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
6Integrated Imaging Center, Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
7Mitochondrial Research Group, Institute for Ageing and Health, Newcastle University, Newcastle upon the Tyne, UK
8Laboratory of Immunopathology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville,

MD 20852, USA
*Correspondence: koehler@chem.ucla.edu (C.M.K.), mteitell@mednet.ucla.edu (M.A.T.)
DOI 10.1016/j.cell.2010.06.035

SUMMARY somes, with mammalian mitochondria importing several


different tRNAs both in vitro and in vivo (Kolesnikova et al.,
RNA import into mammalian mitochondria is consid- 2004; Rubio et al., 2008). Recently, microRNAs were isolated
ered essential for replication, transcription, and from mitochondria (Kren et al., 2009) and the nuclear-encoded
translation of the mitochondrial genome but the 5S rRNA was identified as one of the most abundant RNAs in
pathway(s) and factors that control this import are human mitochondria (Entelis et al., 2001; Smirnov et al., 2008).
poorly understood. Previously, we localized polynu- RNase MRP and RNase P enzyme complexes localize and
function in mammalian mitochondria and may contain RNAs
cleotide phosphorylase (PNPASE), a 30 / 50 exoribo-
that are encoded within the nucleus. RNase MRP functions
nuclease and poly-A polymerase, in the mitochon-
as a site-specific endoribonuclease involved in primer RNA
drial intermembrane space, a location lacking cleavage during the replication of mitochondrial DNA (Chang
resident RNAs. Here, we show a new role for PNPASE and Clayton, 1987). Mammalian RNase P functions in the
in regulating the import of nuclear-encoded RNAs processing of tRNAs during the maturation of mitochondrial
into the mitochondrial matrix. PNPASE reduction transcripts that encode oxidative phosphorylation (OXPHOS)
impaired mitochondrial RNA processing and polycis- components. Large polycistronic RNA transcripts are generated
tronic transcripts accumulated. Augmented import of from the heavy and light strand promoters in mammalian mito-
RNase P, 5S rRNA, and MRP RNAs depended on chondria (Bonawitz et al., 2006). tRNAs often separate the
PNPASE expression and PNPASE–imported RNA coding regions for OXPHOS subunits and are processed and
interactions were identified. PNPASE RNA process- removed by RNase P (Doersen et al., 1985). Earlier studies
showed that the RNA component of RNase P was imported
ing and import activities were separable and a mito-
into mammalian mitochondria (Doersen et al., 1985) and the
chondrial RNA targeting signal was isolated that RNase P RNA and processing activity has been copurified
enabled RNA import in a PNPASE-dependent from mitochondria (Puranam and Attardi, 2001). By contrast,
manner. Combined, these data strongly support an RNase P RNA is encoded in the mitochondrial genome in
unanticipated role for PNPASE in mediating the trans- Saccharomyces cerevisiae (Hollingsworth and Martin, 1986).
location of RNAs into mitochondria. Recently, an additional RNase P enzyme, consisting of three
protein subunits, has been purified from human mitochondria.
INTRODUCTION This alternative RNase P enzyme processes single tRNA 50
precursor sequences in vitro without an RNA component
Much is understood about the mechanisms that regulate (Holzmann et al., 2008; Walker and Engelke, 2008). The identifi-
nuclear-encoded protein import into mitochondria (Chacinska cation of two enzymes with RNase P activity in mammalian
et al., 2009). By contrast, much less is known about the factors mitochondria warrants further investigation.
that regulate mitochondrial RNA import. Almost every organism Critical open questions about RNA import into mammalian
with mitochondria imports tRNAs and aminoacyl-tRNA synthe- mitochondria include the selectivity of RNAs, the factor(s) target-
tases (Alfonzo and Soll, 2009; Duchene et al., 2009). The number ing RNA from the cytosol, and the translocation pathway(s)
of imported tRNAs ranges from one in yeast to all in trypano- across the mitochondrial membranes (Duchene et al., 2009).

456 Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc.


Different import pathways have been proposed for a subset of and mammalian mitochondria into a homo-oligomeric
precursors in different species and the details of the components complex, consisting of a trimer or a ‘dimer of trimers’ (Symmons
involved remain ill-defined. For example, the TOM and TIM et al., 2002). These results strongly suggest that PNPASE
protein import complexes have been implicated in the import assembles and may function similarly in yeast and mammalian
of tRNALys in yeast mitochondria (Tarassov et al., 1995). By mitochondria.
contrast, a protein import pathway-independent mechanism
may exist and involve a 600 kDa multi-subunit RNA import Pnpt1 Knockout Cells Show Altered Mitochondrial
complex (RIC) in Leishmania (Mukherjee et al., 2007). Thus, Morphology and Impaired Respiration
inconsistencies among RNA import systems suggest a critical We used several approaches to determine the function of
need to learn more about the nature of the RNAs that are PNPASE in mitochondria. First, the gene encoding PNPASE
imported and the factor(s) mediating this import. (Pnpt1) was knocked out (KO) in C57BL/6 mice (Figure S2 and
Previously we showed an unexpected location in the mito- Figure S3). Homozygous Pnpt1neo-flox mice, in which exon 2
chondrial intermembrane space (IMS) for mammalian polynucle- was flanked by loxP recombination sites, were viable and fertile.
otide phosphorylase (PNPASE) (Chen et al., 2007; Chen et al., A complete KO of Pnpt1 exon 2 was generated by crossing
2006; Rainey et al., 2006), a 30 / 50 exoribonuclease and CMVCRE expressing mice with Pnpt1WT/neo-flox heterozygotes
poly-A polymerase that uses phosphorolysis to degrade RNA followed by inter-crossing the Pnpt1WT/KO progeny. Pnpt1KO/KO
(Yehudai-Resheff et al., 2001). This was a surprise because mice were embryonic lethal (Figure 1A).
We expected that PNPASE would instead localize in the RNA- Our prior cell line studies showed that RNAi targeting PNPASE
abundant mitochondrial matrix. Therefore, initial studies on reduced ATP production by OXPHOS and slowed cell growth
mammalian PNPASE focused on a general role in maintaining (Chen et al., 2006). Recently, a viable liver KO of the COX10
mitochondrial homeostasis, potentially by regulating adenine gene, which is required for cytochrome c oxidase and ETC func-
nucleotide levels (Chen et al., 2006; Portnoy et al., 2008). Here, tion, was produced, suggesting that targeted disruption of Pnpt1
we show that PNPASE has a central role in augmenting the in hepatocytes might be tolerated (Diaz et al., 2008). Therefore,
import of small RNA components required for DNA replication a liver-specific KO (HepKO) of Pnpt1 was generated by the
and RNA processing into the mitochondrial matrix. We suggest cross AlbCRE/WT/Pnpt1neo-flox/neo-floxx AlbWT/WT/Pnpt1neo-flox/neo-flox,
that PNPASE regulates adenine nucleotide levels and mitochon- which produced fertile progeny at the expected frequency
drial homeostasis at least partly by regulating RNA import to (Figure S2C). Quantitative real-time PCR (QPCR) from HepKO
control the abundance of the electron transport chain (ETC) liver showed reduced Pnpt1 transcripts containing targeted
components. exon 2 compared with those containing untargeted exon 28
(Figure 1B). PNPASE protein expression was also markedly
RESULTS reduced in HepKO liver compared with sex-matched littermate
WT liver (Figure 1B). The expression of Pnpt1 transcripts and
PNPASE Forms a Trimer in Yeast and Mammalian PNPASE protein in HepKO liver likely arises from heterogeneous
Mitochondria liver elements. Also, the AlbCRE deleting strain is hepatocyte-
To examine PNPASE in the IMS, a coimmunoprecipitation (IP) specific but incomplete (Postic and Magnuson, 2000). The
assay was performed to identify potential binding partners. A physiology of these mice will be reported elsewhere.
6XHis-Protein-C (HisPC) tag was added to the C terminus of The ultrastructure of HepKO liver mitochondria was investi-
PNPASE and stable PNPASE-HisPC expressing HEK293 cells gated by transmission electron microscopy (TEM). Rather than
were generated. The PNPASE-HisPC protein was detected in displaying ordered, linear cristae with convolutions as in WT
isolated mitochondria by immunoblot (Figure S1A available mitochondria, the HepKO mitochondria showed disordered
online). PNPASE-HisPC was isolated from mitochondria using circular and smooth IM cristae (Figure 1C), similar to mitochon-
sequential purification over Ni2+ and Protein-C affinity columns, dria that are impaired for OXPHOS (Mandel et al., 2001) and to
followed by elution, and copurifying proteins were identified by Pnpt1 RNAi mammalian cell lines (Chen et al., 2006). These
Sypro Ruby staining (Figure S1B) and liquid chromatography- and prior results suggesting that reduced PNPASE caused a
tandem mass spectrometry (LC-MS/MS) (data not shown). All decrease in ATP production prompted the evaluation of O2
of the identified bands originated from PNPASE, suggesting consumption from HepKO liver mitochondria. Oxygen electrode
that PNPASE lacks partner proteins in vivo. Bands of lower studies showed a 1.5- to 2-fold decrease in the activity of
molecular weight than the 85 kDa of PNPASE monomers Complex IV and Complexes II+III+IV when normalized to citrate
were likely degradation products. The assembly state of synthase activity in HepKO compared to WT mitochondria
PNPASE was also investigated. Mitochondria from yeast ex- (Figure 1D). Combined, these data establish an in vivo role for
pressing human PNPASE (Rainey et al., 2006) were detergent PNPASE in mitochondrial morphogenesis and respiration
solubilized and separated on blue-native (BN) gels. Immunoblot through an unknown mechanism.
showed PNPASE in a complex of 240 kDa (Figure S1C), similar
to the trimeric complex of endogenous mouse hepatocyte PNPASE Is Required for the Processing
PNPASE (Chen et al., 2006) and bacterially-expressed human of Mitochondrial RNA Transcripts
PNPASE (French et al., 2007). PNPASE-HisPC isolated from The data showing decreased respiration in HepKO mitochon-
HEK293 mitochondria also migrated in a similarly-sized complex dria suggested a reduction in functional ETC complexes. There-
(Figure S1D). Overall, PNPASE assembled identically in yeast fore, RNA processing and translation were examined in cells

Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc. 457


Figure 1. Deletion of Pnpt1 in Hepatocytes
Impairs Mitochondrial Function
(A) Breeding strategy (top) and results (bottom) for
attempting to generate a PNPASE KO mouse.
(B) Hepatocyte-specific Pnpt1 KO (HepKO) in
4-week old mice. Top: QPCR for liver Pnpt1
expression using an exon 2 – exon 3 primer pair
versus a primer pair within exon 28. Bottom:
PNPASE immunoblot from 4-week old WT and
HepKO mouse livers.
(C) HepKO mitochondria have altered cristae. Left:
TEM of 6-week old littermate livers shows circular,
smooth HepKO IM cristae in contrast to linear,
stacked cristae of WT mitochondria. Right: Anal-
ysis of cristae morphology in which a single normal
cristae within a mitochondrion was scored as
normal. Indet = indeterminate.
(D) Decreased respiration in isolated HepKO mito-
chondria. Oxygen consumption (nmol/min/mg
protein) for ETC complexes IV and II+III+IV was
measured using an O2 electrode. Mitochondrial
mass was determined by citrate synthase (CS)
activity using a spectrophotometer. Respiratory
activities are shown normalized to CS activity.
(E) Decreased mature mtRNAs in HEK293 cells
with RNAi to PNPT1. Transcripts were quantified
relative to cytosolic GAPDH expression by QPCR
from HEK293 cells 7d post-infection (nadir) with
scramble (Scr) or PNPT1 RNAi retroviral
constructs. See also Figure S1, Figure S2, and
Figure S3.

precursor transcripts. Isolated mitochon-


drial RNAs were examined by RT-PCR. A
900-bp fragment was detected from
HepKO but not from WT liver mitochon-
dria (Figure 2B). Similar results were
obtained using the same primers in
PNPASE KO mouse embryonic fibro-
blasts (MEFs) (Figure S3 and Figure S4A).
with decreased PNPASE. HEK293 cells with > 75% reduced To query RNA processing at a second site, primers were gener-
PNPASE expression were generated by RNAi, followed by ated for adjacent Cox2 and Atp8/6 loci, separated by tRNAlys.
mitochondrial RNA (mtRNA) transcript quantification using Again, polycistronic transcripts accumulated in the PNPASE
QPCR normalized to cytosolic GAPDH RNA. All mtRNAs tested KO MEFs (Figure S4B). The sizes of Cox1 and Cox3 transcripts
were reduced in Pnpt1 RNAi cells compared to WT cells were investigated using specific probes and Northern blot
(Figure 1E). Proteins translated from mtRNAs were decreased (Figure 2C). In addition to the mature Cox1 and Cox3 transcripts,
in HEK293 Pnpt1 RNAi cells (data not shown) and HepKO liver a range of larger precursor transcripts was seen in HepKO liver
cells (Figure 2A), strongly suggesting that a decrease in cells. Also, the mature 0.9-kb Cox3 transcript was more
functional ETC complexes was responsible for decreased abundant in WT than HepKO liver. The abundance of mitochon-
respiration. dria-encoded COX3 and ND6 proteins was determined
The processing of polycistronic mtRNAs was investigated (Figure 2D). In HepKO liver mitochondria, the steady-state
because reduced PNPASE could cause an accumulation of large abundance of PNPASE was decreased by 2-fold compared
precursor transcripts, resulting in reduced ETC proteins. Tran- to the WT, similar to a 2-fold decrease for COX3 and ND6
script processing requires RNase P excision of the tRNAs proteins. Controls TOM40, MORTALIN, TIM23, and BAP37
between ETC gene coding regions. Primers were designed to showed that the amount of nuclear-encoded mitochondrial
test processing between adjacent Cox1 and Cox2 transcripts proteins, and therefore the mitochondrial mass, was similar
that are separated by tRNAser and tRNAasp (Figure 2B). The between HepKO and sex-matched WT littermate liver cells.
primer set Cox1f and Cox1r generates a 450-bp fragment Thus, the processing of polycistronic mtRNAs was impaired in
whereas the primer pair Cox1f and Cox2r generates a 900-bp mitochondria with reduced PNPASE, resulting in fewer mature
fragment when tRNAser and tRNAasp are not excised from large mtRNAs and reduced ETC complexes.

458 Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc.


Figure 2. HepKO Liver Mitochondria Do Not
Efficiently Process mtRNA Precursors
(A) In organello protein synthesis. WT and HepKO
mitochondria (100 mg) were treated with micro-
coccal nuclease S7, and in organello translation
was performed using [35S]-MET. TOM40 immuno-
blot shows equivalent mitochondria in each assay.
(B) RNA was isolated from WT and HepKO liver
mitochondria followed by DNase I treatment to
remove contaminating DNA. RT-PCR was per-
formed for Cox1 and Cox2 with primers shown in
the schematic (upper) and separated on a 1.5%
agarose gel.
(C) Northern blot of mtRNA from WT and HepKO
mouse liver mitochondria using a Cox1 or Cox3
DNA probe. Asterisks mark larger precursor
mtRNAs and the arrow shows the mature mtRNA.
(D) Steady-state expression of nuclear- and mito-
chondrial-encoded proteins in WT and HepKO
liver mitochondria. Equivalent nuclear-encoded
protein expression shows that HepKO reduced
mitochondria-encoded protein expression was
not due to differing mitochondrial content between
WT and HepKO liver cells. See also Figure S4.

did not adventitiously bind RNA because


the RNA transcripts for Cox1, GAPDH,
mitochondrial 12S rRNA and mitochon-
drial tRNAtrp were not bound to PNPASE
(Figure 3B). Furthermore, PNPASE-
HisPC, but not TIM23-HisPC, bound
in vitro transcribed and imported RNase
P and MRP RNAs, but not control mito-
chondrial RNA, in cross linking IP assays
(Figure S5A) Thus, RNase P and MRP
RNA bound specifically to PNPASE.
A protein-only RNase P complex was
shown to process the 50 -terminus of a
single mitochondrial tRNAlys (Holzmann
et al., 2008; Walker and Engelke, 2008).
RNase P RNA Binds to PNPASE and May Function However, most mammalian mitochondrial tRNA genes are
in PNPASE-Dependent mtRNA Processing grouped together in the mitochondrial genome and lack an
Although an in vitro form of the RNase P enzyme can function intervening pre-sequence, causing them to be expressed with
without nucleic acid (Holzmann et al., 2008; Walker and Engelke, the ETC genes in polycistronic transcripts. Therefore, we
2008), the RNA component of RNase P localizes to mitochondria examined whether protein-only RNase P can efficiently process
(Puranam and Attardi, 2001) and has not been excluded from paired mitochondrial tRNAs, as must occur in vivo. As done
mtRNA processing in vivo (Holzmann et al., 2008). Therefore, previously (Holzmann et al., 2008), mitoplast lysates were
the abundance of RNase P RNA in HepKO liver mitochondria untreated or treated with micrococcal nuclease, followed by
was determined by RT-PCR and QPCR (Figure 3A). Reproduc- inactivation with EGTA and EDTA. tRNAlys or abutting tRNAs,
ibly, RNase P RNA was decreased by 75% in HepKO versus tRNAhistRNAser, were then added to the lysates. Nuclease-
WT liver mitochondria, suggesting that PNPASE may help import treated mitoplast lysates cleaved the 50 -precursor sequence of
and/or stabilize RNase P RNA. Therefore, we determined the single and abutting tRNA substrates as efficiently as the
whether RNase P RNA directly bound to PNPASE in HEK293 lysates without treatment, as shown before (Figure 3C). By
cells stably expressing dual-tagged PNPASE-HisPC. Isolated contrast, nuclease-treated lysates were impaired in cleaving
mitochondria were treated with nuclease and tagged PNPASE the two abutting tRNAs into individual tRNAs, strongly sug-
was purified. RNase P RNA was amplified by RT-PCR and gesting an additional nucleic acid component is required for
copurified with PNPASE (Figure 3B, lane 4). Importantly, control efficient processing. Interestingly, mitoplast lysates from
IMS-localized TIM23-HisPC in stably-expressing HEK293 cells HepKO liver showed the same defect on abutting tRNA matura-
did not bind RNase P RNA (Figure 3B, lane 2). PNPASE also tion as the nuclease-treated WT mitoplast lysates (Figure 3D).

Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc. 459


Figure 3. RNase P RNA Binds to PNPASE
and May Function in PNPASE-Dependent
tRNA Processing
(A) Left: RNA was isolated from WT and HepKO
liver mitochondria following nuclease treatment.
RT-PCR was performed with primers that
amplify nuclear-encoded RNase P RNA (212 bp).
Right: QPCR analysis of RNase P RNA expression
relative to TOM40 protein in isolated mitochondria.
(B) PNPASE-HisPC (PNP) or TIM23-HisPC (TIM23)
was purified from stably transfected HEK293 cells.
Candidate interacting RNAs that copurified in the
final eluate with PNPASE-HisPC and TIM23-
HisPC were identified by primer-specific RT-
PCR. T is the total lysate (0.3% of the reaction)
before mitochondrial purification and B is the
bound fraction. Note that only RNase P RNA
bound to PNPASE-HisPC (lane 4).
(C) Mitoplast extract (10 mg) was prepared from
nuclease-treated, intact WT liver mitochondria.
The extract was then treated with nuclease (+),
as indicated, and then inactivated with EDTA
and EGTA. The nuclease-treated or untreated
extract was incubated with abutted tRNAs
(tRNAHistRNASer) or a single tRNA (tRNALys) at
25 C for 10 or 30 min. RNA was separated on an
urea-acrylamide gel and detected by autoradiog-
raphy. A MORTALIN immunoblot shows equiva-
lent mitoplast extract in each assay.
(D) Mitoplast extract was prepared from nuclease-
treated, intact WT or HepKO liver mitochondria.
The enzymatic assay was performed as described
for panel (C). See also Figure S5.

(Figure S6). Mitochondria isolated from


WT yeast or yeast expressing human
PNPASE were incubated with in vitro tran-
scribed human RNase P RNA in import
buffer. The reaction was treated with
nuclease to remove nonimported RNA
followed by RNA isolation and RT-PCR.
RNase P RNA abundance was increased
in mitochondria containing PNPASE com-
pared to WT mitochondria (Figure 4A). This
Furthermore, the in vivo processing and separation of an RNA increase was specific for certain RNAs because cytosolic
endogenous paired tRNAhistRNAser substrate was inhibited in GAPDH RNA was not increased in the same mitochondria
HepKO compared to WT liver mitochondria, whereas a linked (Figure 4B). Osmotic shock was used to identify the location of
12 s rRNA-tRNAval substrate was processed equivalently (Fig- the imported RNase P RNA (Koehler et al., 1998). Mitochondria
ure S5B). These results, combined with prior data (Puranam and were incubated in hypotonic buffer to rupture the outer membrane
Attardi, 2001), suggest protein-only and RNase P RNA-containing and the mitoplasts (P, pellet fraction that contains the matrix and
RNase P complexes coexist in mitochondria, with efficient tRNA IM) and the supernatant (S, contains the soluble IMS contents)
processing requiring PNPASE-dependent RNase P RNA. were separated by centrifugation (Figure 4C). RNase P RNA was
detected by RT-PCR and was localized in the mitochondrial
PNPASE Augments the Import of RNase P, 5S rRNA, matrix. Detergent exposed the matrix to verify that the nuclease
and MRP RNAs into Yeast Mitochondria degraded the RNase P RNA. To confirm that osmotic shock did
Similar PNPASE complexes in yeast and mammalian mitochon- not disrupt the IM, antibodies against cytochrome b2 (cyt b2;
dria (Chen et al., 2006) (Figure S1) suggest yeast as a model for IMS) and a-ketoglutarate dehydrogenase (KDH; matrix) showed
studying the import of nuclear-encoded RNAs. Importantly, added that cyt b2 was sensitive to protease in the IMS, but KDH was
human PNPASE did not alter yeast mitochondrial morphology, resistant to protease until the IM was lysed with Triton X-100.
rate of proliferation, or extent of cell death, supporting this choice Thus, RNase P RNA import was augmented, or RNase P RNA

460 Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc.


Figure 4. PNPASE Augments RNase P, 5S
rRNA, and MRP RNA Import into Yeast Mito-
chondria
(A) Upper: In vitro transcribed human RNase P
RNA was incubated with yeast mitochondria
expressing human PNPT1 (PNP) or an empty
vector (Vec) control. Nonimported RNA was
digested with nuclease and the imported RNA
was detected by RT-PCR. PNPT1-expressing
mitochondria without added RNase P RNA was
included as a specificity control for import and
RT-PCR (lane 2: Std, 1% of the reaction). Lower:
Control showing equivalent total mitochondrial
nucleic acid in each reaction.
(B) Upper: As in panel (A), with cytosolic human
GAPDH RNA used as a substrate. Middle- Control
showing equivalent total mitochondrial nucleic
acid in each reaction. Lower: Western blot
showing PNPASE expression and PORIN immu-
noblot showing equivalent mitochondria in each
import assay.
(C) After import as in panel (A), mitochondria
were subjected to osmotic shock, fractionated
by centrifugation into soluble (S) and pellet (P)
fractions, followed by proteinase K and nuclease
additions where indicated. The pellet fraction
was solubilized with Triton X-100 to expose the
matrix. Localization was determined by RT-PCR
for RNase P RNA and immunoblot for KDH (matrix)
and cyt b2 (IMS) proteins.
(D) Upper: Radiolabeled RNase P, MRP, 5S rRNA,
and GAPDH human RNAs were in vitro transcribed
and then incubated with yeast mitochondria
expressing PNPASE or an empty vector control.
Nonimported RNA was digested with nuclease,
followed by RNA isolation, separation on a urea-
acrylamide gel, and autoradiography. Import reac-
tions were repeated with 13 and 23 amounts of
RNA. Lower: Control showing equivalent total
mitochondrial nucleic acid in each reaction.
(E) Upper: As in panel A except that the mitochon-
drial membrane potential (Dc) was dissipated prior
to import. Lower: Control showing equivalent total
mitochondrial nucleic acid in each reaction.

was stabilized, in the yeast mitochondrial matrix when exogenous biochemical assays (Portnoy et al., 2008), although the effects
PNPASE was present in the IMS. of these mutations on PNPASE in vivo are unknown. To deter-
To confirm the RT-PCR results and assay other imported mine whether the RNA import or stabilization activity of PNPASE
RNAs, we performed the in vitro RNA import assay with yeast was separable from its RNA processing activities, RNase P RNA
mitochondria and radiolabeled human RNAs (Figure 4D). Two import was studied when different PNPASE mutants were
different RNA volumes were used and the imported RNA was expressed in yeast mitochondria (Figure 5A). The point mutants
isolated and separated on a urea-acrylamide gel followed by generated and tested were based on results from Schuster and
autoradiography. RNase P, 5S rRNA, and MRP RNAs showed colleagues (Portnoy et al., 2008). Mutants D135G and S484A
augmented import or stability in mitochondria expressing lacked poly-A polymerase and RNA degradation activities
PNPASE relative to control mitochondria (Figure 4D). Again, in vitro. Mutant D544G and double mutant R445E/R446E
this increase was RNA-type specific as PNPASE did not showed enhanced in vitro poly-A polymerase activity but com-
augment GAPDH RNA levels. When the mitochondrial promised degradation activity. Of the four mutants, PNPASE
membrane potential was dissipated, the RNase P RNA level S484A and R445E/R446E supported the import or stabilization
was not increased in this assay system (Figure 4E). of RNase P RNA, whereas mutants D135G and D544G were
defective in this function (Figure 5A). The abundance of WT
PNPASE Mutations that Inactivate RNA Processing and the four mutant PNPASE proteins were similar between
Do Not Affect RNA Import or Stability yeast strains and all of the PNPASE proteins assembled into
Specific point mutations in conserved regions of PNPASE impair 240 kDa complexes without impairment (Figure 5A, lower
its RNA exonuclease or 30 poly-A polymerase activity in panel). This contrasts with the expectation that mutant D135G

Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc. 461


Figure 5. PNPASE Mutations that Inactivate
RNA Processing Do Not Affect RNA Import
or Stability
(A) Upper: Schematic for the positions of point
mutations made in the PNPASE protein. Listed
are the in vitro effects of mutations on 30 poly-
merase and RNA degrading activities, from (Port-
noy et al., 2008). Middle: Import reactions were
performed as in Figure 4A. Radiolabeled RNase
P RNA was incubated with isolated yeast mito-
chondria expressing an empty vector or the listed
PNPASE constructs. Lower panels: Immunoblot of
WT and point mutant PNPASE yeast transfectants
used in panel A import assay. A PORIN immuno-
blot confirms the colocalization of PNPASE WT
and mutants in yeast mitochondria. The assembly
state of WT and point mutant PNPASE was deter-
mined by solubilization with 1% digitonin and
separation on a 6%–16% BN gel, followed by
PNPASE immunoblot.
(B) Upper: WT and S484A PNPASE IPs from yeast
mitochondria were used to analyze RNA degrada-
tion properties. Lower: WT or S484A mutant
PNPASE was incubated with radiolabeled RNase
P RNA for 10 min at 25 C to assess degradation
activity. The asterisk marks degradation products.
(C) Left: Following in vitro import of radiolabeled
RNase P RNA and nuclease treatment to remove
nonimported RNA, mitochondria were incubated
for up to 90 min at 25 C and aliquots removed at
the indicated time points. The RNA was then
resolved by urea-acrylamide gel electrophoresis.
Right: RNase P RNA that was not degraded was
quantified using FX imager; n = 3.

similar for degradation competent WT


and incompetent mutant PNPASE pro-
teins, supporting a role for PNPASE in
augmenting the import of specific RNAs
into the mitochondrial matrix. This result
further supports PNPASE localizing to
the IMS because a greater amount of
would fail to form a trimeric complex from prior studies (Portnoy WT PNPASE imported into the matrix could cause a relative
et al., 2008). Therefore, the mitochondrial RNA import or stabili- increase in the rate of turnover of matrix localized RNase P RNA.
zation function of PNPASE is separable from its poly-A poly-
merase or exoribonuclease activities. A Predicted Stem-Loop RNA Structure Mediates
To determine whether PNPASE augmented either RNA import PNPASE-Dependent RNA Import
or stabilization in mitochondria, the enzymatic properties of Results showing PNPASE-dependent RNA import into mito-
the WT and S484A mutant protein were examined with respect chondria (Figure 3, Figure 4, and Figure 5) do not exclude an
to RNA turnover in vitro and in isolated yeast mitochondria. indirect role. To establish a direct PNPASE role, a systematic
For in vitro studies, WT and S484A PNPASE were immunoprecip- search was used to identify PNPASE-dependent RNA import
itated from yeast mitochondria and tested in an in vitro degrada- sequences. Primers were designed to generate distinct
tion assay with radiolabeled RNase P RNA (Figure 5B). WT segments of the 340 nucleotide (nt) RNase P RNA full length
PNPASE degraded the RNase P RNA, but the S484A mutant sequence. RPf1 lacked the 50 70 nt, RPf2 lacked the 50 140 nt,
was impaired, consistent with prior results (Portnoy et al., 2008). and RPr1 lacked the 30 148 nt of WT PNPT1 (Figure 6A). Import
In addition, radiolabeled RNase P RNA was imported into mito- assays were performed using full length or truncated in vitro
chondria, followed by incubation at 25 C for up to 90 min transcribed RNase P RNAs (Figures 6B and 6C). Augmented
(Figure 5C). The internalized RNase P RNA was separated on RPf1 and RPr1 import into yeast mitochondria depended upon
a urea-acyrlamide gel and quantified during this time course using PNPASE, as did the full length RNase P RNA. In striking contrast,
a phosphorimager. The rate of degradation of RNase P RNA was RPf2 was not efficiently imported into yeast mitochondria,

462 Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc.


Figure 6. A Stem-Loop Structure Mediates
PNPASE-Dependent RNA Import
(A) Schematic depiction of human RNase P RNA
and deletion fragments.
(B) Import of full length RNase P RNA into yeast
mitochondria expressing PNPASE (PNP) or
control (Vec) vectors, as in Figure 4A.
(C) Import of the indicated RNase P RNA frag-
ments.
(D) Import of RNase P RNA fragments RPf3 and
RPf4.
(E) Import of human GAPDH mRNA or GAPDH
mRNA with control (CR), MRP RNA, or RNase P
RNA 20 nt sequences fused to the 50 end, as
shown in panel (F).
(F) The secondary structures and sequences of
mitochondrial RNA targeting signals in RNase P
(RP) and MRP (MRP) RNAs. A random sequence
(CR) was used as a control.
(G) Isolated mitochondria from HEK293 cells
stably expressing IMS-localized PNPASE-HisPC
or TIM23-HisPC (control) dual-tagged proteins
were incubated with [32P]-CTP labeled CR-tRNAtrp
or RP-tRNAtrp, followed by UV-cross linking,
tag-IP, separation by SDS-PAGE, and autoradiog-
raphy. See also Figure S6.

isolated mouse liver mitochondria, with


the tRNAtrp-PNPASE interaction cap-
tured using UV-cross linking (Figure 6G).
These results strongly implicate the
structural specificity of mitochondrial
RNA import (Figure 6F) and the direct
involvement of PNPASE in this process.

PNPASE Augments RNA Import


into Yeast and Mammalian
Mitochondria In Vivo
To explore in vivo RNA import into mito-
chondria, a construct was generated in
implicating the sequence between nt 71 and 140 in PNPASE- which the human RNase P RNA was expressed from the yeast
augmented RNA import. To further refine this import signal, NME1 promoter (Figure 7A). When expressed in control yeast,
RNA sequences lacking the 50 86 (RPf3) or 102 (RPf4) nts RNase P RNA localized to mitochondria, consistent with
were generated (Figure 6A). Augmented RPf3 and RPf4 import nuclear-encoded RNA import by a PNPASE-independent mech-
into yeast mitochondria was PNPASE-dependent (Figure 6D), anism, since yeast normally lack PNPASE. By contrast, RNase P
further implicating an import signal between nt 103 and 140. RNA import increased by 2-fold in mitochondria from yeast
The most obvious, predicted secondary structure of RNase P expressing PNPASE compared with control cells (Figures 7A
RNA in this region was a 20 nt stem-loop (Figure 6F). Interest- and 7B). Importantly, an RNA similar in size to RNase P RNA
ingly, a similarly-predicted stem-loop structure was also identi- (340 nt), HOT13, that is translated in the cytosol and imported
fied in MRP RNA. To determine whether one or both stem-loop as a protein into mitochondria, was not localized to mitochon-
structures could mediate mitochondrial targeting of non- dria. Also, mitochondrial-encoded RPM1, which codes for the
imported GAPDH RNA, each 20 nt stem-loop sequence was yeast homolog of RNase P RNA, was sequestered in the mito-
fused to the 50 -terminus of the GAPDH RNA, which is not chondrion at a level equivalent to control yeast mitochondria,
imported (Figure 4D and Figure 7D). Strikingly, the RNase P as expected. These data indicate that PNPASE augments the
and MRP stem-loop structures licensed the PNPASE-depen- import of RNase P RNA into yeast mitochondria in vivo. Finally,
dent import of GAPDH RNA into yeast mitochondria (Figure 6E). replacement of the human RNase P RNA stem-loop sequence
By contrast, a control random 20nt sequence could not mediate with the 20nt random sequence blocked augmented RNase P
this import. Human mitochondrial tRNAtrp with the RNase P RNA RNA import into yeast mitochondria in vivo (Figure S7), confirm-
step-loop structure, but not tRNAtrp itself, was imported into ing the role of the stem-loop in PNPASE-regulated import.

Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc. 463


Figure 7. PNPASE Augments RNA Import
into Yeast and Mammalian Mitochondria In
Vivo
(A) Upper: Human RNase P RNA yeast expression
construct is driven by the RPM1 RNA promoter,
NME1. Lower: Mitochondria from yeast express-
ing human RNase P RNA and either PNPASE
(PNP) or an empty vector (Vec) were isolated and
treated with nuclease. RNA was then isolated
from the total cell lysate or from nuclease-treated
mitochondria (Mito) and analyzed by primer-
specific RT-PCR.
(B) QPCR for Cox1 and RNase P RNAs isolated
from mitochondria in panel (A), normalized to the
total mitochondrial RNA obtained.
(C) Radiolabeled, in vitro transcribed RNase P
RNA was imported into mitochondria from MEF
cell lines WT (expressing mouse PNPASE,
mPNP), Pnpt1 knockout (KO), PNPT1 overexpres-
sion (expressing mPNP and hPNP), or Pnpt1
knockout plus PNPT1 overexpression (expressing
hPNP). Upper panel is an immunoblot for mouse
and human PNPASE expression. Middle panel is
an immunoblot of b-ACTIN, a loading control.
Lower panel is an autoradiogram of RNase P
RNA import into isolated MEF mitochondria.
(D) Radiolabeled, in vitro transcribed RNAs were
incubated with WT or HepKO liver mitochondria
for 10 min at 25 C. Nonimported RNA was
removed with nuclease, followed by RNA isolation
and separation on a urea-acrylamide gel. Import
reactions were repeated with 13 and 23 amounts
of synthesized RNAs. TOM40 immunoblot pro-
vides a mitochondrial loading control. See also
Figure S7.

PNPASE augmented import of RNase P RNA into yeast DISCUSSION


mitochondria is nonphysiologic. Therefore, we developed WT,
PNPASE KO, WT expressing human PNPASE, and PNPASE In contrast to the protein translocation, very little is understood
KO expressing human PNPASE MEFs for import assays. about the import of specific RNAs into the mitochondrion. A
PNPASE abundance in each MEF line was confirmed by confounder is that the spectrum of RNAs imported and the
immunoblot (Figure 7C). Radiolabeled RNase P RNA was not import factors and mechanisms seem to vary greatly among
imported into mitochondria from the PNPASE KO MEFs, but different organisms. At an extreme is mammalian mitochondria,
was imported into mitochondria that contained mouse and/or in which despite strong evidence for RNA import (Alfonzo and
human PNPASE. The in vitro import of RNase P, MRP, 5S Soll, 2009; Tarassov et al., 1995), no factors have thus far been
rRNA, and GAPDH RNAs was also tested in liver mitochondria identified. Here we implicate PNPASE as the first RNA import
isolated from the HepKO mouse and WT littermates. Again, factor for mammalian mitochondria. Our results show that
RNase P, 5S rRNA, and MRP RNAs were imported into mito- PNPASE KO disrupts mitochondrial morphology and respiration
chondria expressing PNPASE, whereas cytosolic GAPDH RNA in mouse liver cells, at least partially by inhibiting the import of
was not imported (Figure 7D). As expected, more than half of RNAs that control the transcription and translation of the ETC
the imported MRP RNA was processed into the mature proteins. Our data also suggest that a nucleic acid component
130 nt form (Figure 7D, S7) (Chang and Clayton, 1987). By of the RNase P RNA processing complex, possibly RNase P
contrast, mitochondrial RNA import was severely compromised RNA (Puranam and Attardi, 2001), is imported in vivo to process
in HepKO liver mitochondria. Combined, these results strongly linked tRNAs in long mitochondrial transcripts. PNPASE medi-
support PNPASE as the first RNA import factor that mediates ated RNA delivery into the mitochondrial matrix and this import
the translocation of specific RNAs into the mammalian was augmented over background. Strikingly, PNPASE RNA
mitochondrial matrix. import and RNA processing functions were separable and

464 Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc.


predicted stem-loop structures were identified in two imported PNPASE function in chloroplasts and prokaryotes (Lisitsky
RNAs that could transfer PNPASE-dependent import potential et al., 1996; Yehudai-Resheff and Schuster, 2000) and a stem-
to nonimported RNAs. Combined, these results open a new loop structure protects the RNA from degradation by PNPASE
chapter for studies into the pathway(s) and mechanism(s) of in chloroplasts (Yehudai-Resheff et al., 2001). A detailed dissec-
RNA import into mammalian mitochondria. tion of what constitutes a trigger sequence for processing versus
A key question that this study presages is how PNPASE import activities is clearly warranted. Finally, since the overall
regulates the import of specific cytosolic RNAs. For this, the sequence homology between the identified stem-loop struc-
location of PNPASE first needs to be considered. Previously, tures on RNase P and MRP RNAs is not high, binding interactions
we localized PNPASE to the IMS. Carbonate extraction studies between specific RNAs and PNPASE may be stronger or
also indicated that PNPASE was bound to the IM facing the weaker, allowing or inhibiting detection by standard in vitro
IMS (Chen et al., 2006; Rainey et al., 2006). However, more techniques such as IP, which could make the verification of
recently, others have shown a weak interaction with the additional candidate imported RNAs challenging without the
matrix-localized RNA helicase hSUV3, suggesting a matrix functional import assay system for validation.
localization for PNPASE (Szczesny et al., 2009; Wang et al., Studies and important applications that use RNA import in
2009). Our sub-fractionation studies were highly reproducible mammalian cells have been hampered by the inconsistent
and, in contrast, we favor the idea that hSUV3 bound to PNPASE requirements between systems and their study in vitro versus
after all of the mitochondrial sub-compartments were exposed in vivo. Although the exact mechanism for how PNPASE
during purification. The methods used to identify this interaction augments and licenses RNA import is not yet known, the identifi-
do not exclude this possibility and we have already shown an cation of PNPASE represents the first receptor-like component
unanticipated interaction between PNPASE and the cytosol- that binds RNA in mammalian cells to mediate RNA import into
localized oncoprotein TCL1 using similar limited resolution the mitochondrial matrix. This finding, along with the identification
methods (French et al., 2007). Our failure to again identify a of import signal sequences, should open up additional studies to
hSUV3 and PNPASE interaction using an ultra-sensitive dual- determine what other pathway components are involved and
tag expression and purification system (Claypool et al., 2008) what the RNA sequence or structure rules tell us about how
further supports this interpretation (Figure S1). An alternative PNPASE may decipher between processing and import.
explanation that we cannot exclude or confirm with current
methodologies is that a small amount of PNPASE could get EXPERIMENTAL PROCEDURES
into the matrix and interact with hSUV3.
Protein and RNA Purification
A second interesting area opened by the current results is to
Purification of the PNPASE-HisPC protein complex was performed as before
determine how PNPASE controls RNA import into mitochondria. (Claypool et al., 2008). For protein-RNA interactions, mitochondria (1 mg/ml)
In concept, PNPASE could import RNAs from the cytosol into were solubilized in lysis buffer (300 mM NaCl, 10 mM imidazole, 10% glycerol,
the IMS and then pass this RNA to another protein or complex 0.25% Triton X-100, 2 mM DTT, 20 mM HEPES [pH 6.6]) containing protease
that would assist it through the IM into the matrix (Figure 4C). inhibitor (Roche) and RNase inhibitor (Invitrogen). Insoluble material was
Interestingly, PNPASE augments import in yeast, which does removed by spinning and extracts transferred to Eppendorf tubes. 50 ml of
Ni2+NTA resin (QIAGEN) was incubated in 1 ml lysis buffer with 100 mg/ml
not have a PNPASE homolog, indicating a distinct RNA import
ssDNA for 1h at 4 C. The resin was then mixed with the mitochondrial lysates
mechanism that PNPASE can augment directly or indepen-
in the presence of 100 mg/ml ssDNA for 1 hr at 4 C. After incubation, the resin
dently. In mouse liver and MEF mitochondria, PNPASE KO is was washed 103 with lysis buffer containing RNase inhibitor. The protein-RNA
incomplete because cells without PNPASE are nonviable, so it complex was eluted with elution buffer (300 mM NaCl, 10 mM imidazole, 10%
is unclear whether it is absolutely essential for all RNA import glycerol, 0.25% Triton X-100, 20 mM citrate [pH 5.5]) containing RNase
or whether the amount detected in import assays is still mediated inhibitor. RNA was isolated from the eluate using TRIzol reagent (Invitrogen).
by the minimal residual amount of PNPASE present required
Isolation of Mitochondrial RNA and DNA
for cell survival. Furthermore, it is not clear if this imported
Mitochondria (1 mg/ml) were treated with 25 mg/ml of micrococcal nuclease S7
RNA in PNPASE KO mitochondria is stuck in the IMS and not in nuclease buffer (0.6 M Sorbitol, 20 mM MgCl2, 5 mM CaCl2, 20 mM Tris
in the matrix, which could provide further insight for the detailed [pH 8.0]) for 30 min at 27 C. The reaction was stopped by addition of 20 mM
function of PNPASE in RNA import. EGTA. Mitochondria were collected and solubilized in SDS buffer (100 mM
PNPASE has two external domains (KH and S1) that bind RNA NaCl, 1% SDS, 20 mM Tris pH 7.4) at 65 C for 5 min. RNA was purified using
near the opening of a central processing pore in a trimeric TRIzol reagent, and treated with RNase-free DNase I (Roche) for 1h at 37 C.
DNase I was inactivated by heating at 65 C for 10 min. Phenol-chloroform
complex (Carpousis, 2002; Symmons et al., 2000). It is not clear
(EM Science) extractions were used for DNA purification from the mitochon-
whether the same domains are used indiscriminately or in some drial lysates.
distinct manner to trigger PNPASE RNA processing versus
import functions. It is possible that the stem-loop structures In Vitro Transcription
identified in RNase P and MRP RNAs interact with PNPASE RNAs were synthesized as before (Portnoy et al., 2008). For radiolabeled RNA
in a manner that triggers only import rather than processing. synthesis, [32P]-CTP (MP Biochemicals) was incorporated. The RNAs were
purified using TRIzol reagent.
Interestingly, GAPDH RNA can be a target of PNPASE degrada-
tion activity in vitro (French et al., 2007), although when either
RNA Import Assay
of the identified stem-loop structures is appended to the Yeast mitochondria were isolated from cells grown in selection medium until
50 -terminus, GAPDH RNA is efficiently imported into mitochon- stationary phase and mammalian mitochondria were isolated as previously
dria (Figure 6E). Indeed, RNA structural elements regulate described (Chen et al., 2006; Rainey et al., 2006). In vitro RNA import assays

Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc. 465


were performed in a 200-ml volume containing 0.5 mg of RNA, 100 mg of mito- REFERENCES
chondria, 0.6 M sorbitol, 2 mM KH2PO4, 50 mM KCl, 10 mM MgCl2, 2.5 mM
EDTA, 5 mM L-methionine, 1 mg/ml BSA, 5 mM ATP, 2 mM DTT, 5 mM Alfonzo, J.D., and Soll, D. (2009). Mitochondrial tRNA import–the challenge to
NADH, 50 mM HEPES, [pH 7.1], at RT for 10 min. Mitochondria were spun understand has just begun. Biol. Chem. 390, 717–722.
at 11,000 x g for 5 min and washed once with wash buffer (0.6 M sorbitol, Bonawitz, N.D., Clayton, D.A., and Shadel, G.S. (2006). Initiation and beyond:
20 mM Tris, [pH 8.0]). Mitochondria were spun again and resuspended in multiple functions of the human mitochondrial transcription machinery.
200 ml nuclease buffer containing 25 mg/ml of micrococcal nuclease S7 and Mol. Cell 24, 813–825.
incubated for 30 min at 27 C. Mitochondria were collected and solubilized in
Carpousis, A.J. (2002). The Escherichia coli RNA degradosome: structure,
SDS buffer at 65 C for 5 min. RNA was purified using TRIzol reagent. For
function and relationship in other ribonucleolytic multienzyme complexes.
import into mammalian mitochondria, 0.25 M sucrose instead of 0.6 M sorbitol,
Biochem. Soc. Trans. 30, 150–155.
and 20 mM succinate instead of 5 mM NADH, were used. For import with
radiolabeled RNA, the purified RNAs were analyzed by urea-acrylamide gel Chacinska, A., Koehler, C.M., Milenkovic, D., Lithgow, T., and Pfanner, N.
and autoradiography. (2009). Importing mitochondrial proteins: machineries and mechanisms. Cell
138, 628–644.
Chang, D.D., and Clayton, D.A. (1987). A mammalian mitochondrial RNA
RNA Degradation Assay processing activity contains nucleus-encoded RNA. Science 235, 1178–1184.
The RNA processing activity of WT and mutant PNPASE was done as before Chen, H.W., Koehler, C.M., and Teitell, M.A. (2007). Human polynucleotide
(Portnoy et al., 2008). [32P]-RNA was incubated with the corresponding phosphorylase: location matters. Trends Cell Biol. 17, 600–608.
proteins in buffer E (20 mM HEPES, [pH 7.9], 60 mM KCl, 12.5 mM MgCl2, Chen, H.W., Rainey, R.N., Balatoni, C.E., Dawson, D.W., Troke, J.J., Wasiak,
0.1 mM EDTA, 2 mM DTT, and 17% glycerol, 0.1 mM Pi) at 25 C for 5 min. S., Hong, J.S., McBride, H.M., Koehler, C.M., Teitell, M.A., and French, S.W.
Following incubation, the RNA was isolated and analyzed by urea-acrylamide (2006). Mammalian polynucleotide phosphorylase is an intermembrane
gel and autoradiography. space RNase that maintains mitochondrial homeostasis. Mol. Cell. Biol. 26,
8475–8487.
Claypool, S.M., Oktay, Y., Boontheung, P., Loo, J.A., and Koehler, C.M. (2008).
Additional Procedures
Cardiolipin defines the interactome of the major ADP/ATP carrier protein of the
Osmotic shock was performed by incubating mitochondria for 30 min on ice in
mitochondrial inner membrane. J. Cell Biol. 182, 937–950.
0.03 M sorbitol and 20 mM HEPES-KOH, (pH 7.4) (Claypool et al., 2008). Blue
native gel electrophoresis was performed on a 6%–16% linear polyacrylamide Diaz, F., Garcia, S., Hernandez, D., Regev, A., Rebelo, A., Oca-Cossio, J., and
gradient using 50 mg of digitonin solubilized material (Chen et al., 2006). Moraes, C.T. (2008). Pathophysiology and fate of hepatocytes in a mouse
Northern blotting was performed as previously described (Tollervey, 1987). model of mitochondrial hepatopathies. Gut 57, 232–242.
Total mtRNA was separated on a 12% agarose-formaldehyde gel and Doersen, C.J., Guerrier-Takada, C., Altman, S., and Attardi, G. (1985).
transferred to a nylon membrane. Hybridization was carried out with Characterization of an RNase P activity from HeLa cell mitochondria. Compar-
[32P]-dCTP (MP Biochemicals) labeled DNA probes. In organello protein ison with the cytosol RNase P activity. J. Biol. Chem. 260, 5942–5949.
synthesis assays were performed as before with minor changes (Stuart and Duchene, A.M., Pujol, C., and Marechal-Drouard, L. (2009). Import of tRNAs
Koehler, 2007). 100 mg mouse liver mitochondria were incubated in 100 ml and aminoacyl-tRNA synthetases into mitochondria. Curr. Genet. 55, 1–18.
translation buffer (250 mM sucrose, 100 mM KCl, 1 mM MgCl2, 10 mM Tris
Entelis, N.S., Kolesnikova, O.A., Martin, R.P., and Tarassov, I.A. (2001). RNA
(pH 7.4), 10 mM K2HPO4 (pH 7.4), 10 mM glutamate, 10 mM malate, 5mM
delivery into mitochondria. Adv. Drug Deliv. Rev. 49, 199–215.
NADH, 1 mM ADP, 1 mg/ml BSA, 100 mg/ml emetine, 100 mg/ml cyclohexi-
mide, and 30 mM of amino acid mix without methionine) with 5 ml of L-[35S] French, S.W., Dawson, D.W., Chen, H.W., Rainey, R.N., Sievers, S.A.,
methionine (MP Biochemicals) at 37 C for 30 min. The mitochondria were Balatoni, C.E., Wong, L., Troke, J.J., Nguyen, M.T., Koehler, C.M., and Teitell,
then precipitated and proteins resolved by 12% SDS PAGE. M.A. (2007). The TCL1 oncoprotein binds the RNase PH domains of the
PNPase exoribonuclease without affecting its RNA degrading activity. Cancer
Lett. 248, 198–210.

SUPPLEMENTAL INFORMATION Hollingsworth, M.J., and Martin, N.C. (1986). RNase P activity in the mitochon-
dria of Saccharomyces cerevisiae depends on both mitochondrion and
Supplemental Information includes Extended Experimental Procedures and nucleus-encoded components. Mol. Cell. Biol. 6, 1058–1064.
seven figures and can be found with this article online at doi:10.1016/j.cell. Holzmann, J., Frank, P., Loffler, E., Bennett, K.L., Gerner, C., and Rossmanith,
2010.06.035. W. (2008). RNase P without RNA: identification and functional reconstitution of
the human mitochondrial tRNA processing enzyme. Cell 135, 462–474.
Koehler, C.M., Jarosch, E., Tokatlidis, K., Schmid, K., Schweyen, R.J., and
ACKNOWLEDGMENTS Schatz, G. (1998). Import of mitochondrial carriers mediated by essential
proteins of the intermembrane space. Science 279, 369–373.
We thank Michelle Husain for expertise in TEM. Supported by NIH grants
Kolesnikova, O.A., Entelis, N.S., Jacquin-Becker, C., Goltzene, F., Chrzanow-
R01GM061721 and R01GM073981 (C.M.K.), R01CA90571 (M.A.T.) and
ska-Lightowlers, Z.M., Lightowlers, R.N., Martin, R.P., and Tarassov, I. (2004).
PN2EY018228 (Roadmap for Medical Research Nanomedicine Initiative)
Nuclear DNA-encoded tRNAs targeted into mitochondria can rescue a
(M.A.T.), and K22CA120147 (S.W.F.), the Intramural Research Program of
mitochondrial DNA mutation associated with the MERRF syndrome in cultured
the NIH, National Institute of Allergy and Infectious Diseases (H.C.M. III), the
human cells. Hum. Mol. Genet. 13, 2519–2534.
Muscular Dystrophy Association 022398 (C.M.K.), the American Heart Associ-
Kren, B.T., Wong, P.Y., Sarver, A., Zhang, X., Zeng, Y., and Steer, C.J. (2009).
ation 0640076N (C.M.K.), and the California Institute of Regenerative Medicine
MicroRNAs identified in highly purified liver-derived mitochondria may play
(RS1-00313 and RB1-01397, M.A.T.). C.M.K. is an Established Investigator of
a role in apoptosis. RNA Biol. 6, 65–72.
the American Heart Association and M.A.T. is a Scholar of the Leukemia and
Lymphoma Society. Lisitsky, I., Klaff, P., and Schuster, G. (1996). Addition of destabilizing poly
(A)-rich sequences to endonuclease cleavage sites during the degradation
Received: December 22, 2009 of chloroplast mRNA. Proc. Natl. Acad. Sci. USA 93, 13398–13403.
Revised: March 20, 2010 Mandel, H., Hartman, C., Berkowitz, D., Elpeleg, O.N., Manov, I., and Iancu,
Accepted: May 13, 2010 T.C. (2001). The hepatic mitochondrial DNA depletion syndrome: ultrastruc-
Published: August 5, 2010 tural changes in liver biopsies. Hepatology 34, 776–784.

466 Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc.


Mukherjee, S., Basu, S., Home, P., Dhar, G., and Adhya, S. (2007). Necessary Symmons, M.F., Jones, G.H., and Luisi, B.F. (2000). A duplicated fold is the
and sufficient factors for the import of transfer RNA into the kinetoplast structural basis for polynucleotide phosphorylase catalytic activity, processiv-
mitochondrion. EMBO Rep. 8, 589–595. ity, and regulation. Structure 8, 1215–1226.

Portnoy, V., Palnizky, G., Yehudai-Resheff, S., Glaser, F., and Schuster, G. Symmons, M.F., Williams, M.G., Luisi, B.F., Jones, G.H., and Carpousis, A.J.
(2008). Analysis of the human polynucleotide phosphorylase (PNPase) reveals (2002). Running rings around RNA: a superfamily of phosphate-dependent
differences in RNA binding and response to phosphate compared to its RNases. Trends Biochem. Sci. 27, 11–18.
bacterial and chloroplast counterparts. RNA 14, 297–309. Szczesny, R.J., Borowski, L.S., Brzezniak, L.K., Dmochowska, A., Gewartow-
ski, K., Bartnik, E., and Stepien, P.P. (2009). Human mitochondrial RNA
Postic, C., and Magnuson, M.A. (2000). DNA excision in liver by an albumin-
turnover caught in flagranti: involvement of hSuv3p helicase in RNA surveil-
Cre transgene occurs progressively with age. Genesis 26, 149–150.
lance. Nucleic Acids Res. 38, 279–298.
Puranam, R.S., and Attardi, G. (2001). The RNase P associated with HeLa cell Tarassov, I., Entelis, N., and Martin, R.P. (1995). An intact protein translocating
mitochondria contains an essential RNA component identical in sequence to machinery is required for mitochondrial import of a yeast cytoplasmic tRNA. J.
that of the nuclear RNase P. Mol. Cell. Biol. 21, 548–561. Mol. Biol. 245, 315–323.
Rainey, R.N., Glavin, J.D., Chen, H.W., French, S.W., Teitell, M.A., and Tollervey, D. (1987). A yeast small nuclear RNA is required for normal process-
Koehler, C.M. (2006). A new function in translocation for the mitochondrial ing of pre-ribosomal RNA. EMBO J 6, 4169–4175.
i-AAA protease Yme1: import of polynucleotide phosphorylase into the inter- Walker, S.C., and Engelke, D.R. (2008). A protein-only RNase P in human mito-
membrane space. Mol. Cell. Biol. 26, 8488–8497. chondria. Cell 135, 412–414.

Rubio, M.A., Rinehart, J.J., Krett, B., Duvezin-Caubet, S., Reichert, A.S., Soll, Wang, D.D., Shu, Z., Lieser, S.A., Chen, P.L., and Lee, W.H. (2009).
D., and Alfonzo, J.D. (2008). Mammalian mitochondria have the innate ability to Human Mitochondrial SUV3 and Polynucleotide Phosphorylase Form a
import tRNAs by a mechanism distinct from protein import. Proc. Natl. Acad. 330-kDa Heteropentamer to Cooperatively Degrade Double-stranded RNA
Sci. USA 105, 9186–9191. with a 30 -to-50 Directionality. J. Biol. Chem. 284, 20812–20821.
Yehudai-Resheff, S., Hirsh, M., and Schuster, G. (2001). Polynucleotide
Smirnov, A.V., Entelis, N.S., Krasheninnikov, I.A., Martin, R., and Tarassov, I.A. phosphorylase functions as both an exonuclease and a poly(A) polymerase
(2008). Specific features of 5S rRNA structure - its interactions with macromol- in spinach chloroplasts. Mol. Cell. Biol. 21, 5408–5416.
ecules and possible functions. Biochemistry (Mosc.) 73, 1418–1437.
Yehudai-Resheff, S., and Schuster, G. (2000). Characterization of the E.coli
Stuart, R.A., and Koehler, C.M. (2007). In vitro analysis of yeast mitochondrial poly(A) polymerase: nucleotide specificity, RNA-binding affinities and RNA
protein import. Curr. Protoc. Cell Biol. Chapter 11, Unit 11.19. structure dependence. Nucleic Acids Res. 28, 1139–1144.

Cell 142, 456–467, August 6, 2010 ª2010 Elsevier Inc. 467


Plzf Regulates Germline Progenitor
Self-Renewal by Opposing mTORC1
Robin M. Hobbs,1 Marco Seandel,2 Ilaria Falciatori,3 Shahin Rafii,3 and Pier Paolo Pandolfi1,*
1Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess

Medical Center, Harvard Medical School, Boston, MA 02115, USA


2Department of Surgery
3Howard Hughes Medical Institute, Department of Genetic Medicine

Weill Cornell Medical College, New York, NY 10065, USA


*Correspondence: ppandolf@bidmc.harvard.edu
DOI 10.1016/j.cell.2010.06.041

SUMMARY activated mTORC1 is detrimental to stem cell maintenance


(Castilho et al., 2009; Gan and DePinho, 2009; Yilmaz et al.,
Hyperactivity of mTORC1, a key mediator of cell 2006). It is proposed that inappropriate mTORC1 activation
growth, leads to stem cell depletion, although the drives stem cell depletion through aberrant translation of down-
underlying mechanisms are poorly defined. Using stream targets and subsequent activation of tumor suppressive/
spermatogonial progenitor cells (SPCs) as a model fail-safe mechanisms resulting in cellular senescence or apo-
system, we show that mTORC1 impairs stem cell ptosis (Ito et al., 2009). However, the molecular mechanisms
and targets of mTORC1 in this context are currently unknown.
maintenance by a negative feedback from mTORC1
Interestingly, inhibition of mTORC1 also extends organism life-
to receptors required to transduce niche-derived span (Harrison et al., 2009; Schieke and Finkel, 2006), consistent
signals. We find that SPCs lacking Plzf, a transcrip- with the notion that declining stem cell potential underlies aging
tion factor essential for SPC maintenance, have (Rossi et al., 2008).
enhanced mTORC1 activity. Aberrant mTORC1 acti- Undifferentiated germline cells of the testis (spermatogonial
vation in Plzf / SPCs inhibits their response to progenitor cells; SPCs) are formed from gonocytes during post-
GDNF, a growth factor critical for SPC self-renewal, natal development of the mouse testis and possess self-renewal
via negative feedback at the level of the GDNF potential (de Rooij and Russell, 2000). A major advance in the
receptor. Plzf opposes mTORC1 activity by inducing study of male germline biology was the development of culture
expression of the mTORC1 inhibitor Redd1. Thus, we systems allowing long-term SPC expansion in vitro while main-
identify the mTORC1-Plzf functional interaction as taining stem cell potential. Key to this was the observation that
mice heterozygous for the glial cell-derived neurotrophic factor
a critical rheostat for maintenance of the spermato-
(GDNF) cytokine gene had a depletion of SPC activity (Meng
gonial pool and propose a model whereby negative et al., 2000). GDNF is produced by Sertoli cells within the testis,
feedback from mTORC1 to the GDNF receptor and signals via the GFRa1/c-Ret receptor to promote SPC self-
balances SPC growth with self-renewal. renewal and growth through activation of Src family kinases and
Akt (Lee et al., 2007b; Oatley et al., 2007). Culture of SPCs with
INTRODUCTION GDNF plus a variety of additional factors (including basic fibro-
blast growth factor; bFGF) preserves self-renewal capabilities
Maintenance of a wide array of adult tissues is dependent on the and allows essentially unlimited cell expansion while maintaining
presence of a resident stem cell pool with self-renewal potential in vivo differentiation potential; assessed by the ability to repopu-
that generates differentiating progeny. Factors regulating the late depleted recipient testis (Kanatsu-Shinohara et al., 2003;
balance between stem cell self-renewal and differentiation Kubota et al., 2004; Seandel et al., 2007). Although some cellular
ensure tissue homeostasis whereas disruption of these regula- signaling pathways involved in SPC self-renewal have been
tory mechanisms can lead to tissue degeneration or cancer (Ito described, it remains unclear how SPCs integrate signals from
et al., 2009). One factor central to stem cell homeostasis is general mitogenic stimuli with those required for self-renewal
mammalian TOR complex 1 (mTORC1), a signaling complex to balance stem cell maintenance and differentiation.
that promotes protein translation and cell growth by phosphory- A limited number of cell intrinsic factors have also been impli-
lating components of the translation machinery (Ma and Blenis, cated in SPC function, foremost among which is promyelocytic
2009). mTORC1 is regulated in response to diverse stimuli leukemia zinc finger (PLZF). PLZF was identified from the trans-
including nutrient availability, energy status, growth factors, location breakpoint in t(11;17) acute promyelocytic leukemia
and cellular stress. Persistent mTORC1 activation in certain (Chen et al., 1993) and encodes a transcription factor belonging
tissues leads to increased proliferation but subsequent exhaus- to the POZ-Krüppel (POK) family. PLZF binds DNA through
tion of the stem cell compartment, demonstrating that aberrantly carboxy-terminal Krüppel-type zinc fingers and recruits histone

468 Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc.


deacetylases (HDACs) via an amino-terminal POZ domain (David compared the ability of avneg Thy-1low c-Kitneg and unfractio-
et al., 1998). Recruitment of HDACs to target promoters can nated cells to repopulate recipient depleted testis (Ogawa
result in gene repression although PLZF is also able to activate et al., 1997). Donor cells were isolated from transgenic mice
gene expression (Doulatov et al., 2009; Labbaye et al., 2002). expressing GFP from the b-actin promoter, allowing visualization
Male mice lacking Plzf expression undergo progressive germ of donor-derived colonies (Figure 1D, left). Numbers of GFP-
cell loss and testis atrophy with age causing infertility (Buaas positive colonies were scored 2 months posttransplant (Fig-
et al., 2004; Costoya et al., 2004). Plzf is expressed by SPCs ure 1D, right), representing numbers of engrafted stem cells.
and is needed in a cell autonomous fashion for maintenance Stem cell activity was enriched 25-fold in the avneg Thy-1low
of the germ lineage. A male patient with biallelic PLZF loss-of- c-Kitneg population compared to unfractionated cells. As the
function and gonad hypoplasia has been recently reported avneg Thy-1low c-Kitneg fraction represents 3%–5% of total
(Fischer et al., 2008), emphasizing the role played by PLZF in cells (1/25th of testis cellularity) from 10 to 14 days postnatal
germ cell biology. testis (Figures 1B, 1C, and 1G), a 25-fold enrichment of stem
SPC maintenance is dependent on Plzf plus key growth cell activity indicates that almost all stem cells are contained
factors such as GDNF. As mTORC1 is activated in response to within this fraction. Given the number of colonies obtained
growth factor signaling we hypothesized that both Plzf and from avneg Thy-1low c-Kitneg cells (Figure 1D) and an estimated
mTORC1 are involved in the GDNF response of SPCs and that 10% engraftment efficiency (Nagano et al., 1999), there are
Plzf and mTORC1 may crosstalk. To assess the roles of Plzf 1265 stem cells per 105 cells in this fraction (1 in 80 cells
and mTORC1 in GDNF-dependent SPC maintenance, we devel- have stem cell potential).
oped systems for isolation and culture of SPCs from wild-type We next analyzed prepubertal Plzf +/+ and Plzf / testis by
(WT) and Plzf / prepubertal mice (subsequent to formation flow-cytometry to determine if Plzf / mice display aberrant
of the SPC pool and prior to overt germ cell depletion in the SPC activity before overt germ cell depletion. Although the char-
Plzf / mouse). We find that Plzf opposes mTORC1 activity acteristic av/Thy-1 flow profile was present in juvenile Plzf /
and define a cellular signaling network controlling SPC homeo- testis (2 and 3 weeks postnatal), the avneg Thy-1low population
stasis whereby mTORC1 integrates mitogenic signals received was depleted relative to the WT (Figure 1E and Figure S1C)
by SPCs and determines their sensitivity to self-renewal stimuli. and the percentage of c-Kitpos cells within the Plzf / avneg
Thy-1low fraction was increased (Figure 1F and Figure S1D).
RESULTS The decreased frequency and numbers of avneg Thy-1low c-Kitneg
cells in Plzf / testis (Figure 1G) confirms that Plzf loss is detri-
Isolation, Culture, and Comparative Analysis mental to SPCs whereas the increase in c-Kitpos cells within the
of Plzf +/+ and Plzf / SPCs avneg Thy-1low fraction is suggestive of increased differentiation
The testis is composed of a heterogeneous mix of mitotic and commitment. As Plzf directly represses c-Kit expression
meiotic germ cells plus multiple types of somatic cells. SPCs (Filipponi et al., 2007), this can be reflective of a de-repression
are rare within the testis and systems for their purification are of the c-Kit locus. However, the Plzf / avneg Thy-1low fraction
poorly developed. We therefore sought to take advantage of still contains c-Kitneg cells (Figure 1F and Figure S1D), indicating
the expression of Plzf in SPCs to enrich for this cell type. Plzf- that aberrant c-Kit expression is unlikely to fully explain the
expressing cells were identified from juvenile testis by intracel- Plzf / phenotype. Additionally, both Plzf +/+ and Plzf / avneg
lular staining and flow cytometry using a newly developed Thy-1low c-Kitneg cells show substantial enrichment in expres-
monoclonal antibody (Figure 1A). Our aim was to correlate the sion of the SPC-associated factors Pou5f1/Oct4, Ngn3, Bcl6b,
Plzf-expressing population to sets of cell surface markers that and Pou3f1 (Figure 1H and Figure S1E) (Oatley et al., 2006;
would allow subsequent purification of those live cells. By adapt- Ohbo et al., 2003; Wu et al., 2010; Yoshida et al., 2006), confirm-
ing markers able to enrich for stem cell activity from cryptorchid ing the identity of this fraction. We also noted that Plzf / avneg
testis (Kubota et al., 2003; Shinohara et al., 2000), we found Thy-1low cells had decreased levels of b1 and a6 integrins but
that the Plzf-positive population was av-integrin negative and equivalent levels of CD9 compared to controls (Figures S1F
expressed low levels of Thy-1 (Figure 1B). Negative selection and S1G) (Kanatsu-Shinohara et al., 2004b; Shinohara et al.,
for av-integrin combined with Thy-1 positive selection allowed 1999). Our data indicate that SPCs from juvenile Plzf / mice
isolation of Plzf-positive cells and minimized somatic cell have an impaired ability to maintain an undifferentiated state,
contamination (Figures S1A and S1B available online) (Virtanen which precedes overt germ cell loss.
et al., 1986). c-Kit expression is associated with SPC differentia- We next attempted in vitro culture of isolated Plzf +/+ and
tion (Schrans-Stassen et al., 1999) and the avneg Thy-1low popu- Plzf / SPCs, adapting previously developed culture systems
lation was composed primarily of c-Kit negative cells with (Kubota et al., 2004; Seandel et al., 2007). As anticipated, germ
a smaller c-Kit positive fraction (Figure 1C, left). The c-Kitneg cells cell colony-forming activity under SPC culture conditions was
were largely in G1/G0 phase of cell cycle whereas the c-Kitpos essentially entirely contained within the avneg Thy-1low fraction
population contained more cells in S plus G2 phases (Figure 1C, (Figure S2A). We successfully derived SPC lines from both
right). As SPCs are more quiescent than differentiating sper- Plzf +/+ and Plzf / avneg Thy-1low cells, which grew as discrete
matogonia (Takubo et al., 2008), the avneg Thy-1low population colonies on mouse embryonic fibroblast (MEF) feeder cells
contains both SPCs (c-Kitneg, quiescent) and cells undergoing (Figure 2A) and maintained their cell surface marker identity
differentiation (c-Kitpos, proliferating). To confirm that the avneg during exponential growth over 1 year of culture (Figure S2B).
Thy-1low c-Kitneg fraction was enriched for stem cells, we WT SPC lines expressed Plzf (Figure 2B) whereas Plzf / SPC

Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc. 469


+/+
Figure 1. Isolation and Analysis of Plzf
and Plzf / SPCs
(A) Detection of Plzf-expressing spermatogonia
from juvenile testis (10–14 days postnatal) by intra-
cellular staining and flow cytometry using anti-Plzf
antibody.
(B) Plzf expressing cells correlate to a discrete av-
integrin negative, Thy-1 low population by flow cy-
tometry. CD45 marker excludes contaminating
leukocytes.
(C) Left panel: flow cytometric analysis of c-Kit
within the Plzf-expressing av-integrin negative,
Thy-1 low population. Right panels: cell cycle
status of c-Kit negative and positive populations
plus percentage of cells in G1/S/G2 cell cycle
phases from a representative sample.
(D) Left panels: seminiferous tubules repopulated
with donor GFP-positive av integrin negative,
Thy-1 low, c-Kit negative cells in testis transplant
assay. Fluorescent image (top) and brightfield
image of same recipient testis (bottom) are shown.
Right panel: numbers of GFP positive colonies ob-
tained from av integrin negative, Thy-1 low, c-Kit
negative (sorted) and unsorted testis cells in recip-
ient testes 2 months posttransplant. Data is pre-
sented as mean number of colonies per 1 3 105
donor cells together with standard error of the
mean (SEM). Fold enrichment of stem cell activity
in sorted populations is indicated (***p < 0.0001).
Values are averaged from two independent exper-
iments. Thirteen recipient testes were analyzed for
sorted populations and 12 for unsorted.
(E) Representative av-integrin/Thy-1 flow profiles
of Plzf +/+ and Plzf / littermate mouse testes at
2 weeks postnatal age. The av-integrin negative,
Thy-1 low gate, containing Plzf-positive cells in
WT testis is indicated.
(F) Representative flow cytometry analysis of c-Kit
expression within the av-integrin negative, Thy-1
low testis cell fractions from (E).
(G) Quantification of frequency and absolute
numbers of av integrin negative, Thy-1 low, c-Kit
negative testis cells from mice of the indicated
postnatal ages and Plzf genotypes (n = 3 per geno-
type and age group, **p < 0.002, *p < 0.02).Graphs
represent mean values ± SEM.
(H) Quantitative RT-PCR analysis of Pou5f1 mRNA
expression in testis cell populations from juvenile
Plzf +/+ and Plzf / littermate mice: total (unfrac-
tionated testis cells), SPCs (av-integrin negative, Thy-1 low, c-Kit negative), early differentiating spermatogonia (av-integrin negative, Thy-1 low, c-Kit positive),
late differentiating spermatogonia (av-integrin negative, Thy-1 negative, c-Kit positive), and somatic cells (av-integrin positive). mRNA levels are normalized to
those of b-actin and standard deviations from duplicate reactions are shown.
See also Figure S1.

lines were more difficult to establish (not shown) consistent with Shinohara et al., 2004a; Seandel et al., 2007). We observed
a role for Plzf in SPC function. Early passages of Plzf / SPCs spontaneous formation of MASC colonies from two of seven
had a slower growth rate than WT cells (Figures S2C and S2D) WT lines (Figure S2E). MASC formation was not observed in
but could be maintained long-term and growth became more four Plzf / SPC lines, possibly indicating a defective capability
comparable to WT cells at later passages (Figure 5C). The ability to generate MASCs. The multipotent capabilities of MASC lines
to culture Plzf / SPCs seemed counterintuitive, but the gener- were verified by teratoma formation assay (Figure S2F). On SPC
ation of these cultures became critical for further dissection of to MASC conversion, expression of the pluripotency-associated
Plzf function and to the solution of this apparent contradiction. factors Pou5f1/Oct4, Sox2 and Nanog were substantially
An additional indicator of SPC potential is the ability to derive increased whereas Plzf expression was lost (Figure S2G and
embryonic stem-like cells from the cultured lines (multipotent data not shown) (Seandel et al., 2007). Formation of MASCs indi-
adult spermatogonial-derived stem cells; MASCs) (Kanatsu- cates that SPC potential is maintained in our culture system,

470 Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc.


Figure 2. Increased mTORC1 Pathway
Activity in Plzf / SPCs
(A) SPC lines established from culture of av-integ-
rin negative, Thy-1 low cells from juvenile Plzf +/+
and Plzf / littermate mice immunostained for
the germ cell marker Mvh and counterstained
with DAPI to detect DNA. Scale bar represents
100 mm.
(B) Cultured SPCs of the indicated genotype
analyzed for Plzf expression by intracellular stain-
ing and flow cytometry (left panel) and by western
blot (right panel). The germ cell marker Mvh plus
actin are used as loading controls.
(C) Analysis of SPC size by flow cytometry.
Forward-scatter (FSC) profiles of two indepen-
dently derived sets of littermate Plzf +/+ and
Plzf / SPC lines (1 and 2) are shown.
(D) Quantification of flow cytometry profiles from
(C) showing averaged mean FSC with standard
deviation and p value.
(E) Control (DMSO) and rapamycin-treated Plzf +/+
and Plzf / SPCs were fixed and permeabilized
48 hr post-treatment and levels of Phospho-
RPS6 analyzed by flow cytometry.
(F) Normalization of increased Plzf / SPC size by
mTORC1 inhibition. FSC profiles of Plzf / SPCs
treated with DMSO (control) or rapamycin for
48 hr compared to untreated Plzf +/+ SPCs.
(G) Overlay of representative av-integrin negative,
Thy-1 low, c-Kit negative testis cell fraction FSC
profiles from Plzf +/+ and Plzf / littermate mice
treated from 10 days postnatal age for a period
of 1 week with vehicle or rapamycin.
(H) Quantification of relative mean FSC values of
av-integrin negative, Thy-1 low, c-Kit negative
testis cells of mice treated as in (G) (n = 4 per geno-
type and treatment group, *p < 0.05, **p < 0.01).
Horizontal bars represent mean values.
See also Figure S2.

whereas the apparent inability of Plzf / SPCs to form MASCs elevated mTORC1 activity was responsible for the size increase
can be consistent with their defective function. (Figure 2F). Rapamycin was confirmed to inhibit RPS6 phos-
phorylation in Plzf / SPCs (Figure 2E). Increased mTORC1
Plzf / SPCs Show Enhanced mTORC1 Activity activity in Plzf / SPCs was associated with elevated levels of
From analysis of cultured SPC lines, we noticed that Plzf / cells cellular protein (Figure S2H), consistent with the role of mTORC1
were physically larger than WT SPCs, measured by the FSC in regulating protein translation. Importantly, freshly isolated
parameter of flow cytometry (Figures 2C and 2D). Alterations in Plzf / SPCs were also physically larger (Figures 2G and 2H)
cell size are associated with changes in activity of mTORC1 and rapamycin treatment of Plzf / mice normalized SPC size
acting through its downstream targets S6Kinase1 (S6K1), which (Figures 2G and 2H). Together, our data indicate that Plzf inhibits
phosphorylates ribosomal S6 protein (RPS6), and 4EBP1 (Fingar mTORC1 activation in SPCs.
et al., 2002; Ruvinsky et al., 2005). Given the role of mTORC1 in
hematopoietic stem cells (Gan and DePinho, 2009), we consid- Growth Factor-Mediated Regulation of mTORC1 in SPCs
ered that aberrant mTORC1 activity in Plzf / SPCs could Given the possibility that mTORC1 activity can regulate SPC
contribute to their defective maintenance. Importantly, Plzf / function, we next defined key regulatory inputs of mTORC1 in
SPCs had elevated levels of phosphorylated RPS6 compared WT SPCs. Growth factor signaling represents a major activating
to WT cells, confirming increased mTORC1 activity (Figure 2E input of mTORC1 (Ma and Blenis, 2009; Shaw and Cantley,
and Figure 3C). Inhibition of mTORC1 with rapamycin decreased 2006) and mTORC1 inhibition interferes with mitogenic stimuli
the size of Plzf / SPCs to that of WT cells suggesting that causing cell cycle arrest (Brown et al., 1994). Indeed, rapamycin

Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc. 471


Figure 3. Upstream Pathways and Growth Factors
Regulating mTORC1 in SPCs
(A) WT SPCs fed with complete SPC medium containing
inhibitors to the indicated signaling pathways were har-
vested 5 hr after treatment and analyzed by western blot
for the indicated proteins and phospho (P)-proteins.
(B) WT SPCs were starved overnight in basal SPC medium
lacking cytokine supplements. The indicated cytokines
were then added and SPCs harvested 20 min later then
analyzed by western blot. Cytokines were used at the
concentrations present in complete SPC medium. Neure-
gulin1 (NRG1) is not a standard supplement for SPC
medium although SPCs are responsive to it (see text).
(C) Western blot analysis of two independently derived
sets of littermate Plzf +/+ and Plzf / SPC lines (1 and 2)
under steady-state conditions is shown to the left. Quanti-
fication of the relative levels of phospho-Akt, Erk, and
RPS6 (corrected to the total levels of respective proteins)
are shown in panels on the right.
See also Figure S3.

higher levels of phospho-RPS6 (Figure 3B). The


relative abilities of GDNF and bFGF to activate
mTORC1 correlated to their efficiency of Akt
and Erk activation (Figure 3B). However, this
correlation did not hold true for epidermal
growth factor (EGF) stimulation, possibly re-
flecting additional pathways by which EGF acti-
vates mTORC1 (Fan et al., 2009). In summary,
the self-renewal signal GDNF poorly activates
mTORC1 when compared to more general
mitogens for SPCs (bFGF, EGF), in agreement
with the Src kinase inhibition data (see above).
However, we note that although GDNF is
considered the key SPC self-renewal signal,
suppressed SPC colony growth and caused an accumulation of SPC expansion and self-renewal in vitro requires a combination
cells in G1 phase of the cell cycle (Figures S3A and S3B). of GDNF plus other factors (e.g., bFGF) that stimulate mTORC1
Growth factor receptors activate mTORC1 by signaling more efficiently (Lee et al., 2007b).
through phosphoinositide 3-kinase (PI3K)/Akt and Ras/Erk
MAPK, which inhibit the TSC1/TSC2 complex (Huang and Plzf Uncouples mTORC1 Activity from Growth Factor
Manning, 2008); TSC1/TSC2 negatively regulates mTORC1 via Signaling in SPCs through Redd1 Modulation
its GTPase-activating protein activity toward the small G protein Given that mTORC1 activity in SPCs is regulated by growth
Rheb. Treatment of WT SPCs with PI3K/Akt or Erk MAPK factors present in the culture medium and resultant PI3K/Akt
pathway inhibitors significantly reduced levels of phosphory- and Erk-MAPK activation, we next assessed whether the
lated RPS6 (Figure 3A), indicating that efficient mTORC1 activa- elevated mTORC1 activity of Plzf / SPCs correlated with an
tion in SPCs requires both PI3K/Akt and Erk MAPK. Inhibition of enhanced growth factor response. However, in comparison to
Src family kinases, implicated in the GDNF response of SPCs WT cells, the Akt and Erk activities of steady-state Plzf /
(Oatley et al., 2007), had modest effects on phospho-RPS6 SPCs were decreased whereas phospho-RPS6 was increased
(Figure 3A). Therefore, despite the ability of Src family kinases (Figure 3C), indicating that the elevated mTORC1 activity is not
to activate PI3K/Akt downstream GDNF (Encinas et al., 2001; due to increased activity of growth factor pathways.
Oatley et al., 2007), this SPC self-renewal pathway couples inef- We next considered alternative mechanisms by which
ficiently to mTORC1. mTORC1 activity would be increased in Plzf / SPCs. Cellular
SPCs are maintained with a cocktail of growth factors stress-response pathways provide additional regulatory inputs
(Kanatsu-Shinohara et al., 2003; Seandel et al., 2007) and are resulting in mTORC1 inhibition when conditions such as low
responsive to others (Hamra et al., 2007); thus we next assessed energy availability or hypoxia require temporary arrest of cell
contributions of these factors to mTORC1 activation in WT growth (Huang and Manning, 2008). We hypothesized that
SPCs. We found that GDNF triggered a modest phosphorylation mTORC1 hyperactivity in Plzf / SPCs could be due to reduced
of RPS6 whereas other mitogens (including bFGF) induced much activity of stress response pathways. Accordingly, we first

472 Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc.


Figure 4. Plzf Regulates Expression of the
mTORC1 Pathway Inhibitor Redd1
(A) Western blot analysis for components of the
mTORC1 pathway in two independently derived
sets of littermate Plzf +/+ and Plzf / SPC lines
(1 and 2) under steady-state conditions.
(B) Quantitative RT-PCR analysis of Redd1 mRNA
expression in independently derived Plzf +/+ and
Plzf / SPC lines. mRNA levels are normalized
to those of b-actin and standard deviations from
duplicate reactions are shown.
(C) Quantitative RT-PCR analysis of Redd1 mRNA
expression in spermatogonial fractions from
juvenile Plzf +/+ and Plzf / littermate mice:
SPCs (av-integrin negative, Thy-1 low, c-Kit nega-
tive), early differentiating spermatogonia (av-
integrin negative, Thy-1 low, c-Kit positive), late
differentiating spermatogonia (av-integrin nega-
tive, Thy-1 negative, c-Kit positive). mRNA levels
are normalized to those of b-actin and standard
deviations from duplicate reactions are shown.
Dashed line indicates Redd1 expression levels in
unfractionated WT testis cells.
(D) WT SPCs infected with control shRNA (against
GFP) or two independent shRNA constructs
against Redd1 were analyzed by western blot for
the indicated proteins. Quantification of relative
levels of phospho-RPS6 (corrected to total RPS6
levels) is shown under respective lanes. Cells
were under steady-state culturing conditions
(see also Figure S4).
(E) Top panel: Redd1 promoter (from translation
start site at +1 to 2 kb upstream) with location of
previously described transcription factor binding
sites. The promoter is divided into proximal and
distal regions (PP and DP, respectively). Positions
of ChIP amplicons are indicated. Bottom panel:
ChIP assay. Quantitative PCR for Redd1 promoter
regions from WT SPC chromatin pulled down
with Plzf antibody. Fold enrichment is shown rela-
tive to background of chromatin pulled down from
Plzf / SPCs with the same antibody. Standard
deviations from duplicate PCR reactions are
indicated.
(F) Luciferase reporter assay with constructs con-
taining the PP+DP Redd1 promoter regions or PP
alone. 293HEK cells were transfected with the
appropriate luciferase reporter together with PLZF constructs or vector control as indicated. Reporter activities were normalized to that of TK-Renilla and empty
vector luciferase reporter controls (*p < 0.001). Mean values from triplicate wells and associated standard deviations are indicated.

assessed levels of upstream regulatory proteins as stress fractions and found a relative enrichment of Redd1 mRNA
pathways converge on mTORC1 at the level of TSC1/TSC2. in SPCs (Figure 4C). We also confirmed that freshly isolated
However, we did not find significant differences in levels of Plzf / SPCs had lower Redd1 expression compared to controls
Tsc1, Tsc2, Rheb, or the mTOR kinase itself in Plzf / SPCs (Figure 4C). Furthermore, shRNA knockdown of Redd1 in WT
compared to WT cells (Figure 4A). SPCs increased mTORC1 activity (Figure 4D and Figure S4).
By contrast, we noticed that levels of Redd1 (also known as Together, our data suggest that Plzf opposes mTORC1 by main-
Ddit4, Rtp801, and Dig2) were substantially lower in Plzf / taining Redd1 expression and that Redd1 is a key mTORC1
SPCs compared to controls, at both protein and mRNA levels regulator in SPCs.
(Figures 4A and 4B). Redd1 is induced by multiple types of cell
stress (e.g., hypoxia, DNA damage) and by developmental Plzf Is a Transcriptional Activator of the mTORC1
signals and inhibits mTORC1 through regulation of TSC1/TSC2 Inhibitor Redd1 in SPCs
(Brugarolas et al., 2004; Corradetti et al., 2005; Ellisen et al., Redd1 expression was decreased at the mRNA level in
2002). As a role for Redd1 in SPCs is not described, we analyzed Plzf / SPCs. We therefore tested whether Plzf could directly
the distribution of Redd1 expression in isolated spermatogonial induce Redd1 expression. We first performed a chromatin

Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc. 473


Figure 5. A Negative Feedback Loop from
mTORC1 to the GDNF Receptor Is Activated
in Plzf / SPCs
(A) Plzf +/+ and Plzf / SPCs starved overnight in
basal SPC medium supplemented with DMSO
(vehicle control) or rapamycin were stimulated
with 10ng/ml GDNF for 20 min before harvesting
for western blot analysis.
(B) Quantification of relative levels of phospho-Akt
and phospho-RPS6 (corrected to total levels of
respective proteins) from (A).
(C) Plzf +/+ and Plzf / SPCs were plated in
medium containing decreasing concentrations of
GDNF, harvested one week later and counted to
determine fold cell recovery as an indicator of
growth. The concentration of GDNF was varied
from 4 ng/ml (left bars) to 1 ng/ml (right bars). Stan-
dard deviations from duplicate wells are indicated
(**p < 0.01, *p < 0.02).
(D) Quantitative RT-PCR analysis of GDNF
receptor components in Plzf +/+ and Plzf /
SPCs treated with DMSO (vehicle control) or rapa-
mycin for 48 hr. mRNA levels are normalized to
those of b-actin and standard deviations from
duplicate reactions are shown.
(E) Flow cytometry analysis of Gfra1 levels from
SPC lines treated as in (D).
(F) Quantitative RT-PCR analysis of GDNF
receptor components in av-integrin negative,
Thy-1 low, c-Kit negative testis cell fractions
pooled from 2-week-old Plzf +/+ and Plzf /
mice. mRNA levels are normalized to those of
b-actin and standard deviations from duplicate
reactions are shown.
See also Figure S5.

immunoprecipitation (ChIP) assay to assess whether Plzf was from mTORC1 could prevent Plzf / cells from activating Akt
bound to the Redd1 promoter in SPCs. We scanned a 2-kb in response to growth factors. In cells lacking TSC1/TSC2,
region upstream Redd1 that contains binding sites for multiple increased mTORC1-signaling induces a negative feedback
transcription factors known to regulate Redd1 expression loop from the mTORC1 downstream target S6K to upstream
(Figure 4E) (Ellisen et al., 2002; Lin et al., 2005; Shoshani et al., signaling components causing inhibition of PI3K/Akt (Harrington
2002). Importantly, we could detect Plzf association with distal et al., 2004; Shah et al., 2004). Therefore, we tested whether
promoter (DP) regions but not proximal promoter (PP) regions aberrant mTORC1 activity inhibited the response of Plzf /
(Figure 4E), indicating that Plzf regulates Redd1 expression SPCs to GDNF, a growth factor required for SPC self-renewal,
through recruitment to the DP. To confirm this we performed which could explain the maintenance defect of Plzf / SPCs.
luciferase assays with REDD1 promoter constructs containing We compared the response of starved Plzf +/+ and Plzf /
both DP+PP elements and the PP element alone (Figure 4F). SPCs to GDNF in the absence or presence of rapamycin to
In agreement with our ChIP results, PLZF activated the DP+PP vary mTORC1 activity (Figures 5A and 5B). Consistent with
construct but not the PP construct. Further, deletion of the previous data, Plzf / SPCs showed lower levels of basal
PLZF POZ domain prevented PP+DP reporter activation, indi- and GDNF-stimulated Akt activity when compared to WT
cating a requirement for this protein-protein interaction domain. SPCs, indicating a substantially reduced ability of Plzf / cells
We conclude that Plzf directly activates Redd1 through the DP to to activate PI3K/Akt. However, on rapamycin treatment, the
inhibit mTORC1 in SPCs. ability of Plzf / SPCs to activate Akt in response to GDNF
was significantly increased and became comparable to that of
Active mTORC1 Inhibits SPC Self-Renewal Pathways WT cells. This rescue of GDNF responsiveness by rapamycin
via a Negative Feedback Loop indicates that a negative feedback response from aberrantly
As Plzf / SPCs show increased mTORC1 but decreased Akt activated mTORC1 suppresses the response of Plzf / SPCs
activities (Figure 3C), we considered that negative feedback to GDNF. Indeed, if GDNF levels in the media were reduced,

474 Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc.


Plzf / SPC growth was inhibited more substantially than that of and numbers of avneg Thy-1low c-Kitneg cells in Plzf / testis,
the WT cells (Figure 5C). A decreased sensitivity to GDNF due approaching those values found in vehicle-treated WT controls
to negative feedback from mTORC1 can explain the defect in (Figures 6A and 6B). Rapamycin treatment of Plzf / mice
Plzf / SPC maintenance in vivo when levels of GDNF are increased both the frequency of avneg Thy-1low cells and normal-
limiting (Meng et al., 2000) and how Plzf / SPCs can be ized the increased percentage of c-Kitpos cells within this fraction
cultured in vitro when GDNF is supplied to excess. Importantly, compared to controls (Figure 6A). We conclude that aberrant
GDNF expression in Plzf / testis was equivalent to that of mTORC1 activation can explain, at least in part, the SPC mainte-
the WT (Figure S5), ruling out noncell autonomous defects in nance defect of Plzf / mice.
production of niche factors. Intriguingly, rapamycin also increased the frequency of SPCs
in WT mice (Figure 6B), suggesting that mTORC1 inhibition
Activated mTORC1 Suppresses Expression of GDNF stimulates SPC function in a WT setting. Indeed, in WT SPCs,
Receptor Components rapamycin enhanced Akt activation in response to GDNF
We next sought to determine the mechanism by which activated (Figures 5A and 5B) and increased GDNF receptor expression
mTORC1 inhibits response of Plzf / SPCs to GDNF. Negative (Figures 5D and 5E). Importantly, prolonged rapamycin treat-
feedback from mTORC1 to PI3K/Akt was originally character- ment of juvenile WT mice increased the number of cells with
ized in the context of insulin responsiveness and signaling high levels of Plzf expression in the testis (Figures 6C and 6D
through IRS1/2 proteins (Harrington et al., 2005). Although IRS and Figure S6B) and enhanced GDNF receptor expression
proteins have been implicated in activation of PI3K by the (Figure S6C), suggesting that physiological mTORC1 activity
GDNF receptor component c-Ret (Hennige et al., 2000), we controls size and function of the SPC pool in vivo.
noticed that expression of the GDNF receptor components
GFRa1 and c-Ret in SPCs were responsive to rapamycin DISCUSSION
(Figures 5D and 5E). Cultured Plzf / SPCs expressed lower
levels of GFRa1 and c-Ret compared to WT cells (correlating Disruption of genes encoding for negative regulators of the PI3K/
with reduced GDNF-responsiveness) but on mTORC1 inhibition, Akt/mTORC1 pathway (e.g., Pten, Tsc1, and Pml) leads to loss of
expression of these GDNF receptor components was increased hematopoietic stem cell (HSC) quiescence and subsequent
back to WT levels. Expression of GFRa1 and c-Ret were also exhaustion. Rapamycin prevents HSC depletion, demonstrating
lower in freshly isolated Plzf / SPCs compared to WT SPCs that aberrant mTORC1 activation is responsible for stem cell loss
(Figure 5F), confirming that Plzf loss disrupts GDNF receptor (Chen et al., 2008; Gan et al., 2008; Ito et al., 2008; Yilmaz et al.,
expression in vivo. Our data suggest that aberrantly activated 2006). The negative effects of mTORC1 on HSC maintenance
mTORC1 inhibits the response of Plzf / SPCs to GDNF by have been attributed to its downstream targets in cell growth
opposing expression of the receptor. Interestingly, mTORC1 pathways (Chen et al., 2008) and aberrant mTORC1 activity
inhibits upstream signaling events in MEFs through transcrip- elicits activation of tumor suppressive mechanisms that can
tional inhibition of the PDGF receptor (Zhang et al., 2007). In contribute to stem cell failure (Figure S6D) (Alimonti et al.,
SPCs, negative feedback between mTORC1 and the GDNF 2010; Lee et al., 2007a). By studying Plzf function in SPCs, we
receptor will lead to loss of self-renewal when mTORC1 is hyper- now implicate negative feedback responses from the mTORC1
activated, such as occurs on loss of Plzf expression (Figure S6A). pathway to receptors required to transduce niche-derived self-
This feedback loop would inversely couple cell growth to self- renewal signals in the loss of stem cell potential (Figure S6E).
renewal, so that on expansion of the stem cell pool by mitogenic HSC maintenance is also linked to niche signals acting
stimulation and resultant mTORC1 activation, response of the through growth factor receptors (Arai et al., 2004; Yoshihara
cells to self-renewal signals is inhibited, restricting stem cell et al., 2007) thus it will be of interest to translate our findings
numbers. Interestingly, GDNF activates mTORC1 weakly in into the hematopoietic system. In addition, the decreased sensi-
SPCs when compared to other mitogens (e.g., bFGF) (Figure 3B), tivity of Plzf / SPCs to GDNF mimics a reduced production of
suggesting that GDNF would trigger a limited activation of the niche-signals; the decline in stem cell numbers in aging mouse
negative feedback response whereas bFGF could cause more testis is due partly to declining niche function and GDNF produc-
extensive inhibition of GDNF signaling via mTORC1. tion (Ryu et al., 2006; Zhang et al., 2006), thus loss of Plzf expres-
sion can represent a premature aging phenotype. Our studies
Rapamycin Treatment Attenuates the SPC Maintenance therefore provide further insight into the association between
Defect of Plzf / Mice and Increases SPCs in WT stem cell maintenance, mTORC1, and aging (Harrison et al.,
Control Mice 2009; Rossi et al., 2008).
As inhibition of aberrant mTORC1 activity in Plzf / SPCs in vitro A correlation between aging and cancer is also apparent, likely
rescues their response to GDNF, we next asked whether due to the accumulation of multiple genetic hits over time for
mTORC1 inhibition in vivo could rescue the Plzf / SPC mainte- tumor development (Rossi et al., 2008). As stem cells are resi-
nance defect. We initiated rapamycin treatment of Plzf / and dent long-term in tissues and possess self-renewal capability
WT control mice during early stages of postnatal development (a feature of cancer stem cells), they are suggested to be the
(10 days postnatal) and continued daily treatment for 1 week, source of many cancers. However, loss of tumor suppressors
a regimen able to normalize the increased size of Plzf / SPCs (e.g., Pten, Pml) trigger stem cell depletion (Ito et al., 2009) and
(Figures 2G and 2H). Subsequent analysis of SPC compartment subsequent cancer development is associated with additional
status demonstrated that rapamycin increased both frequency genetic hits (Guo et al., 2008). It is proposed that oncogenic

Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc. 475


Figure 6. mTORC1 Inhibition Attenuates
SPC Maintenance Defect of Plzf / Mice
(A) Top panels: representative av-integrin/Thy-1
flow profiles of testis cells from Plzf +/+ and
Plzf / mice treated with vehicle or rapamycin
from 10 days postnatal age for a period of
1 week and analyzed the day after completing
treatment. Bottom panels: Flow cytometry anal-
ysis of c-Kit expression within the corresponding
av-integrin negative, Thy-1 low fractions.
(B) Quantification of frequency and absolute
numbers of av integrin negative, Thy-1 low, c-Kit
negative testis cells from mice of the indicated
genotypes treated with Vehicle or rapamycin as
in (A) (n R 5 per genotype and treatment group,
**p < 0.02, *p < 0.05). Graphs indicate mean
values ± SEM.
(C) WT juvenile mice (2–3 weeks postnatal) were
treated daily for 2 weeks with rapamycin or vehicle.
Testes were harvested and subject to immunohis-
tochemistry for Plzf. Representative images are
203.
(D) Quantification of cells showing strong positivity
for Plzf in seminiferous tubules of testis from (C),
representing SPCs. Results from duplicate exper-
iments (1 and 2) are shown together with SEM
(**p < 0.01).
(E) Illustration of the effects of aberrant mTORC1
activity or inhibition of physiological mTORC1
activation with rapamycin on SPC numbers and
function.
See also Figure S6.

PLZF was first identified from involve-


ment in acute promyelocytic leukemia
(APL) (Chen et al., 1993). Thus, our model
of Plzf-dependent SPC maintenance
can have implications for leukemia patho-
genesis. The ability of Plzf to inhibit
mTORC1 through Redd1 induction may
be relevant in this context given that
perturbed PLZF function and elevated
mTORC1 activity are associated with
leukemia development (He et al., 2000;
Martelli et al., 2007; McConnell and Licht,
2007). In addition, REDD1 is involved in
hits in stem cells elicit activation of fail-safe mechanisms that leukemic and myeloid cell differentiation (Gery et al., 2007;
have to be evaded to allow cancer development (Figure S6D). Nishioka et al., 2009), suggesting that this gene can be a
These fail-safe mechanisms prevent propagation of clones of relevant PLZF target in hematopoietic cells. PML, also identified
cells derived from stem cells with oncogenic mutations and the from its involvement in chromosomal translocations with the
consequent risk of acquiring additional mutations that lead to RARA gene in APL, is required for HSC maintenance and inhibits
cancer. The mTORC1 hyperactivation that occurs in response mTORC1 (Bernardi et al., 2006; Ito et al., 2008); thus illustrating
to loss of tumor suppressors such as Pten and Pml is likely a striking commonality of function between the RARA partners
a trigger of this fail-safe mechanism as rapamycin prevents of APL.
stem cell exhaustion. Based on our new model, mTORC1 We have identified a role for Plzf-mediated Redd1 expression
hyperactivation in response to oncogenic stimulation may also in inhibiting mTORC1 activation in SPCs. As mTORC1 activity
desensitize the cancer stem cell to niche-signals that are has a significant impact on the response of SPCs to GDNF, the
required for self-renewal; thus additional mutations supporting tight regulation of this pathway is critical. Redd1 is also induced
niche-independent self-renewal could allow cancer to develop in response to cellular stress and in particular, by hypoxia
(Figure S6E). (Brugarolas et al., 2004; Shoshani et al., 2002). Interestingly,

476 Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc.


the HSC niche has been suggested to exist in a hypoxic state Chromatin Immunoprecipitation (ChIP)
(Kubota et al., 2008; Parmar et al., 2007) that, if the SPC niche SPCs were trypsinized and resuspended in SPC medium then incubated in
tissue culture dishes for 30 min to remove contaminating (adherent) MEFs.
had similar characteristics, could be responsible for Redd1
Preparation and immunoprecipitation of chromatin was performed using
induction. However, SPCs reside close to the vasculature a SimpleChIP Enzymatic Chromatin IP kit (Cell Signaling Technology)
in vivo (Yoshida et al., 2007) and Redd1 expression is maintained according to manufacturers instructions and monoclonal anti-PLZF antibody
when SPCs are cultured at atmospheric oxygen levels. Thus (Calbiochem). DNA samples were analyzed by quantitative PCR using a
Redd1 expression in this case depends on a Plzf tonic transcrip- QuantiTect SYBR Green PCR kit (QIAGEN) and Light Cycler (Roche). Primer
tional activity and its induction is seemingly independent of sequences for the Redd1 promoter are included in the Extended Experimental
hypoxia. Procedures.

As mTORC1 inhibition enhances SPC function even in a WT


Luciferase Assay
setting, our data also have important therapeutic implications. REDD1 promoter constructs are described (Lin et al., 2005) and were cloned
It is tempting to speculate that rapamycin could be used to from U2OS genomic DNA into the pGL3-enhancer vector (Promega). Lucif-
enhance SPC activity in humans; potentially allowing improved erase assays were performed with 293HEK cells as described (Barna et al.,
SPC culture from testicular biopsies and generation of therapeu- 2002). Please refer to Extended Experimental Procedures for details.
tically relevant MASCs (Figure 6E). Furthermore, our findings
provide a novel and straightforward explanation for the require- Quantitative Reverse Transcription Polymerase Chain Reaction
(qRT-PCR)
ment of Plzf in germline maintenance.
RNA was harvested using Trizol reagent (Invitrogen) then used for first strand
cDNA synthesis and subsequent quantitative PCR analysis as described
EXPERIMENTAL PROCEDURES
(Filipponi et al., 2007). For primer sequences and detailed methodology please
see Extended Experimental Procedures.
Mouse Maintenance and Manipulation
Plzf / mice are previously described (Costoya et al., 2004). Mice were treated
Statistical Analysis
with rapamycin (LC Laboratories) at a daily dose of 4 mg/kg as described
Results from cell growth assays, testis transplants, and flow cytometry anal-
(Yilmaz et al., 2006). Transgenic mice expressing EGFP from the b-actin
ysis were assessed for statistical significance by a standard two-tailed t test.
promoter were from Jackson Laboratories. Testis transplant assays were
performed as described (Seandel et al., 2007) with 50 3 103 unfractionated
or 5 3 103 avneg Thy-1low c-Kitneg testis cells injected per recipient testis. SUPPLEMENTAL INFORMATION
Teratoma formation assays in NOD/SCID mice (Jackson Laboratories) were
performed as described (Seandel et al., 2007). Supplemental Information includes Extended Experimental Procedures and
six figures and can be found with this article online at doi:10.1016/j.cell.
2010.06.041.
Antibody Generation
Monoclonal anti-PLZF antibody (clone 9E12) was raised in Armenian hamster ACKNOWLEDGMENTS
against a pool of keyhole limpet hemocyanin (KLH)-conjugated peptides at the
Memorial Sloan-Kettering Cancer Center (MSKCC) antibody facility and reacts We thank current and past members of the Pandolfi lab and in particular Taka-
to a peptide within the hinge domain of mouse Plzf (RSKEGPGTPTRRSVIT hiro Maeda and Rosa Bernardi for helpful discussion and advice. We would
SARE). Antibody was directly conjugated to Alexa 488 or Alexa 647 for use also like to thank Antonella Papa for generating luciferase reporters, Sharmila
in flow cytometry (MSKCC antibody facility). Fagoonee for experimental help, Leif Ellisen for Redd1 promoter constructs,
and the flow cytometry facilities of BIDMC and MSKCC for expert support.
M.S. is a Stanley and Fiona Druckenmiller Fellow of the New York Stem Cell
Flow Cytometry
Foundation. This work was supported by NIH grants to P.P.P.
Single cell suspensions were prepared from testis as described (Ogawa et al.,
1997) and resuspended in phosphate-buffered saline (PBS) containing 2%
Received: October 5, 2009
fetal bovine serum (FBS) for subsequent staining and analysis (Filipponi
Revised: April 1, 2010
et al., 2007). For a detailed description of flow cytometry methods and anti-
Accepted: May 28, 2010
bodies used, refer to the Extended Experimental Procedures.
Published: August 5, 2010

Cell Culture and Treatment REFERENCES


SPC fractions from testis were plated directly onto mitotically inactivated MEF
feeder cells. Medium for SPC culture was as previously described (Seandel Alimonti, A., Nardella, C., Chen, Z., Clohessy, J.G., Carracedo, A., Trotman,
et al., 2007) supplemented with 10 ng/ml IGF-I (Peprotech). MASCs and L.C., Cheng, K., Varmeh, S., Kozma, S.C., Thomas, G., et al. (2010). A novel
mouse ESCs (v6.5) were cultured in embryonic stem cell medium on MEF type of cellular senescence that can be enhanced in mouse models and human
feeders (Seandel et al., 2007). Knockdown of Redd1 expression in SPCs by tumor xenografts to suppress prostate tumorigenesis. J. Clin. Invest. 120,
shRNA was performed using pLKO.1 lentiviral vectors (Open Biosystems). 681–693.
A detailed description of methods is included in the Extended Experimental Arai, F., Hirao, A., Ohmura, M., Sato, H., Matsuoka, S., Takubo, K., Ito, K., Koh,
Procedures. G.Y., and Suda, T. (2004). Tie2/angiopoietin-1 signaling regulates hematopoi-
etic stem cell quiescence in the bone marrow niche. Cell 118, 149–161.
Immunofluorescence, Immunohistochemistry, and Western Blot Barna, M., Merghoub, T., Costoya, J.A., Ruggero, D., Branford, M., Bergia, A.,
Tissue and cultured cells were fixed in 4% paraformaldehyde before Samori, B., and Pandolfi, P.P. (2002). Plzf mediates transcriptional repression
processing for immunofluorescence (IF) and immunohistochemistry (IHC) as of HoxD gene expression through chromatin remodeling. Dev. Cell 3, 499–510.
described (Costoya et al., 2004; Filipponi et al., 2007). For western blot anal- Bernardi, R., Guernah, I., Jin, D., Grisendi, S., Alimonti, A., Teruya-Feldstein, J.,
ysis, cells were lysed in RIPA buffer and processed as described (Costoya Cordon-Cardo, C., Simon, M.C., Rafii, S., and Pandolfi, P.P. (2006). PML
et al., 2008). Detailed methods are included in the Extended Experimental inhibits HIF-1alpha translation and neoangiogenesis through repression of
Procedures. mTOR. Nature 442, 779–785.

Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc. 477


Brown, E.J., Albers, M.W., Shin, T.B., Ichikawa, K., Keith, C.T., Lane, W.S., and Gan, B., Sahin, E., Jiang, S., Sanchez-Aguilera, A., Scott, K.L., Chin, L.,
Schreiber, S.L. (1994). A mammalian protein targeted by G1-arresting rapamy- Williams, D.A., Kwiatkowski, D.J., and DePinho, R.A. (2008). mTORC1-depen-
cin-receptor complex. Nature 369, 756–758. dent and -independent regulation of stem cell renewal, differentiation, and
Brugarolas, J., Lei, K., Hurley, R.L., Manning, B.D., Reiling, J.H., Hafen, E., mobilization. Proc. Natl. Acad. Sci. USA 105, 19384–19389.
Witters, L.A., Ellisen, L.W., and Kaelin, W.G., Jr. (2004). Regulation of mTOR Gery, S., Park, D.J., Vuong, P.T., Virk, R.K., Muller, C.I., Hofmann, W.K., and
function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor Koeffler, H.P. (2007). RTP801 is a novel retinoic acid-responsive gene associ-
suppressor complex. Genes Dev. 18, 2893–2904. ated with myeloid differentiation. Exp. Hematol. 35, 572–578.
Buaas, F.W., Kirsh, A.L., Sharma, M., McLean, D.J., Morris, J.L., Griswold, Guo, W., Lasky, J.L., Chang, C.J., Mosessian, S., Lewis, X., Xiao, Y., Yeh, J.E.,
M.D., de Rooij, D.G., and Braun, R.E. (2004). Plzf is required in adult male Chen, J.Y., Iruela-Arispe, M.L., Varella-Garcia, M., et al. (2008). Multi-genetic
germ cells for stem cell self-renewal. Nat. Genet. 36, 647–652. events collaboratively contribute to Pten-null leukemia stem-cell formation.
Castilho, R.M., Squarize, C.H., Chodosh, L.A., Williams, B.O., and Gutkind, Nature 453, 529–533.
J.S. (2009). mTOR mediates Wnt-induced epidermal stem cell exhaustion Hamra, F.K., Chapman, K.M., Nguyen, D., and Garbers, D.L. (2007). Identifica-
and aging. Cell Stem Cell 5, 279–289. tion of neuregulin as a factor required for formation of aligned spermatogonia.
Chen, C., Liu, Y., Liu, R., Ikenoue, T., Guan, K.L., and Zheng, P. (2008). TSC- J. Biol. Chem. 282, 721–730.
mTOR maintains quiescence and function of hematopoietic stem cells by Harrington, L.S., Findlay, G.M., Gray, A., Tolkacheva, T., Wigfield, S., Rebholz,
repressing mitochondrial biogenesis and reactive oxygen species. J. Exp. H., Barnett, J., Leslie, N.R., Cheng, S., Shepherd, P.R., et al. (2004). The
Med. 205, 2397–2408. TSC1-2 tumor suppressor controls insulin-PI3K signaling via regulation of
Chen, Z., Brand, N.J., Chen, A., Chen, S.J., Tong, J.H., Wang, Z.Y., Waxman, IRS proteins. J. Cell Biol. 166, 213–223.
S., and Zelent, A. (1993). Fusion between a novel Kruppel-like zinc finger gene Harrington, L.S., Findlay, G.M., and Lamb, R.F. (2005). Restraining PI3K:
and the retinoic acid receptor-alpha locus due to a variant t(11;17) transloca- mTOR signalling goes back to the membrane. Trends Biochem. Sci. 30, 35–42.
tion associated with acute promyelocytic leukaemia. EMBO J. 12, 1161–1167. Harrison, D.E., Strong, R., Sharp, Z.D., Nelson, J.F., Astle, C.M., Flurkey, K.,
Corradetti, M.N., Inoki, K., and Guan, K.L. (2005). The stress-inducted proteins Nadon, N.L., Wilkinson, J.E., Frenkel, K., Carter, C.S., et al. (2009). Rapamycin
RTP801 and RTP801L are negative regulators of the mammalian target of fed late in life extends lifespan in genetically heterogeneous mice. Nature 460,
rapamycin pathway. J. Biol. Chem. 280, 9769–9772. 392–395.
Costoya, J.A., Hobbs, R.M., Barna, M., Cattoretti, G., Manova, K., Sukhwani, He, L.Z., Bhaumik, M., Tribioli, C., Rego, E.M., Ivins, S., Zelent, A., and
M., Orwig, K.E., Wolgemuth, D.J., and Pandolfi, P.P. (2004). Essential role of Pandolfi, P.P. (2000). Two critical hits for promyelocytic leukemia. Mol. Cell
Plzf in maintenance of spermatogonial stem cells. Nat. Genet. 36, 653–659. 6, 1131–1141.
Costoya, J.A., Hobbs, R.M., and Pandolfi, P.P. (2008). Cyclin-dependent Hennige, A.M., Lammers, R., Arlt, D., Hoppner, W., Strack, V., Niederfellner,
kinase antagonizes promyelocytic leukemia zinc-finger through phosphoryla- G., Seif, F.J., Haring, H.U., and Kellerer, M. (2000). Ret oncogene signal trans-
tion. Oncogene 27, 3789–3796. duction via a IRS-2/PI 3-kinase/PKB and a SHC/Grb-2 dependent pathway:
David, G., Alland, L., Hong, S.H., Wong, C.W., DePinho, R.A., and Dejean, A. possible implication for transforming activity in NIH3T3 cells. Mol. Cell. Endo-
(1998). Histone deacetylase associated with mSin3A mediates repression by crinol. 167, 69–76.
the acute promyelocytic leukemia-associated PLZF protein. Oncogene 16, Huang, J., and Manning, B.D. (2008). The TSC1-TSC2 complex: a molecular
2549–2556. switchboard controlling cell growth. Biochem. J. 412, 179–190.
de Rooij, D.G., and Russell, L.D. (2000). All you wanted to know about Ito, K., Bernardi, R., Morotti, A., Matsuoka, S., Saglio, G., Ikeda, Y., Rosenblatt,
spermatogonia but were afraid to ask. J. Androl. 21, 776–798. J., Avigan, D.E., Teruya-Feldstein, J., and Pandolfi, P.P. (2008). PML targeting
Doulatov, S., Notta, F., Rice, K.L., Howell, L., Zelent, A., Licht, J.D., and Dick, eradicates quiescent leukaemia-initiating cells. Nature 453, 1072–1078.
J.E. (2009). PLZF is a regulator of homeostatic and cytokine-induced myeloid Ito, K., Bernardi, R., and Pandolfi, P.P. (2009). A novel signaling network as
development. Genes Dev. 23, 2076–2087. a critical rheostat for the biology and maintenance of the normal stem cell
Ellisen, L.W., Ramsayer, K.D., Johannessen, C.M., Yang, A., Beppu, H., and the cancer-initiating cell. Curr. Opin. Genet. Dev. 19, 51–59.
Minda, K., Oliner, J.D., McKeon, F., and Haber, D.A. (2002). REDD1, a develop- Kanatsu-Shinohara, M., Inoue, K., Lee, J., Yoshimoto, M., Ogonuki, N., Miki,
mentally regulated transcriptional target of p63 and p53, links p63 to regulation H., Baba, S., Kato, T., Kazuki, Y., Toyokuni, S., et al. (2004a). Generation of
of reactive oxygen species. Mol. Cell 10, 995–1005. pluripotent stem cells from neonatal mouse testis. Cell 119, 1001–1012.
Encinas, M., Tansey, M.G., Tsui-Pierchala, B.A., Comella, J.X., Milbrandt, J., Kanatsu-Shinohara, M., Ogonuki, N., Inoue, K., Miki, H., Ogura, A., Toyokuni,
and Johnson, E.M., Jr. (2001). c-Src is required for glial cell line-derived neuro- S., and Shinohara, T. (2003). Long-term proliferation in culture and germline
trophic factor (GDNF) family ligand-mediated neuronal survival via a phospha- transmission of mouse male germline stem cells. Biol. Reprod. 69, 612–616.
tidylinositol-3 kinase (PI-3K)-dependent pathway. J. Neurosci. 21, 1464–1472. Kanatsu-Shinohara, M., Toyokuni, S., and Shinohara, T. (2004b). CD9 is
Fan, Q.W., Cheng, C., Knight, Z.A., Haas-Kogan, D., Stokoe, D., James, C.D., a surface marker on mouse and rat male germline stem cells. Biol. Reprod.
McCormick, F., Shokat, K.M., and Weiss, W.A. (2009). EGFR signals to mTOR 70, 70–75.
through PKC and independently of Akt in glioma. Sci. Signal. 2, ra4. Kubota, H., Avarbock, M.R., and Brinster, R.L. (2003). Spermatogonial stem
Filipponi, D., Hobbs, R.M., Ottolenghi, S., Rossi, P., Jannini, E.A., Pandolfi, cells share some, but not all, phenotypic and functional characteristics with
P.P., and Dolci, S. (2007). Repression of kit expression by Plzf in germ cells. other stem cells. Proc. Natl. Acad. Sci. USA 100, 6487–6492.
Mol. Cell. Biol. 27, 6770–6781. Kubota, H., Avarbock, M.R., and Brinster, R.L. (2004). Growth factors essential
Fingar, D.C., Salama, S., Tsou, C., Harlow, E., and Blenis, J. (2002). Mamma- for self-renewal and expansion of mouse spermatogonial stem cells. Proc.
lian cell size is controlled by mTOR and its downstream targets S6K1 and Natl. Acad. Sci. USA 101, 16489–16494.
4EBP1/eIF4E. Genes Dev. 16, 1472–1487. Kubota, Y., Takubo, K., and Suda, T. (2008). Bone marrow long label-retaining
Fischer, S., Kohlhase, J., Bohm, D., Schweiger, B., Hoffmann, D., Heitmann, cells reside in the sinusoidal hypoxic niche. Biochem. Biophys. Res. Commun.
M., Horsthemke, B., and Wieczorek, D. (2008). Biallelic loss of function of 366, 335–339.
the promyelocytic leukemia zinc finger (PLZF) gene causes severe skeletal Labbaye, C., Quaranta, M.T., Pagliuca, A., Militi, S., Licht, J.D., Testa, U., and
defects and genital hypoplasia. J. Med. Genet. 45, 731–737. Peschle, C. (2002). PLZF induces megakaryocytic development, activates
Gan, B., and DePinho, R.A. (2009). mTORC1 signaling governs hematopoietic Tpo receptor expression and interacts with GATA1 protein. Oncogene 21,
stem cell quiescence. Cell Cycle 8, 1003–1006. 6669–6679.

478 Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc.


Lee, C.H., Inoki, K., Karbowniczek, M., Petroulakis, E., Sonenberg, N., Henske, Schieke, S.M., and Finkel, T. (2006). Mitochondrial signaling, TOR, and life
E.P., and Guan, K.L. (2007a). Constitutive mTOR activation in TSC mutants span. Biol. Chem. 387, 1357–1361.
sensitizes cells to energy starvation and genomic damage via p53. EMBO J.
Schrans-Stassen, B.H., van de Kant, H.J., de Rooij, D.G., and van Pelt, A.M.
26, 4812–4823.
(1999). Differential expression of c-kit in mouse undifferentiated and differen-
Lee, J., Kanatsu-Shinohara, M., Inoue, K., Ogonuki, N., Miki, H., Toyokuni, S., tiating type A spermatogonia. Endocrinology 140, 5894–5900.
Kimura, T., Nakano, T., Ogura, A., and Shinohara, T. (2007b). Akt mediates
Seandel, M., James, D., Shmelkov, S.V., Falciatori, I., Kim, J., Chavala, S.,
self-renewal division of mouse spermatogonial stem cells. Development
Scherr, D.S., Zhang, F., Torres, R., Gale, N.W., et al. (2007). Generation of
134, 1853–1859.
functional multipotent adult stem cells from GPR125+ germline progenitors.
Lin, L., Stringfield, T.M., Shi, X., and Chen, Y. (2005). Arsenite induces a cell
Nature 449, 346–350.
stress-response gene, RTP801, through reactive oxygen species and tran-
scription factors Elk-1 and CCAAT/enhancer-binding protein. Biochem. J. Shah, O.J., Wang, Z., and Hunter, T. (2004). Inappropriate activation of the
392, 93–102. TSC/Rheb/mTOR/S6K cassette induces IRS1/2 depletion, insulin resistance,
and cell survival deficiencies. Curr. Biol. 14, 1650–1656.
Ma, X.M., and Blenis, J. (2009). Molecular mechanisms of mTOR-mediated
translational control. Nat. Rev. Mol. Cell Biol. 10, 307–318. Shaw, R.J., and Cantley, L.C. (2006). Ras, PI(3)K and mTOR signalling controls
Martelli, A.M., Tazzari, P.L., Evangelisti, C., Chiarini, F., Blalock, W.L., Billi, tumour cell growth. Nature 441, 424–430.
A.M., Manzoli, L., McCubrey, J.A., and Cocco, L. (2007). Targeting the phos- Shinohara, T., Avarbock, M.R., and Brinster, R.L. (1999). beta1- and alpha6-
phatidylinositol 3-kinase/Akt/mammalian target of rapamycin module for integrin are surface markers on mouse spermatogonial stem cells. Proc.
acute myelogenous leukemia therapy: from bench to bedside. Curr. Med. Natl. Acad. Sci. USA 96, 5504–5509.
Chem. 14, 2009–2023.
Shinohara, T., Orwig, K.E., Avarbock, M.R., and Brinster, R.L. (2000).
McConnell, M.J., and Licht, J.D. (2007). The PLZF gene of t (11;17)-associated Spermatogonial stem cell enrichment by multiparameter selection of mouse
APL. Curr. Top. Microbiol. Immunol. 313, 31–48. testis cells. Proc. Natl. Acad. Sci. USA 97, 8346–8351.
Meng, X., Lindahl, M., Hyvonen, M.E., Parvinen, M., de Rooij, D.G., Hess,
Shoshani, T., Faerman, A., Mett, I., Zelin, E., Tenne, T., Gorodin, S., Moshel, Y.,
M.W., Raatikainen-Ahokas, A., Sainio, K., Rauvala, H., Lakso, M., et al.
Elbaz, S., Budanov, A., Chajut, A., et al. (2002). Identification of a novel
(2000). Regulation of cell fate decision of undifferentiated spermatogonia by
hypoxia-inducible factor 1-responsive gene, RTP801, involved in apoptosis.
GDNF. Science 287, 1489–1493.
Mol. Cell. Biol. 22, 2283–2293.
Nagano, M., Avarbock, M.R., and Brinster, R.L. (1999). Pattern and kinetics of
mouse donor spermatogonial stem cell colonization in recipient testes. Biol. Takubo, K., Ohmura, M., Azuma, M., Nagamatsu, G., Yamada, W., Arai, F.,
Reprod. 60, 1429–1436. Hirao, A., and Suda, T. (2008). Stem cell defects in ATM-deficient undifferen-
tiated spermatogonia through DNA damage-induced cell-cycle arrest. Cell
Nishioka, C., Ikezoe, T., Yang, J., Gery, S., Koeffler, H.P., and Yokoyama, A.
Stem Cell 2, 170–182.
(2009). Inhibition of mammalian target of rapamycin signaling potentiates the
effects of all-trans retinoic acid to induce growth arrest and differentiation of Virtanen, I., Kallajoki, M., Narvanen, O., Paranko, J., Thornell, L.E., Miettinen,
human acute myelogenous leukemia cells. Int. J. Cancer 125, 1710–1720. M., and Lehto, V.P. (1986). Peritubular myoid cells of human and rat testis
Oatley, J.M., Avarbock, M.R., and Brinster, R.L. (2007). Glial cell line-derived are smooth muscle cells that contain desmin-type intermediate filaments.
neurotrophic factor regulation of genes essential for self-renewal of mouse Anat. Rec. 215, 10–20.
spermatogonial stem cells is dependent on Src family kinase signaling. J. Wu, X., Oatley, J.M., Oatley, M.J., Kaucher, A.V., Avarbock, M.R., and Brinster,
Biol. Chem. 282, 25842–25851. R.L. (2010). The POU domain transcription factor POU3F1 is an important
Oatley, J.M., Avarbock, M.R., Telaranta, A.I., Fearon, D.T., and Brinster, R.L. intrinsic regulator of GDNF-induced survival and self-renewal of mouse
(2006). Identifying genes important for spermatogonial stem cell self-renewal spermatogonial stem cells. Biol. Reprod. 82, 1103–1111.
and survival. Proc. Natl. Acad. Sci. USA 103, 9524–9529. Yilmaz, O.H., Valdez, R., Theisen, B.K., Guo, W., Ferguson, D.O., Wu, H., and
Ogawa, T., Arechaga, J.M., Avarbock, M.R., and Brinster, R.L. (1997). Trans- Morrison, S.J. (2006). Pten dependence distinguishes hematopoietic stem
plantation of testis germinal cells into mouse seminiferous tubules. Int. J. Dev. cells from leukemia-initiating cells. Nature 441, 475–482.
Biol. 41, 111–122.
Yoshida, S., Sukeno, M., and Nabeshima, Y. (2007). A vasculature-associated
Ohbo, K., Yoshida, S., Ohmura, M., Ohneda, O., Ogawa, T., Tsuchiya, H., niche for undifferentiated spermatogonia in the mouse testis. Science 317,
Kuwana, T., Kehler, J., Abe, K., Scholer, H.R., et al. (2003). Identification and 1722–1726.
characterization of stem cells in prepubertal spermatogenesis in mice small
star, filled. Dev. Biol. 258, 209–225. Yoshida, S., Sukeno, M., Nakagawa, T., Ohbo, K., Nagamatsu, G., Suda, T.,
and Nabeshima, Y. (2006). The first round of mouse spermatogenesis is
Parmar, K., Mauch, P., Vergilio, J.A., Sackstein, R., and Down, J.D. (2007).
a distinctive program that lacks the self-renewing spermatogonia stage.
Distribution of hematopoietic stem cells in the bone marrow according to
Development 133, 1495–1505.
regional hypoxia. Proc. Natl. Acad. Sci. USA 104, 5431–5436.
Yoshihara, H., Arai, F., Hosokawa, K., Hagiwara, T., Takubo, K., Nakamura, Y.,
Rossi, D.J., Jamieson, C.H., and Weissman, I.L. (2008). Stems cells and the
Gomei, Y., Iwasaki, H., Matsuoka, S., Miyamoto, K., et al. (2007). Thrombo-
pathways to aging and cancer. Cell 132, 681–696.
poietin/MPL signaling regulates hematopoietic stem cell quiescence and
Ruvinsky, I., Sharon, N., Lerer, T., Cohen, H., Stolovich-Rain, M., Nir, T., Dor,
interaction with the osteoblastic niche. Cell Stem Cell 1, 685–697.
Y., Zisman, P., and Meyuhas, O. (2005). Ribosomal protein S6 phosphorylation
is a determinant of cell size and glucose homeostasis. Genes Dev. 19, Zhang, H., Bajraszewski, N., Wu, E., Wang, H., Moseman, A.P., Dabora, S.L.,
2199–2211. Griffin, J.D., and Kwiatkowski, D.J. (2007). PDGFRs are critical for PI3K/Akt
activation and negatively regulated by mTOR. J. Clin. Invest. 117, 730–738.
Ryu, B.Y., Orwig, K.E., Oatley, J.M., Avarbock, M.R., and Brinster, R.L. (2006).
Effects of aging and niche microenvironment on spermatogonial stem cell Zhang, X., Ebata, K.T., Robaire, B., and Nagano, M.C. (2006). Aging of male
self-renewal. Stem Cells 24, 1505–1511. germ line stem cells in mice. Biol. Reprod. 74, 119–124.

Cell 142, 468–479, August 6, 2010 ª2010 Elsevier Inc. 479


Myc-Nick: A Cytoplasmic Cleavage
Product of Myc that Promotes a-Tubulin
Acetylation and Cell Differentiation
Maralice Conacci-Sorrell,1 Celine Ngouenet,1 and Robert N. Eisenman1,*
1Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA

*Correspondence: eisenman@fhcrc.org
DOI 10.1016/j.cell.2010.06.037

SUMMARY Myc family proteins contain highly conserved regions termed


Myc boxes (MB) that are essential for Myc’s biological activities
The Myc oncoprotein family comprises transcription (see Figure 1E). A major determinant of Myc transcriptional
factors that control multiple cellular functions and function is MBII, which is the site of recruitment of coactivator
are widely involved in oncogenesis. Here we report complexes containing histone acetyltransferases (HATs) such
the identification of Myc-nick, a cytoplasmic form of as GCN5 (McMahon et al., 2000) and TIP60 (Frank et al.,
Myc generated by calpain-dependent proteolysis at 2003). MBI functions as a phosphorylation-dependent binding
site for the ubiquitin ligase Fbw7 (Welcker et al., 2004), whereas
lysine 298 of full-length Myc. Myc-nick retains
MBII is one of the binding sites for the ligase SKP2 (Kim et al.,
conserved Myc box regions but lacks nuclear locali- 2003; von der Lehr et al., 2003). Fbw7 and Skp2 both contribute
zation signals and the bHLHZ domain essential for to the rapid degradation of Myc protein (t1/2 20 min). The C
heterodimerization with Max and DNA binding. Myc- terminus of Myc harbors nuclear localization signals and the
nick induces a-tubulin acetylation and altered cell bHLHZ motif that mediates dimerization with Max and DNA
morphology by recruiting histone acetyltransferase binding.
GCN5 to microtubules. During muscle differentiation, Several variant forms of Myc protein have been previously
while the levels of full-length Myc diminish, Myc- identified. All of them are nuclear-localized, low-abundance
nick and acetylated a-tubulin levels are increased. proteins generated by alternative translation initiation. A weak
Ectopic expression of Myc-nick accelerates myo- CUG translational initiation site, upstream and in-frame of the
blast fusion, triggers the expression of myogenic predominant AUG codon, produces an N-terminally extended
form of c-Myc called c-Myc1 (Hann et al., 1988). Another Myc
markers, and permits Myc-deficient fibroblasts to
protein variant is MycS, generated by internal translational initia-
transdifferentiate in response to MyoD. We propose tions at two AUG codons located 100 amino acids from the
that the cleavage of Myc by calpain abrogates the normal N terminus (Spotts et al., 1997). MycS lacks MBI but
transcriptional inhibition of differentiation by full- contains MBII and retains much of full-length Myc’s biological
length Myc and generates Myc-nick, a driver of cyto- activity (Xiao et al., 1998).
plasmic reorganization and differentiation. As expected, given their broad role as transcriptional regula-
tors, Myc family proteins are predominantly localized to the
cell nucleus during proliferation. Surprisingly, however, there
INTRODUCTION have been multiple reports of cytoplasmically localized Myc,
mostly in differentiated cells. For example, N-Myc localiza-
The Myc family (c-Myc, N-Myc, and L-Myc) of basic-helix-loop- tion was shown to change from nuclear to cytoplasmic in
helix-zipper (bHLHZ) transcription factors controls the expres- differentiating neurons of the neural crest, retinal ganglion
sion of a large number of target genes and noncoding RNA cells, neurons of spinal ganglia (Wakamatsu et al., 1993, 1997),
loci. These Myc targets mediate the physiological effects of and Purkinje cells (Okano et al., 1999; Wakamatsu et al.,
Myc on cell proliferation, metabolism, apoptosis, growth, and 1993). Cytoplasmic Myc was also reported in tumors with
differentiation (Eilers and Eisenman, 2008). To promote tran- diverse origins (Bai et al., 1994; Calcagno et al., 2009; Pietilai-
scriptional activation at target genes, Myc forms heterodimers nen et al., 1995). These studies relied on immunostaining pro-
with its partner Max and recruits chromatin-modifying com- tocols and the form of the Myc protein involved was not char-
plexes to E-box-containing promoters. Myc is also involved in acterized.
transcriptional repression through the inhibition of the transcrip- Interestingly, association of Myc with several cytoplasmic
tional activator Miz1 (Kleine-Kohlbrecher et al., 2006). Aberrant proteins has been reported. The best characterized is the
elevation of Myc levels has been shown to contribute to the interaction of c-Myc with tubulins (Alexandrova et al., 1995)
genesis of many types of human tumors (Hanahan and Wein- (Koch et al., 2007; Niklinski et al., 2000). Myc has also been
berg, 2000). reported to interact with other proteins that are predominantly

480 Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc.


A B IB: anti Myc N262 C IB: anti Myc 9E10
IB: anti Myc 143
c-Myc c-Myc c-Myc

Vector
Sparse Dense Cultures
Cell density Cell density
+ - + c-Myc
62
62 62 Myc
Myc Myc 49
49 49
Myc-nick 38
Myc-nick 38
38 28
17
14
Tubulin Tubulin
Tubulin
Total lysates
Sin3A Sin3A
50 5 .5 50 50 5 .5 X 105 Cells 50 5 .5 50 5 .5 X 105 Cells

Nuclear Cytoplasmic Nuclear Cytoplasmic

D IB: anti Myc N262 E


Nuclear Cytoplasmic Myc-nick
Myc antibodies
2

N262 ....................................... +
26

26
G

G
A

IP:
Ig

Ig
H
N

H
N

Myc
143......................................... +
274................................ +
Myc-nick 9E10 -
MycS
C19 -

Max

F
Sparse Dense
c-Myc
DAPI

20µM

Ab: N262 9E10

Figure 1. Identification of Myc-Nick in the Cytoplasm of Cells Grown at High Density


(A) Total cell lysates of Rat1 myc null fibroblasts infected with c-Myc or empty retroviral vectors were prepared for western blot by adding boiling sample buffer.
(B and C) Nuclear and cytoplasmic fractions of HFF cells expressing c-Myc were prepared 48 hr after plating at the indicated increasing densities.
(D) Immunoprecipitation of HA-c-Myc (N-terminal tag) with anti-N262, anti-HA, and normal IgG from nuclear and cytoplasmic fractions of HFFs. Note that Max is
only coimmunoprecipitated along with nuclear c-Myc.
(E) Schematic representation of antibody mapping.
(F) HFF cells infected with c-myc-expressing retrovirus were cultured for 4 days after reaching confluency (middle and right panels) and compared with a
subconfluent culture (left panel) by immunofluorescence using N262 and 9E10 antibodies.
See also Figure S1.

cytoplasmic, such as cdr2 (Okano et al., 1999) and AMY-1 (Taira RESULTS
et al., 1998). However the nature of the cytoplasmic Myc protein
and its potential function remains an enigma. Here we report the Myc-Nick Is a Truncated Form of Myc Localized
identification of Myc-nick, a cytoplasmically localized cleavage Predominantly in the Cytoplasm
product of Myc, and provide evidence for its role in cytoskeletal While studying regulation of c-Myc degradation, we noticed an
organization and cell differentiation. inverse correlation between the levels of full-length c-Myc and

Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc. 481


a cytoplasmic 42 kDa protein in anti-Myc immunoblots derived and Lactacystein failed to block the formation of Myc-nick
from confluent fibroblast cultures (Figures 1A and 1B). As in vitro and in vivo (Figures 2G and 2H). In addition, inhibition
described below, this protein, which we have named Myc-nick, of either the Fbw7 or Skp2 degradation pathways, known to be
is a cytoplasmic cleavage product of full-length c-Myc gener- responsible for proteasomal Myc turnover, had no effect on
ated at high cell density (Figure 1B). Myc-nick is recognized by Myc-nick formation (data not shown). Moreover, incubation of
three antibodies against the N-terminal two-thirds of c-Myc Myc with 20S and 26S proteasomes failed to produce Myc-
(anti-Myc N262, 274, 143; Figures 1A and 1B and Figure S1A nick (not shown). These results indicate that full-length c-Myc
available online) but not by anti- C-terminal antibodies (anti- is converted into Myc-nick by a cytoplasmic protease that is
Myc 9E10, C19; Figure 1C). Furthermore, an anti-HA antibody independent of the proteasome.
immunoprecipitates Myc-nick from cytoplasmic extracts of cells
expressing N-terminally HA-tagged c-Myc (Figure 1D). In addi- Myc Is Cleaved by a Calcium-Activated Calpain
tion, cytoplasmic Myc bearing N-terminal but not C-terminal to Produce Myc-Nick
epitopes is detected by imunofluorescence in confluent cultures In a systematic search for the proteases mediating cytoplasmic
(see below, Figure 1F). Together, these results indicate that cleavage of Myc, we ruled out both caspases and lysosomal
Myc-nick is a truncated protein lacking the C-terminal portion proteases on the basis of inhibitors and cleavage conditions
of c-Myc while preserving an intact N terminus comprising (Figure S2C). However, we found that all calpain inhibitors tested
Myc boxes I–III (Figure 1E). This makes Myc-nick distinct from including calpeptin, calpain inhibitor XII (Figure 3A), and calpain
any other previously identified form of Myc (see Introduction). inhibitor VI (Figure 3B) blocked the formation of Myc-nick in vitro
We have detected endogenously expressed Myc-nick in the and in vivo. Moreover, MG132, although well known as a protea-
cytoplasm of a large number of cell lines including human some inhibitor, has also been shown to inhibit calpains (Mailhes
foreskin fibroblasts (HFFs), Wi38, L cells, HCT116, SW480, HeLa et al., 2002). Calpains had been linked to Myc stability earlier, but
(Figure S1B), C2C12 (Figure 7D), 293T, A431, Rat1, U2OS, ES the antibodies used in those studies would not have detected
cells, and mouse neurospheres (not shown), in addition to mouse Myc-nick (Gonen et al., 1997; Small et al., 2002). Calpains
tissues such as muscle, brain, and cerebellum (Figure 7A and comprise a large family of calcium-dependent cytoplasmic
Figure S7A). We also observed Myc cytoplasmic localization cysteine proteases that function at neutral pH and are primarily
by immunofluorescence of confluent cultures of HFFs (Figure 1F associated with partial protein cleavage rather than complete
and Figure S5A) and Rat1 myc null cells (not shown) expressing protein degradation. The most well studied members of this
c-myc. Therefore a protein with the size and properties of Myc- family are mcalpain and mcalpain. These ubiquitously expressed
nick is very widely expressed. In some settings we observe catalytic subunits form functional heterodimers with a calpain
relatively low and variable amounts of Myc-nick in the nucleus regulatory subunit (calpain r) to bind calcium. To determine
(e.g., Figure 2A). whether Myc-cleavage is regulated by calcium-activated cal-
pains, we employed siRNA to knock down calpain r in cells
Myc-Nick Is Generated by Proteolytic Cleavage expressing c-Myc either endogenously or under control of
of c-Myc in the Cytoplasm a retroviral vector. In both settings a partial silencing of calpain
We considered the possibility that Myc-nick is generated in the r correlated with decreased Myc-nick and increased full-length
cytoplasm because the nuclear export inhibitor Leptomycin B c-Myc (Figure 3C). Knockdown of m or mcalpains alone did not
had no effect on the production or cytoplasmic localization of block the formation of Myc-nick, most likely due to the presence
Myc-nick (Figure 2A). To determine whether a cytoplasmic of other redundant calpains (not shown). Because calpain
activity could convert full-length c-Myc into Myc-nick, we incu- activity is calcium dependent we next examined the effects on
bated in vitro-translated [35S]-methionine-labeled c-Myc or puri- Myc-nick of modulating calcium levels. Treatment with the
fied full-length recombinant c-Myc with nuclear or cytoplasmic calcium chelators Bapta or EGTA reduced the ability of CE to
extracts from Rat1 myc null cells. Only cytoplasmic extracts cleave c-Myc (Figures 3D and 3E). Incubation of either IVT
(CE) were capable of producing a protein (Figures 2B–2D) having c-Myc (Figure 3F) or recombinant c-Myc (Figure 3G) with purified
the same apparent molecular weight as Myc-nick that was m or mcalpain (together with calpain r) produced Myc-nick.
recognized by antibodies against the N terminus but not C These results demonstrate that Myc-nick is directly generated
terminus of c-Myc (Figure S2A). Increasing the incubation time by calcium-dependant calpain cleavage of full-length c-Myc.
with CE augmented Myc-nick production and decreased the
amounts of full-length c-Myc input (Figure 2C). In agreement Lysine 298 Is the Primary Calpain Cleavage Site in c-Myc
with our observations made in vivo, CE of dense cultures were To map the calpain cleavage region on c-Myc, we used a series
more efficient in cleaving c-Myc than CE of sparse cultures of internal deletion mutants lacking 60 residue segments (c-Myc
(Figure S2B). In experiments designed to characterize the cyto- DA-DG) (Tworkowski et al., 2002) (Figure 3H) and determined
plasmic activity responsible for formation of Myc-nick, we found whether any failed to generate a shorter c-Myc protein. Only
that inhibitors of transcription or translation had no effect on the deletion of residues 252–315 (DE) resulted in loss of a Myc-
Myc-nick formation (Figure 2E) whereas heating or adding nick-like product (Figure 3I). This region contains a high scoring
protease inhibitors to the CE blocked Myc-nick, consistent PEST domain, often present in unstable proteins and frequently
with proteolysis (Figure 2F). Whereas MG132, an inhibitor of the associated with calpain cleavage sites. We further narrowed our
proteasome (and other cysteine proteases), blocked Myc nick search to residues 270–315, based on the ability of antibody 274
formation, specific proteasome inhibitors such as Epoxomycin to detect Myc-nick (Figure S1A and Figure 3H). Although there is

482 Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc.


A B C - - - - - - + MG
IB: N262 - - - - - + - NE
- - - + CE 10' 1 2 4 16 16 16 hours
Nuclear Cytoplasmic - + - - NE + + + + + - + CE
0 10 30 0 10 30 ng LB
Myc Myc Myc
Myc-nick Myc-nick Myc-nick

D E F
- - + - - - Boiled CE
mg/µl CHX mg/µl ActD - - - - - + PI
yc
W

.01 .1 .5 .01 .1 .5
- + + - + + CE
M
M

- + + CE - + + + + + + + CE

Myc Myc Myc


Myc-nick Myc-nick Myc-nick

Silver IB: 143 1 2 3 4 5 6 7 8

G H IB: N262
µM MG132 mM Lacta mM Epoxo MG132 (80nM) Lacta (200nM) Epoxo (400nM)

0 1 10 100 0 0.1 1 10 0 0.1 1 10 - + - + - + - + - + - +

Myc
Myc-nick

N C N C N C
in vitro
in vivo

Figure 2. Myc-Nick Is a Product of c-Myc Cytoplasmic Cleavage Independent of the Proteasome


(A) CRM1-dependant nuclear export is not involved in the formation or localization of Myc-nick. Rat1 myc null cells infected with c-Myc were treated with
leptomycin B (LB) for 4 hr before harvesting.
(B) c-Myc gives rise to Myc-nick in vitro when incubated with cytoplasmic extracts. Radiolabeled IVT c-Myc was incubated for 2 hr with cytoplasmic (CE) or
nuclear extracts (NE) from Rat1 c-myc null cells.
(C) Timecourse of in vitro cleavage of c-Myc.
(D) Recombinant c-Myc is cleaved in the presence of CE. One microgram of recombinant c-Myc was incubated with 30 mg of CE and processed for western blot.
(E) IVT c-Myc was incubated with CE for 4 hr in the presence of of Actinomycin D (ActD) or cyclohexamide (CHX).
(F) The cleavage of c-Myc is inhibited by protease inhibitors and by heat inactivation. CE was boiled prior to incubation with IVT c-Myc and the protease inhibitor
(PI) was added to the incubation mixture.
(G and H) The cleavage of c-Myc is inhibited by MG132, but not by Lactacystein or Epoxomycin. (G) IVT c-Myc was incubated with CE, in the presence of
increasing amounts of MG132, Lactacystein, and Epoxomycin for 1 hr. (H) Rat1 myc null cells expressing c-Myc were incubated with MG132, Lactacystein,
or Epoxomycin for 2 hr prior to harvesting. Nuclear (N) and cytoplasmic (C) fractions are shown.
See also Figure S2.

no universal consensus for a calpain cleavage site, a comparison are cleaved (Figures S3B and S3C). To determine whether this
of 106 sites present in 49 known calpain substrates indicated region functions as a calpain cleavage site, we synthesized
a preference for calpain cleavage after K or R, and to lesser a 10 amino acid peptide corresponding to the putative c-Myc
extent Y, especially when these amino acids are flanked by P, calpain cleavage region (291–300) and to another nearby region
V, and L (Figure 3J) (Tompa et al., 2004). We noted a region local- of c-Myc (236–245) (Figure 3H, in red and blue, respectively) and
ized C-terminal to the PEST domain containing the sequence asked whether they could act as competitive inhibitors of Myc-
PLVLKRC (Figure 3H, marked in red). This region is evolutionarily nick formation in vitro. Addition of increasing amounts of the
conserved in c-Myc and N-Myc but not L-Myc (Figure S3A), peptide containing the putative calpain cleavage site blocked
consistent with the fact that c-Myc and N-Myc, but not L-Myc, the formation of Myc-nick in vitro whereas the control peptide

Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc. 483


A B C Figure 3. Myc-Nick Is Generated by Calpain
μM Calpeptin μM inhibitor XII IB: N262 IB: N262 Cleavage of Full-Length Myc
0 0 0.1 1 10 100 0 1 10 100 N C HCT116 HFF-Myc
- + - + Reg siRNA
(A) IVT c-Myc was incubated with CE for 1 hr in the
- + + + + + + + + + CE - + - + inhibitor VI
Myc Myc Myc presence of the calpain inhibitors calpeptin and
Myc-nick Myc-nick Myc-nick calpain inhibitor XII.
Tubulin Calpain r (B) Dense cultures of HFF cells infected with a
Sin 3A Tubulin c-Myc retroviral vector were incubated with cal-
pain inhibitor VI for 2 hr prior to harvesting.
D (C) siRNA for calpain regulatory subunit (Reg)
E F μ m Calpain
0 1 1 1 ng Calpain
G IB: 143 reduces the formation of Myc-nick in HCT116
- + - Bapta 0 0 .1 1 10 100 μM EGTA - - + - - Calpeptin + + Purified c-Myc and HFF-Myc cells.
- + + CE - + + + + + CE - + - - - CE - + μCalpain (D) Rat1 myc null cells were incubated for 2 hr in the
Myc Myc Myc
presence of the calcium chelant Bapta, and then
Myc
Myc-nick Myc-nick Myc-nick cytoplasmic extracts were prepared and incubated
Myc-nick
with IVT c-Myc (as in 2B).
(E) IVT c-Myc was incubated with CE for 1 hr in the
presence of increasing amounts of EGTA.
(F) IVT c-Myc was incubated with purified recombi-
H nant mcalpain or mcalpain and r subunit for 1 hr
1 63 126 189 252 315 378 439 on ice.
MBI MBII MBIII NLS BHLH LZ

A B C D E F G (G) One hundred nanograms of purified mcalpain


and r subunit was incubated with 2 mg of recombi-
274 antibody
SPEPLVLHEETPPTTSSDSEEEQEDEEEIDVVSVEKRQAPGKRSESGSPSAGGHSKPPHSPLVLK RCHVSTHQHNYAAPPST nant c-Myc for 30 min on ice.
* ** * (H) Schematic representation of c-Myc protein
PEST domain
indicating A-G deletion regions (deletion end-
points indicated above), putative calpain cleavage
I K region in red.
PHSPLVLKRC SPEPLVLHEE
(aa 291-300) (aa 236-245)
(I) c-Myc and the deletion mutants DA-DG were
c

ΔG
c- c ΔC

M D
M A
M B

M E
M F
c- My

J
c- yc Δ
c- c Δ

c- Δ
c- c Δ
c- yc Δ

0 .1 1 10 μg expressed in 293T cells, and 48 hr later the pres-


c- yc

0 .1 1 10
c-

yc

yc
y
y

y
M
M

M
A-

- + + + - + + + CE
ence of a Myc-nick-like protein in cytoplasmic
H

P3 P2 P1 extracts was determined.


V,L,F L,V RKY
V L K Myc (J) Amino acid preference for calpain cleavage
Myc-nick region according to Tompa et al. (2004). P1–P3
IB: 143+274
indicate the position of preferred residues in rela-
tion to the cleavage site, bold letters indicate the
c-Myc calpain cleavage site.
(K) IVT c-Myc was incubated with CE in the pres-
L M N IB: 143+274
ence of a peptide that contains the potential
- + - + Δ291-300 calpain cleavage site (amino acids 291–300 in
98 0

WT L295A K298A R299A


K2 -30
K2 A
A

+ - + -
W or

Wt c-Myc red) or a nearby sequence (amino acids 236–245


91

97
ct
T
Δ2

0 2 4 0 2 4 0 2 4 0 2 4 h incubation
Ve

- - + + CE
in blue).
Myc Myc Myc
Myc-nick (L) WT and D291–300 IVT c-Myc were incubated
Myc-nick Myc-nick
with CE for 1 hr (as in 2B).
(M) IVT WT, L295A, K298A, and K299A c-Myc were
incubated with CE for the indicated time points.
(N) Cleavage products produced from the indi-
cated c-Myc mutants in 293T cells.
See also Figure S3.

had no effect (Figure 3K). Next, we generated a deletion mutant Myc that become more pronounced in the cleavage-deficient
of Myc lacking residues 291–300 and found it to be resistant to mutants.
cleavage by CE (Figure 3L). When this mutant was ectopically
expressed in 293T cells the cleavage was also reduced in Expression of Myc-Nick Alters Cell Morphology
comparison to the wild-type (WT) c-Myc (Figure 3N), indicating and Increases Acetylation of a-Tubulin
that this is the major calpain cleavage site within c-Myc. Next, To study Myc-nick function, we generated a truncated form of
we made point mutations in the calpain cleavage region (labeled Myc containing amino acids 1–298 (referred to as Myc-nick*).
with asterisks in Figure 3H) and assessed their cleavage. We We found that ectopically expressed Myc-nick* is localized
found that the K298A mutation reduced the cleavage of c-Myc predominantly to the cytoplasm and migrates on SDS-PAGE
in vitro (Figure 3M) and in vivo when ectopically expressed in with the same apparent molecular weight as Myc-nick generated
293T cells (Figure 3N). Although K298 appears to be the major by cleavage of full-length c-Myc (Figure 4A). Myc-nick is
calpain cleavage site, a K298A mutant was still cleaved in vivo degraded at a comparable rate to full-length c-Myc with a half-
most likely because when this residue is mutated the cleavage life of about 30 min (data not shown). Because Myc-nick
is shifted to R299 (and perhaps L297, V296). We also identified contains the MBI phosphodegron, with its GSK3b phosphoryla-
weaker calpain cleavage sites localized in the C terminus of tion and Fbw7-binding sites, we would expect Myc-nick to be

484 Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc.


A B
IB: N262

vector Myc Myc-nick*


- - + - MG132
Myc
C
Myc-nick

Myc Vector Myc-nick * Myc-nick *


N

α-tubulin
Myc-nick
1 2 3 4

DAPI/α
Vector Myc-nick *

60μM
Vector Myc Myc-nick* Myc-nick*

Figure 4. The Expression of Myc-Nick* Promotes Changes in Cell Morphology


(A) Myc-nick* (1–298) is cytoplasmic and has the same apparent molecular weight as Myc-nick derived from full-length c-Myc (compare lanes 2 and 4, upper
panel). Rat1 myc null cells expressing empty vector, full-length c-Myc, and Myc-nick were fractionated into nuclear (N) and cytoplasmic (C) fractionations.
(B) Myc-nick*-expressing cells extend protrusions at the wound edge. A confluent monolayer of Rat1 myc null cells expressing either vector or Myc-nick was
scratched using a 100 ml tip and phase contrast images were taken at 12 hr.
(C) Rat1 myc null cells expressing empty vector, c-Myc, and Myc-nick at 14 days after selection.
See also Figure S4.

degraded through similar proteasomal pathways as full-length bodies against a-tubulin and acetylated a-tubulin. Figure 5A
Myc. Indeed, blocking proteasome activity by pharmacological shows that although acetylated a-tubulin immunostaining
inhibitors or by silencing E3 ligases and components of the pro- in vector and c-Myc-expressing cells is low, Myc-nick* cells
teasome induced the stabilization of both full-length Myc and display intense staining of elongated structures (Figures 5A
Myc-nick (data not shown). Calpain inhibitors are incapable of and 5F). This was confirmed by immunoblotting for acetylated
inducing accumulation of Myc-nick, indicating that calpains are a-tubulin in Rat1 myc null or 293T cells expressing Myc-nick*
unlikely to play a major role in Myc-nick turnover in vivo. (Figure 5C, Figure S6B). Importantly neither the levels of total
Whereas overexpression of full-length c-Myc in Rat1a myc cellular acetylated lysine (Figure S6A) nor the levels of tyrosi-
null fibroblasts is associated with increased proliferation nated tubulin (not shown) were affected. To ask whether Myc-
and apoptosis, the ectopic expression of Myc-nick* had no nick* expression increased microtubule stability, we treated cells
detectable effect on either cell doubling time or survival (data expressing Myc-nick*, c-Myc, or vector with nocodozole for
not shown). However, Myc-nick*-expressing cells displayed 15 min. This treatment disrupted unstable microtubules with
dramatic morphological changes—they appeared spindle-like a half-life of about 10 min but not stable microtubules with
with long protrusions that occasionally formed intercellular con- a half-life >2 hr. Only Myc-nick*-expressing cells possessed
tacts (Figure 4C, Figure S4, Figure S5C). This morphology was microtubules that survived nocodozole treatment and these
specific for Myc-nick* expression and could not be produced stained strongly with antiacetylated a-tubulin (Figure 5B).
by expressing the C-terminal 100 residues of c-Myc (not shown).
Introducing a scratch wound across a confluent cell monolayer Myc-Nick Interacts with a- and b-Tubulins
showed that whereas control cells migrated into the wound by We observed partial cytoplasmic colocalization between Myc
extending lamellipodia, Myc-nick*-expressing cells aligned and a-tubulin in several cell types expressing c-Myc or Myc-
parallel to each other and extended long protrusions into the nick (Figure 5E; Figures S5A–S5C). Using 293T cells expressing
wound (Figure 4B). GFP-tubulin and Myc-nick we performed immunoprecipitation
The morphological changes induced by Myc-nick* are with anti-GFP and immunobloted with anti-Myc. Myc-nick was
suggestive of altered cell-cell contacts and/or major cytoskeletal coimmunoprecipitated with GFP-tubulin but not GFP-EB1,
reorganization. The elongated cellular protrusions present in a microtubule-binding protein (Figure S5D). In addition, immuno-
Myc-nick-expressing cells resemble specialized structures precipitation of either c-Myc or endogenous a-tubulin from cyto-
formed by stable microtubules. Because stable microtubules plasmic extracts indicated that Myc-nick interacts with a-tubulin
display increased acetylation of a-tubulin on lysine 40 (Hubbert in vivo (Figure 5D). To examine in vitro interactions we incubated
35
et al., 2002), we stained Myc-nick*-expressing cells with anti- [S]-methionine-labeled IVT c-Myc with purified brain tubulins

Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc. 485


A Figure 5. Myc-Nick* Cells Display In-
Vector c-Myc Myc-nick* creased Levels of Acetylated a-Tubulin
and Microtubule Stabilization
α−tubulin/DAPI

(A) Immunofluorescence for a-tubulin (upper


panels) and acetylated a-tubulin (lower panels) of
Rat1 myc null cells infected with empty vector,
c-Myc, or Myc-nick*.
(B) As for (A) but incubated in the presence of
nocodazole for 15 min prior to fixation.
Vector c-Myc Myc-nick* (C) Immunoblotting of Rat1 cell extracts using anti-
acetyl. α−tub/DAPI

bodies against the indicated proteins.


(D) NE or CE of Rat1 myc null cells express-
ing c-Myc were immunoprecipitated with anti
a-tubulin or normal mouse IgG and immunoblotted
for Myc (top panel), or immunoprecipitated with
60μM anti-c-Myc N262 antibody, and immunoblotted
with anti-a-tubulin.
B (E) Immunofluorescence for Myc and acetylated
Vector c-Myc Myc-nick*
a-tubulin in A431 lung epithelial cells cell trans-
fected with Myc-nick.
α−tubulin

(F) Detail of Myc-nick-expressing cell stained for


10μM Nocodazole 15'

acetylated a-tubulin.
See also Figure S5.

Vector c-Myc Myc-nick*


recruits an acetyltransferase to microtu-
acetyl. α−tub

bules. We observed marked acetylation


of a-tubulin in cytoplasmic extracts incu-
bated in the presence of either recombi-
nant c-Myc (Figure 6A) or IVT c-Myc
60μM (Figure 6B and Figure S6C). Importantly,
Myc proteins are known to associate
C
yc k*
k*

Myc-nick*
M nic
ic

D with the acetyltransferases GCN5 and


-n

E F
M c
y
-
ro

yc
M
Pu

N C N C N C
c-

acetyl α-tub
TIP60 (Frank et al., 2003; Sterner and
IB: Myc

Myc
Berger, 2000) via TRRAP that interacts
α-tubulin
Myc-nick
*
acetyl α-tub with MBII (residues 106–143) (McMahon
Input α-tub IgG
γ-tubulin IP et al., 1998). We therefore tested a Myc-
nick deletion mutant lacking Myc box II
IB: α-tub

actin N C N C
α-tubulin Merge
HDAC 3
(DMBII) and found that its ability to pro-
Input IP:Myc
mote a-tubulin acetylation was reduced
HDAC 6 10μM
(Figures 6C and 6D). In addition, the
acetyl. α−tub/DAPI
DMBII Myc-nick* mutant failed to induce
the cell morphological changes that we
for 1 hr, then immunoprecipitated a, b, g, bIII and acetylated had detected with WT Myc-nick* whereas a comparably sized
a-tubulin and exposed the gel to detect radioactive Myc. Full- deletion in a region adjacent to MBII was similar to WT (Figure 6E).
length c-Myc was coimmunoprecipitated mostly with b-tubulin The dependence on MBII for tubulin acetylation and altered
(Figure S5E, middle panel). We also incubated IVT c-Myc with cell morphology suggests involvement of the acetyltranferases
CE for 1 hr to produce Myc-nick and then performed immuno- known to bind this region. We detected substantial amounts of
precipitation for tubulins as above. The results showed that GCN5 and TRRAP in the cytoplasm (Figure S6D), whereas
Myc-nick interacts with a- and b-tubulin (Figure S5E, upper Tip60 was predominantly nuclear (not shown). To test GCN50 s
panel). The binding of Myc-nick to tubulin is consistent with the involvement in a-tubulin acetylation we performed siRNA-medi-
previous report of an N-terminal region of Myc capable of asso- ated knockdown of TRRAP and GCN5 in Myc-nick*-expressing
ciating with tubulin (Alexandrova et al., 1995). cells and found that decreasing either one of these proteins
reduced the levels of acetylated a-tubulin (Figure 6F) and
Myc-Nick Directly Regulates a-Tubulin Acetylation reverted the changes in cell morphology induced by Myc-nick
through GCN5 to that of vector-infected cells (Figure 6G). Moreover, GCN5
Because Myc-nick lacks the Myc C-terminal dimerization and coimmunoprecipated with a-tubulin and Myc in cytoplasmic
DNA-binding domains, we surmised that the increase in acety- extracts of 293T cells transfected with GCN5 (Figure 6H).
lated a-tubulin induced by Myc-nick* expression is independent Both ectopic expression of GCN5 in 293T cells (Figures 6D
of Myc transcriptional activity. One possibility is that Myc-nick and 6I) and addition of full-length recombinant GCN5 to

486 Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc.


I
BI
*
ni ick
A C E

M

c- r

-n
M c
o
y
ct

yc

ck
M
+ + + + CE Vector Myc-nick*

ve
- - 0.1 1 μg c-Myc acetyl α-tub

in vitro CE
- + + + GTP
acetyl α-tub. α-tubulin

Myc IVT

I
BI
B D

N *
G ick

M
Myc-nick* Δ106-143


ni 5
M r
-n
o
+ + - - vector IVT

ct
yc

ck
Myc-nick* Δ145-160

C
(ΔMBII)

ve
- - + + c-Myc IVT
acetyl α-tub

in vivo 293T cells


acetyl α-tub
α-tubulin
α-tubulin Myc-nick
GCN5

F G
G P
TR l
ro

5
A
N
R
nt

si RNA
C
co

Control si RNA GCN5 si RNA TRRAP si RNA


TRRAP

GCN5

acetyl α-tub

α-tubulin 60μM

Myc-nick

HDAC6

H J L - + + + + + - microtubules 5μg
- - - - + + + GCN5 0.1μg
r
5

GCN5
to
N
c

+ - + + - + - c-Myc 0.5μg
C
Ve

- - - + + + c-Myc 0.2μg
G

GCN5
10% input
acetyl α-tub
Myc *
30μg CE

1.0 1.6 1.9 1.8 2.4 2.7 fold change Tubulin


IP: α-tubulin
IB: GCN5 p300
IP: Myc - - - + + + c-Myc 0.2μg GCN5 catalyctic
IB: GCN5 acetyl α-tub Ponceau domain
1.0 1.1 1.1 2.4 2.6 2.5 fold change

acetyl α-tub
I K
GCN5
G r
5
o
N
ct
C
Ve

- - - + + c-Myc
acetyl α-tub α-tubulin
5μg microtubules

acetyl α-tub
1.3 2.7 1.0 3.1 4.2 fold change β-tubulin
GCN5 + p300 1 2 3 4 5 6 7
- - + + c-Myc
α-tubulin acetyl α-tub
0.9 1.0 1.2 1.1 fold change

Figure 6. c-Myc and the HAT GCN5 Promote a-Tubulin Acetylation


(A) CE was incubated with recombinant c-Myc and c-Myc dilution buffer or (B) with c-Myc IVT or vector IVT for 1 hr at 37 C. The samples were immunoblotted as
indicated.
(C) CE was incubated with IVT vector, c-Myc, Myc-nick, and Myc-nick DMBII (D106–143) for 30 min at 37 C, then processed for immunoblotting.
(D) 293T cells were transfected with empty vector, GCN5, Myc-nick, and Myc-nick DMBII and processed for immunoblotting after 48 hr.
(E) Rat1 myc null cells were infected as indicated and photographed 10 days after selection.
(F and G) Rat1 myc null cells expressing Myc-nick were transfected with 100 nM of control, TRRAP, or GCN5 siRNA and 76 hr later processed for immunoblotting
(F) or photographed (G).
(H) CE of 293T cells transfected with empty or GCN5 vectors were immunoprecipitated with anti-a-tubulin or anti-Myc (143+274) and immunoblotted for GCN5.
(I) 293T cells were transfected with control or GCN5-expressing vectors and 48 hr later processed for immunoblotting.
(J–L) GCN5 induces acetylation of a-tubulin. (J) CE were incubated with 200 ng of Myc, 100 ng or 300 ng of GCN5 (upper panel), or 100 ng or 500 ng of p300 (lower
panel). (K) Assembled microtubules were incubated with 100 ng or 500 ng of recombinant full-length GCN5 (upper panel) or p300 (lower panel) in the presence or
absence of 200 ng of purified c-Myc. (L) Purified assembled microtubules were incubated with recombinant c-Myc and GCN5-catalytic domain. The asterisk
indicates a nonspecific bacterial band copurified with GCN5-catalytic domain.
See also Figure S6.

cytoplasmic extracts (Figure 6J, upper panel) resulted in nant GCN5 also induced the acetylation of a-tubulin present
increased levels of acetylated a-tubulin. The addition of Myc in assembled microtubules (Figure 6K) and synergized with
further increased the levels of a-tubulin acetylation induced by c-Myc to promote further acetylation (Figure 6K, compare
GCN5 in cytoplasmic extracts (Figure 6J). Full-length recombi- lanes 1 to 4 and 2 to 5). The GCN5 catalytic domain alone was

Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc. 487


A B GM DM
C D GM DM
1 2 3 8 16 weeks S D D Density primary myoblasts S M D D Density

α T58P α N262
Myc

primary mouse myoblasts


Myc 1.8
Myc

Calpain activity
1.6
Myc-nick 1.4
Myc-nick
Myc-nick TroponinC 1.2
1.0 Myc
Calpain3 0.8 Myc-nick
Tropomyosin 0.6
acetyl α-tub 0.4 Tropomyosin
mouse hindlimb muscles
0.2 Tubulin
Tubulin
GM DM C2C12

E F
c-Myc c-Myc/DAPI acetyl α-tub /DAPI acetyl α-tub /DAPI G
Rhabdomyosarcoma
HFF RHJT RD RH1
S D S D S D S D

20μM 60μM alveolar embryonal


C2C12 DM GM DM Subtype
ni r

H J L M
o
*
ct
ck
ve

Vector Myc-nick Vector GM DM


human myoblasts (pectoral)

Troponin C
- + - + Myc-nick*
(rectus abdominus)

Desmin
TroponinC
human myoblast

Myogenin
Myc-nick* Tropomyosin
acetyl α-tub
Myogenin
α-tubulin
C2C12

C2C12
Caveolin 3
I
r

K
BI
to

ΔM *

I
ck
c
or

ve
ni
*
ct
ck

Myc-nick*
RD Rhabdomyosarcoma

Troponin C
ve

ni

Desmin
(rectus abdominus)

Troponin C Tropomyosin
human myoblast

acetyl α-tub acetyl α-tub


Myc-nick*
Myc
MyoD
α-tubulin nick*
nick* ΔMBII
α-tubulin 60μM α-tubulin
N
O myc -/- myc +/+
TroponinC /DAPI

C MyoD MyoD C
M
M
M
M
M
M

-S
-S

G
G
D
G
G
D

TroponinC
Myogenin
60μM α-tubulin

P nick MyoD MyoD+nick


Phase

M
M

M
G

G
D

D
Rat1 myc -/-

TroponinC
Myogenin
20μM α-tubulin
MyoD MyoD+Myc-nick*
Rat1 myc+/+

Figure 7. Myc-Nick Accelerates Muscle Cell Differentiation


(A) Hindlimb muscles dissected from 1-, 2-, 3-, 8-, and 16-week-old mice were processed for immunoblotting using N-terminal anti-Myc sera (anti 143+274).
(B) Mouse primary myoblasts isolated from hindlimb muscles of 8-week-old mice were cultured as sparse cultures for 24 hr or as dense cultures in the presence of
either growth medium (GM) or differentiation medium (DM) for 3 days. Total cell extracts were immunoblotted for c-Myc (anti 143+274) and indicated proteins.
(C) Mouse primary myoblasts were cultured in GM or DM for 3 days, lysed in buffer A, and total calpain activity was measured using Suc-LLVY-AMC synthetic
substrate (n = 2; calpain activity in DM was compared with calpain activity in GM; set to 1 ± standard error of the mean [SEM]).
(D) C2C12 mouse myoblasts cultured at sparse (S), medium (M), or high densities (D). Dense cultures were harvested or switched to DM for 7 days. Total cell
extracts were immunoblotted for c-Myc using antibodies against total c-Myc (N262) or against phosphorylated T58 Myc, a signal for Myc degradation.
(E) C2C12 cells cultured in DM for 7 days and stained for endogenous c-Myc (anti-N262).
(F) C2C12 cells grown in the presence of GM or DM for 5 days and stained with anti-acetylated a-tubulin.
(G) Western blotting for Myc in rhabdomyosarcoma cell lines grown as dense or sparse cultures for 3 days.
(H and I) Human myoblasts expressing vector, or Myc-nick were cultured in DM and processed for immunoblotting after 4 days.
(J) Human myoblasts expressing vector or Myc-nick were cultured in DM and photographed after 2 days.

488 Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc.


sufficient to induce a-tubulin acetylation, but no synergy with When stimulated to differentiate, these cells displayed low
Myc was detected, most likely because association between levels of full-length c-Myc and high levels of Myc-nick when
these two proteins occurs outside of GCN50 s active site (Fig- compared to undifferentiated cycling cells (Figures 7B and 7D;
ure 6L). As a further control, we tested p300, a HAT that binds Figure S7B). Interestingly, Myc-nick levels were higher in a rhab-
the C terminus of Myc (Faiola et al., 2005). p300 neither induced domyosarcoma cell line from the alveolar subtype (Figure 7G),
tubulin acetylation nor did it synergize with Myc (Figures 6J and a very aggressive tumor derived from partially differentiated
6K, bottom panels). These data indicate that GCN5 can specifi- muscle cells.
cally acetylate tubulin and that Myc augments tubulin acetylation The increased levels of Myc-nick in differentiated primary
by binding to tubulin and recruiting GCN5. mouse myoblasts correlate with an increase in calpain 3 levels
(Figure 7B; Figure S7B) and in total calpain activity (Figure 7C).
Myc-Nick Is Produced in Differentiating Cells Similarly, when C2C12 cells were stimulated to differentiate,
and Tissues they also displayed an increase in total calpain activity (Fig-
When examining the expression of Myc-nick in adult mouse ure S7C) and in the ability to cleave Myc in vitro (Figure S7D).
tissues, we found that brain, cerebellum, and skeletal muscle Importantly, the levels of acetylated a-tubulin were also elevated
express significantly higher levels of Myc-nick than any other during the process of differentiation (Figure 7B; Figures S7B and
tissue (Figure 7A, Figure S7A). Interestingly, both neuronal and S7F). Acetylation of a-tubulin is accompanied by myoblast fusion
muscle differentiation require major cytoskeletal rearrangements to form multinucleated myotubes (Figure 7F; Figure S7F). When
that have been associated with increased microtubule stability myoblasts are stimulated to differentiate they fuse into multinu-
and elevated levels of acetylated a-tubulin. For example, it has cleated myotubes. In addition to displaying increased levels of
been demonstrated that the acetylation of a-tubulin by the acetylated a-tubulin, these myotubes show decreased immu-
Elp3 acetyltransferase is required for proper cortical neuronal nostaining for nuclear Myc and increased cytoplasmic Myc
migration and differentiation (Creppe et al., 2009). Increased staining (Figure 7E). Conversely, undifferentiated satellite cells
levels of acetylated a-tubulin are found during myogenic differ- in the same culture display nuclear staining for Myc (Figure S7E).
entiation (Gundersen et al., 1989). Importantly, inhibiting the This is consistent with our results showing conversion of full-
activity of the deacetylases HDAC6 and Sirt2 (known to deace- length Myc into predominantly cytoplasmic Myc-nick.
tylate a-tubulin) generates augmented levels of acetylated In C2C12 cells, concomitant with increased Myc-nick abun-
a-tubulin and promotes differentiation of myoblasts (Iezzi et al., dance, we observed a decrease in Myc-nick phosphorylation
2004). Furthermore, the presence of the primary cilium, a micro- at threonine 58 (T58) after the switch to differentiation conditions
tubule-based antenna-like structure composed of acetylated (Figure 7D). Phospho-T58 mediates Fbw7-dependent degrada-
a-tubulin, is essential for cardiomyocyte differentiation (Clement tion of Myc by the proteasome. Decreased phosphorylation at
et al., 2009). this site is consistent with the notion that stabilization of Myc-
The extensive cytoskeletal changes that occur during muscle nick contributes to its elevated levels. Interestingly we found
differentiation are regulated by calcium and calpains (Dedieu that Myc-nick phosphorylation at T58 is also reduced in adult
et al., 2004). The inhibition of calpain activity either by pharmaco- muscle, brain, and cerebellum (Figure S7A and data not shown).
logical inhibitors or by overexpression of Calpastatin (an endog- In summary, we found that during muscle differentiation there
enous inhibitor of calpains) blocks muscle cell differentiation is an increase in Myc-nick, concomitant with an elevation in cal-
(Dedieu et al., 2004). In addition, both types of ubiquitous pain activity and tubulin acetylation (Figures 7B–7D; Figure 7F;
calpains (m and m) were shown to regulate muscle cell differen- Figures S7B–7D; Figure S7F).
tiation in vitro (Moyen et al., 2004). Moreover, mutations in the
calpain 3 gene (the muscle-specific calpain) cause limb girdle Myc-Nick Accelerates Muscle Differentiation
muscle dystrophy 2A (LGMD2A) (Richard et al., 1995). Finally, We examined the effects of ectopic expression of Myc-nick* in
knocking out calpain r (the regulatory subunit shared by all human primary myoblasts isolated from the pectoral girdle
calcium-dependent calpains) is lethal due to impaired cardiovas- (Figure 7H) and rectus abdominus (Figures 7I–7J), from the
cular development (Arthur et al., 2000). human rhabdomyosarcoma cell line RD (Figure 7K), and in
To study the cleavage of Myc to Myc-nick during the process C2C12 cells (Figures 7L and 7M). In all cells we found that
of muscle differentiation, we employed human primary myo- Myc-nick* expression accelerated muscle cell differentiation,
blasts purified from pectoral girdle, mouse myoblasts purified augmented a-tubulin acetylation, and elevated expression of
from hindlimb muscles, and C2C12 cultured mouse myoblasts. muscle-specific proteins (Figures 7H–7M).

(K) RD rhabdomyosarcoma cells expressing vector, Myc-nick, or Myc-nick DMBII (D106–143) were grown in DM for 4 days and processed for immunobloting.
(L) C2C12 cells expressing vector or Myc-nick were grown as dense cultures and stimulated to differentiate for 4 days and then photographed.
(M) C2C12 cells expressing vector or Myc-nick were grown to confluency and harvested or stimulated to differentiate for 3 days and then harvested. Total cell
lysates were immunoblotted with antibodies against the indicated proteins.
(N) Rat1 (myc+/+) cells expressing MyoD or MyoD + Myc-nick were cultured in DM for 3 days and photographed (lower panels) or stained for Troponin C and DAPI
(upper panels).
(O) Rat1 (myc+/+) or Rat1 myc null (myc/) cells expressing vector (lane C) or MyoD were cultured in GM, DM, or serum-free medium (lane S) for 4 days and
processed for immunobloting.
(P) Rat myc null (myc/) cells expressing Myc-nick, MyoD, or MyoD + Myc-nick were grown in GM or DM for 3 days and processed for immunoblotting.
See also Figure S7.

Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc. 489


The expression of Myc-nick* in C2C12 myoblasts promoted commonly employed when studying Myc proteins. Third, most
an increase in cell fusion not only in confluent cultures (Fig- studies use total protein or nuclear extracts and have not analyzed
ure 7L; Figure S7H) but also in sparse cultures (Figure S7G). the cytoplasmic pool of Myc. Fourth, Myc-nick may have been
The role of Myc-nick in promoting muscle cell differentiation is mistaken for the similarly sized MycS protein (Hann et al., 1988),
partially dependent on MBII, as the differentiation of RD and a nuclear localized product of internal translation initiation.
C2C12 cells by Myc-nick can be delayed but not inhibited by
the deletion of MBII (Figure 7K; Figure S7I). Calpain Cleavage as a Posttranslational Functional
To examine the requirement for Myc-nick production during Switch
differentiation further, we first tested calpain inhibitor XII and We found that Myc-nick is generated in the cytoplasm by
found that it reduces differentiation and tubulin acetylation calpain-mediated cleavage of full-length Myc. Cytoplasmic
(Figure S7J). Next we employed c-Myc mutant (D291–300), cleavage by calpains has been reported for over 100 proteins
which is deficient in cleavage to Myc-nick. Similar to full-length including many cytoskeletal proteins, membrane receptors,
WT c-Myc, this mutant can induce proliferation, apoptosis, and transcription factors (Tompa et al., 2004). This number is
fibrillarin expression, and phosphorylated H2AX in Rat1 myc probably an underestimate because the lack of a clearly defined
null cells (Figures S7L–S7M). However the D291–300 c-Myc consensus cleavage site for calpains makes the identification of
mutant significantly reduces C2C12 cell differentiation when novel calpain substrates difficult. For most substrates the role of
compared to c-Myc, even though the proteins are expressed calpain cleavage is not known. However, there are several note-
at equal levels. Whereas full-length c-Myc only blocks the worthy examples of calpain cleavage operating as a functional
fusion of C2C12 myoblasts, the c-Myc mutant (D291–300) with switch (Abe and Takeichi, 2007; Yousefi et al., 2006). Here we
reduced ability to generate Myc-nick dramatically reduces the have shown that the cleavage of Myc by calpain converts this
expression of muscle markers in addition to blocking fusion predominantly nuclear transcription factor into Myc-nick, a cyto-
(Figures S7N–S7O). solic factor that regulates a-tubulin acetylation. Based on our
findings and the examples in the literature, we surmise that the
Myc-Nick Renders Rat1 myc Null Cells Competent partial proteolytic cleavage of proteins by calpains functions as
to Differentiate an irreversible posttranslational modification.
MyoD has been long known to induce transdifferentiation in
diverse cell types. We found that MyoD induces expression of A Role for Myc-Nick in a-Tubulin Acetylation
muscle-specific markers in Rat1 fibroblasts but not in Rat1 We have shown that Myc-nick mediates the acetylation of
myc null fibroblasts (Figure 7O), indicating that myc is necessary a-tubulin by forming a complex with microtubules and the HAT
for the transdifferentiation process. Expressing Myc-nick GCN5. GCN5 has been long known to associate with nuclear
together with MyoD in myc+/+ Rat1 fibroblasts further elevated Myc through the highly conserved Myc box II, a region retained
the levels of muscle-specific markers and promoted cell fusion in Myc-nick. Although tubulin acetylation was first described
compared to MyoD alone (Figure 7N). Importantly, we find that 20 years ago, little is known about the enzymes that catalyze
in Rat1 myc null cells, the coexpression of Myc-nick (Figure S7K) this reaction. Recently Elp3, a histone acetyltransferase, was
permitted transdifferentiation to muscle in response to MyoD demonstrated to be critical in acetylation of a-tubulin in cortical
(Figure 7P). This experiment indicates that Myc-nick is sufficient projection neurons, an event linked to neural differentiation and
for MyoD-induced transdifferentiation, supporting the idea that migration (Creppe et al., 2009). We surmise that GCN5 repre-
conversion of full-length Myc to Myc-nick is important in sents another acetyltransferase targeting tubulin. Deacetylation
MyoD-induced differentiation. of a-tubulin is mediated by both HDAC6 and Sirt2 (Hubbert
et al., 2002; Matsuyama et al., 2002; North et al., 2003). However
DISCUSSION the increase in acetylated a-tubulin by Myc-nick does not occur
through modulation of HDAC activity (Figures S6E and S6F).
Recent studies have demonstrated that Myc directly regulates A connection between a-tubulin acetylation and calpain acti-
transcriptional activation through the three RNA polymerases, vation was previously suggested by two independent studies.
as well as transcriptional repression, and DNA replication (Eilers First, calcium depletion, which inactivates calpains, was shown
and Eisenman, 2008). Here we identified and characterized Myc- to decrease the levels of acetylated a-tubulin in epithelial cells
nick, a novel form of Myc performing transcription-independent (Ivanov et al., 2006). Second, the ectopic expression of calpain
functions in the cytoplasm. One of these functions is to regulate 6 causes microtubule stabilization, elevates the levels of acety-
a-tubulin acetylation in cooperation with the HAT GCN5. More- lated a-tubulin, and impairs cytokinesis in HeLa cells (Tonami
over, Myc-nick levels are elevated in differentiated muscle et al., 2007). Our findings suggest that the induction of a-tubulin
tissues and Myc-nick overexpression accelerates muscle cell acetylation by calpains could be mediated at least in part by
differentiation. Myc-nick and GCN5.
Despite its widespread expression, Myc-nick has not been
previously characterized. There are several likely reasons for this. The Role of Myc-Nick in Terminal Differentiation
First, the most commonly used antibodies against Myc (such as The role of Myc family members in differentiation is complex.
9E10) recognize its C terminus and would not detect Myc-nick. Endogenous Myc is strongly downregulated during terminal
Second, the proportion of Myc-nick to Myc increases when differentiation of many cell types. Moreover, ectopic expression
cells are grown as confluent cultures, conditions that are not of Myc has been shown to block terminal differentiation. The

490 Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc.


ability of Myc to negatively regulate differentiation is consistent and recruiting GCN5 to mediate a-tubulin acetylation. Myc-nick
with its role in maintaining pluripotency in ES cells and in the is also likely to have additional functions and partners in the cyto-
generation of induced pluripotent stem (iPS) cells (Cartwright plasm as the deletion of MBII in Myc-nick, which should block
et al., 2005; Takahashi et al., 2007). However, Myc has also the interaction with GCN5, only partially reduced the ability of
been implicated in promoting both proliferation and differentia- Myc-nick to promote muscle differentiation. We predict that
tion in specific cellular contexts such as in progenitor cells of the functions carried out by Myc-nick in the cytoplasm coop-
the skin (Gandarillas and Watt, 1997; Gebhardt et al., 2006), B erate to promote cytoskeletal changes that can further drive
lymphocytes (Habib et al., 2007), and hematopoietic stem cells terminal differentiation. In our model, the cleavage of Myc by
(Wilson et al., 2004). Our data suggest that whereas full-length calpains has two roles. First, it helps diminish nuclear Myc abun-
Myc blocks differentiation at the transcriptional level, Myc-nick dance preventing newly synthesized Myc from entering the
may be involved in promoting differentiation through transcrip- nucleus, therefore eliminating the transcriptional blockade to
tion-independent mechanisms. In agreement with this hypoth- differentiation caused by Myc. Second, the production of Myc-
esis there have been numerous reports of Myc antigenicity nick influences cytoskeletal organization and facilitates terminal
localized predominantly in the cytoplasm of differentiated cells differentiation. We predict that Myc-nick will play a general role
(see Introduction). In addition, we show that ectopic expression linking myc to both nuclear functions and cytoplasmic organiza-
of Myc-nick accelerates muscle cell differentiation and renders tion during differentiation of a wide variety of cell types.
Rat1 myc null cells competent to differentiate into muscle
following introduction of MyoD. EXPERIMENTAL PROCEDURES
A number of studies have shown a strong correlation between
muscle cell differentiation, calcium influx, and calpain activation Retroviral Infection and Transfection
(Dedieu et al., 2004; Kumar et al., 1992). In addition, limb girdle For retroviral production, 293T cells were cotransfected with pBabe-puro
vector expressing Myc clones and amphotropic helper. Infected cells were
muscular dystrophy 2A (LGMD2A) is caused by mutations in
selected with 2 mg/ml puromycin for 4 days. Rat1 myc null cells were harvested
calpain 3 that affect its activity (Richard et al., 1995). During 10–14 days after infection and C2C12 cells were stimulated to differentiate
the process of normal differentiation, myoblasts elongate and after selection. For overexpression experiments, 293T (transfected with
fuse into syncytial myotubes. An early event during this process Fugene-Roche) cells were harvested 3 days after transfection with a change
is the remodeling of the microtubule cytoskeleton, involving in culture medium 24 hr before harvesting. Sparse cultures (S) are about
disassembly of the centrosome and the alignment of stable 20% confluent, medium cultures (M) are about 40%–60% confluent, and
dense cultures (D) are grown as confluent cultures for 2–4 days.
microtubules into a parallel array along the long axis of the cell.
We observed an increase in the levels of acetylated a-tubulin
Total Cell Lysates and Nuclear and Cytoplasmic Fractionation
(an indicator of microtubule stabilization and cytoskeletal reorga-
For total extracts, cells were lysed in either boiling sample buffer or in RIPA
nization) and an elevation in Myc-nick abundance during muscle buffer. For cellular fractionation, cells were lysed in buffer A on ice for 20 min,
cell differentiation. Induction of a-tubulin acetylation and micro- centrifuged for 3 min, and the supernatant employed as the cytoplasmic frac-
tubule stabilization are likely to be important events in terminal tion. The pellet was resuspended in buffer B, rotated for 20 min, sonicated, and
differentiation especially when cells must establish and maintain centrifuged for 10 min. The supernatant was employed as the nuclear fraction.
a new shape. Acetylation of a-tubulin was also demonstrated to Ten to twenty micrograms of total extracts or 10 mg of nuclear and 30 mg of cyto-
plasmic extracts were probed overnight with indicated antibodies. See
regulate cortical neuron differentiation and migration (Creppe
Extended Experimental Procedures for composition of buffers.
et al., 2009).
We propose a model for the regulation of Myc by calpains and
Immunofluorescence
for the function of Myc-nick. During proliferation, full-length Myc Cells were grown on glass coverslips and fixed with 4% paraformaldehyde
is rapidly synthesized and transported to the nucleus where it for 20 min, permeabilized with 0.5% Triton X-100 for 5 min, and blocked
transcriptionally activates growth- and proliferation-related with Image IT FX signal enhancer (Invitrogen) for 30 min. Primary and
genes and represses genes involved in differentiation. External secondary antibodies were diluted in PBS at 1:200 and incubated for 1 hr.
cues that stimulate differentiation, such as cell-cell contact,
and calcium influx can lead to the activation of calpains. Acti- In Vitro Cleavage of Myc
vated calpains interact with and may retain Myc in the cytoplasm All cDNAs were cloned into pCS2+ and were transcribed from the SP6
promoter using the Promega wheat germ system, in the presence of cold
where it is cleaved to produce Myc-nick. As the cleavage
(Promega) or 35[S]-labeled methionine (Perkin Elmer) according to the manu-
removes the nuclear localization sequence (NLS) and the DNA-
facturer’s instructions. For the in vitro cleavage experiments, Myc (1 ml IVT
and Max-binding domains, Myc-nick is predominantly cyto- Myc or 1 mg recombinant Myc) was incubated with 30 mg of nuclear or cyto-
plasmic and transcriptionally inactive. Indeed we have shown plasmic extracts in 20 ml of buffer G at 37 C and the samples were processed
that Myc-nick, when expressed in cells devoid of full-length for autoradiography or western blot as specified. Nuclear and cytoplasmic
Myc, does not stimulate proliferation, growth, or apoptosis— fractions were dissolved in the same buffer and adjusted to identical salt
the transcription-dependent functions of full-length Myc. In concentration for these experiments. For Myc cleavage using recombinant
calpains, 1 ml IVT c-Myc or 0.25 mg of purified c-Myc was incubated with the
such Myc-nick-expressing cells we instead observe morpholog-
indicated amounts of calpains in the presence of 20 ml buffer G. Total calpain
ical changes and increased tubulin acetylation, which are unique activity was measured using a Calbiochem kit.
to Myc-nick. During muscle differentiation Myc-nick appears to
be stabilized by dephosphorylation of threonine 58 within the In Vitro Tubulin Acetylation Assays
Myc box I phosphodegron. We propose that one of the functions CE (30 mg) or purified assembled microtubules (1 mg) were incubated with 1 ml
carried out by Myc-nick in the cytoplasm involves binding tubulin of IVT c-Myc or IVT vector or with recombinant Myc for 1 hr in the presence of

Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc. 491


acetylation buffer. Assembled microtubules were incubated at 30 C and CE Faiola, F., Liu, X., Lo, S., Pan, S., Zhang, K., Lymar, E., Farina, A., and Martinez,
at 37 C. E. (2005). Dual regulation of c-Myc by p300 via acetylation-dependent control
See Extended Experimental Procedures for buffer composition, constructs, of Myc protein turnover and coactivation of Myc-induced transcription. Mol.
siRNA sequence, pharmacological inhibitors, antibodies, cell lines, and culture Cell. Biol. 25, 10220–10234.
media. Frank, S.R., Parisi, T., Taubert, S., Fernandez, P., Fuchs, M., Chan, H.M.,
Livingston, D.M., and Amati, B. (2003). MYC recruits the TIP60 histone
SUPPLEMENTAL INFORMATION acetyltransferase complex to chromatin. EMBO Rep. 4, 575–580.
Gandarillas, A., and Watt, F.M. (1997). c-Myc promotes differentiation of
Supplemental Information includes Extended Experimental Procedures and human epidermal stem cells. Genes Dev. 11, 2869–2882.
seven figures and can be found with this article online at doi:10.1016/j.cell.
Gebhardt, A., Frye, M., Herold, S., Benitah, S.A., Braun, K., Samans, B., Watt,
2010.06.037.
F.M., Elsasser, H.P., and Eilers, M. (2006). Myc regulates keratinocyte
adhesion and differentiation via complex formation with Miz1. J. Cell Biol.
ACKNOWLEDGMENTS
172, 139–149.

We are grateful to Nao Ikegaki, William Tansey, John Sedivy, and Peter Hurlin Gonen, H., Shkedy, D., Barnoy, S., Kosower, N.S., and Ciechanover, A. (1997).
for reagents essential for this work. We also thank Stephen Tapscott, Maura On the involvement of calpains in the degradation of the tumor suppressor
Parker, Hector Rincon, Bill Carter, Linda Wordeman, and members of the protein p53. FEBS Lett. 406, 17–22.
Eisenman lab for advice and discussion. M.C.-S. was a recipient of an Gundersen, G.G., Khawaja, S., and Bulinski, J.C. (1989). Generation of
EMBO Long-Term Fellowship. This work was supported by NIH/NCI grant a stable, posttranslationally modified microtubule array is an early event in
CA20525 (R.N.E.). myogenic differentiation. J. Cell Biol. 109, 2275–2288.
Habib, T., Park, H., Tsang, M., de Alboran, I.M., Nicks, A., Wilson, L.,
Received: November 9, 2009 Knoepfler, P.S., Andrews, S., Rawlings, D.J., Eisenman, R.N., et al. (2007).
Revised: March 2, 2010 Myc stimulates B lymphocyte differentiation and amplifies calcium signaling.
Accepted: May 18, 2010 J. Cell Biol. 179, 717–731.
Published: August 5, 2010
Hanahan, D., and Weinberg, R.A. (2000). The hallmarks of cancer. Cell 100,
57–70.
REFERENCES
Hann, S.R., King, M.W., Bentley, D.L., Anderson, C.W., and Eisenman, R.N.
Abe, K., and Takeichi, M. (2007). NMDA-receptor activation induces calpain- (1988). A non-AUG translational initiation in c-myc exon 1 generates an
mediated beta-catenin cleavages for triggering gene expression. Neuron 53, N-terminally distinct protein whose synthesis is disrupted in Burkitt’s
387–397. lymphomas. Cell 52, 185–195.
Alexandrova, N., Niklinski, J., Bliskovsky, V., Otterson, G.A., Blake, M., Kaye, Hubbert, C., Guardiola, A., Shao, R., Kawaguchi, Y., Ito, A., Nixon, A., Yoshida,
F.J., and Zajac-Kaye, M. (1995). The N-terminal domain of c-Myc associates M., Wang, X.F., and Yao, T.P. (2002). HDAC6 is a microtubule-associated
with alpha-tubulin and microtubules in vivo and in vitro. Mol. Cell. Biol. 15, deacetylase. Nature 417, 455–458.
5188–5195. Iezzi, S., Di Padova, M., Serra, C., Caretti, G., Simone, C., Maklan, E., Minetti,
Arthur, J.S., Elce, J.S., Hegadorn, C., Williams, K., and Greer, P.A. (2000). G., Zhao, P., Hoffman, E.P., Puri, P.L., et al. (2004). Deacetylase inhibitors
Disruption of the murine calpain small subunit gene, Capn4: calpain is essen- increase muscle cell size by promoting myoblast recruitment and fusion
tial for embryonic development but not for cell growth and division. Mol. Cell. through induction of follistatin. Dev. Cell 6, 673–684.
Biol. 20, 4474–4481.
Ivanov, A.I., McCall, I.C., Babbin, B., Samarin, S.N., Nusrat, A., and Parkos,
Bai, M.K., Costopoulos, J.S., Christoforidou, B.P., and Papadimitriou, C.S. C.A. (2006). Microtubules regulate disassembly of epithelial apical junctions.
(1994). Immunohistochemical detection of the c-myc oncogene product BMC Cell Biol. 7, 12.
in normal, hyperplastic and carcinomatous endometrium. Oncology 51,
Kim, S.Y., Herbst, A., Tworkowski, K.A., Salghetti, S.E., and Tansey, W.P.
314–319.
(2003). Skp2 regulates Myc protein stability and activity. Mol. Cell 11,
Calcagno, D.Q., Guimaraes, A.C., Leal, M.F., Seabra, A.D., Khayat, A.S., 1177–1188.
Pontes, T.B., Assumpcao, P.P., De Arruda Cardoso Smith, M., and Burbano,
Kleine-Kohlbrecher, D., Adhikary, S., and Eilers, M. (2006). Mechanisms of
R.R. (2009). MYC insertions in diffuse-type gastric adenocarcinoma.
transcriptional repression by Myc. Curr. Top. Microbiol. Immunol. 302, 51–62.
Anticancer Res. 29, 2479–2483.
Cartwright, P., McLean, C., Sheppard, A., Rivett, D., Jones, K., and Dalton, S. Koch, H.B., Zhang, R., Verdoodt, B., Bailey, A., Zhang, C.D., Yates, J.R., 3rd,
(2005). LIF/STAT3 controls ES cell self-renewal and pluripotency by a Myc- Menssen, A., and Hermeking, H. (2007). Large-scale identification of c-MYC-
dependent mechanism. Development 132, 885–896. associated proteins using a combined TAP/MudPIT approach. Cell Cycle 6,
205–217.
Clement, C.A., Kristensen, S.G., Mollgard, K., Pazour, G.J., Yoder, B.K.,
Larsen, L.A., and Christensen, S.T. (2009). The primary cilium coordinates Kumar, A., Shafiq, S., Wadgaonkar, R., and Stracher, A. (1992). The effect of
early cardiogenesis and hedgehog signaling in cardiomyocyte differentiation. protease inhibitors, leupeptin and E64d, on differentiation of C2C12 myoblasts
J. Cell Sci. 122, 3070–3082. in tissue culture. Cell. Mol. Biol. 38, 477–483.

Creppe, C., Malinouskaya, L., Volvert, M.L., Gillard, M., Close, P., Malaise, O., Mailhes, J.B., Hilliard, C., Lowery, M., and London, S.N. (2002). MG-132, an
Laguesse, S., Cornez, I., Rahmouni, S., Ormenese, S., et al. (2009). Elongator inhibitor of proteasomes and calpains, induced inhibition of oocyte maturation
controls the migration and differentiation of cortical neurons through acetyla- and aneuploidy in mouse oocytes. Cell Chromosome 1, 2.
tion of alpha-tubulin. Cell 136, 551–564. Matsuyama, A., Shimazu, T., Sumida, Y., Saito, A., Yoshimatsu, Y.,
Dedieu, S., Poussard, S., Mazeres, G., Grise, F., Dargelos, E., Cottin, P., and Seigneurin-Berny, D., Osada, H., Komatsu, Y., Nishino, N., Khochbin, S.,
Brustis, J.J. (2004). Myoblast migration is regulated by calpain through its et al. (2002). In vivo destabilization of dynamic microtubules by HDAC6-medi-
involvement in cell attachment and cytoskeletal organization. Exp. Cell Res. ated deacetylation. EMBO J. 21, 6820–6831.
292, 187–200. McMahon, S.B., Van Buskirk, H.A., Dugan, K.A., Copeland, T.D., and Cole,
Eilers, M., and Eisenman, R.N. (2008). Myc’s broad reach. Genes Dev. 22, M.D. (1998). The novel ATM-related protein TRRAP is an essential cofactor
2755–2766. for the c-Myc and E2F oncoproteins. Cell 94, 363–374.

492 Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc.


McMahon, S.B., Wood, M.A., and Cole, M.D. (2000). The essential cofactor Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K.,
TRRAP recruits the histone acetyltransferase hGCN5 to c-Myc. Mol. Cell. and Yamanaka, S. (2007). Induction of pluripotent stem cells from adult human
Biol. 20, 556–562. fibroblasts by defined factors. Cell 131, 861–872.
Moyen, C., Goudenege, S., Poussard, S., Sassi, A.H., Brustis, J.J., and Cottin, Tompa, P., Buzder-Lantos, P., Tantos, A., Farkas, A., Szilagyi, A., Banoczi, Z.,
P. (2004). Involvement of micro-calpain (CAPN 1) in muscle cell differentiation. Hudecz, F., and Friedrich, P. (2004). On the sequential determinants of calpain
Int. J. Biochem. Cell Biol. 36, 728–743. cleavage. J. Biol. Chem. 279, 20775–20785.
Niklinski, J., Claassen, G., Meyers, C., Gregory, M.A., Allegra, C.J., Kaye, F.J., Tonami, K., Kurihara, Y., Aburatani, H., Uchijima, Y., Asano, T., and Kurihara,
Hann, S.R., and Zajac-Kaye, M. (2000). Disruption of Myc-tubulin interaction H. (2007). Calpain 6 is involved in microtubule stabilization and cytoskeletal
by hyperphosphorylation of c-Myc during mitosis or by constitutive hyper- organization. Mol. Cell. Biol. 27, 2548–2561.
phosphorylation of mutant c-Myc in Burkitt’s lymphoma. Mol. Cell. Biol. 20,
Tworkowski, K.A., Salghetti, S.E., and Tansey, W.P. (2002). Stable and
5276–5284.
unstable pools of Myc protein exist in human cells. Oncogene 21, 8515–8520.
North, B.J., Marshall, B.L., Borra, M.T., Denu, J.M., and Verdin, E. (2003). The
von der Lehr, N., Johansson, S., Wu, S., Bahram, F., Castell, A., Cetinkaya, C.,
human Sir2 ortholog, SIRT2, is an NAD+-dependent tubulin deacetylase. Mol.
Hydbring, P., Weidung, I., Nakayama, K., Nakayama, K.I., et al. (2003). The
Cell 11, 437–444.
F-box protein Skp2 participates in c-Myc proteosomal degradation and acts
Okano, H.J., Park, W.Y., Corradi, J.P., and Darnell, R.B. (1999). The cyto-
as a cofactor for c-Myc-regulated transcription. Mol. Cell 11, 1189–1200.
plasmic Purkinje onconeural antigen cdr2 down-regulates c-Myc function:
implications for neuronal and tumor cell survival. Genes Dev. 13, 2087–2097. Wakamatsu, Y., Watanabe, Y., Shimono, A., and Kondoh, H. (1993). Transition
of localization of the N-Myc protein from nucleus to cytoplasm in differentiating
Pietilainen, T., Lipponen, P., Aaltomaa, S., Eskelinen, M., Kosma, V.M., and
neurons. Neuron 10, 1–9.
Syrjanen, K. (1995). Expression of c-myc proteins in breast cancer as related
to established prognostic factors and survival. Anticancer Res. 15, 959–964. Wakamatsu, Y., Watanabe, Y., Nakamura, H., and Kondoh, H. (1997). Regula-
tion of the neural crest cell fate by N-myc: promotion of ventral migration and
Richard, I., Broux, O., Allamand, V., Fougerousse, F., Chiannilkulchai, N.,
neuronal differentiation. Development 124, 1953–1962.
Bourg, N., Brenguier, L., Devaud, C., Pasturaud, P., Roudaut, C., et al.
(1995). Mutations in the proteolytic enzyme calpain 3 cause limb-girdle Welcker, M., Orian, A., Jin, J., Grim, J.E., Harper, J.W., Eisenman, R.N., and
muscular dystrophy type 2A. Cell 81, 27–40. Clurman, B.E. (2004). The Fbw7 tumor suppressor regulates glycogen syn-
Small, G.W., Chou, T.Y., Dang, C.V., and Orlowski, R.Z. (2002). Evidence for thase kinase 3 phosphorylation-dependent c-Myc protein degradation. Proc.
involvement of calpain in c-Myc proteolysis in vivo. Arch. Biochem. Biophys. Natl. Acad. Sci. USA 101, 9085–9090.
400, 151–161. Wilson, A., Murphy, M.J., Oskarsson, T., Kaloulis, K., Bettess, M.D., Oser,
Spotts, G.D., Patel, S.V., Xiao, Q., and Hann, S.R. (1997). Identification of G.M., Pasche, A.C., Knabenhans, C., Macdonald, H.R., and Trumpp, A.
downstream-initiated c-Myc proteins which are dominant-negative inhibitors (2004). c-Myc controls the balance between hematopoietic stem cell self-
of transactivation by full-length c-Myc proteins. Mol. Cell. Biol. 17, 1459–1468. renewal and differentiation. Genes Dev. 18, 2747–2763.
Sterner, D.E., and Berger, S.L. (2000). Acetylation of histones and transcrip- Xiao, Q., Claassen, G., Shi, J., Adachi, S., Sedivy, J., and Hann, S.R. (1998).
tion-related factors. Microbiol. Mol. Biol. Rev. 64, 435–459. Transactivation-defective c-MycS retains the ability to regulate proliferation
Taira, T., Maeda, J., Onishi, T., Kitaura, H., Yoshida, S., Kato, H., Ikeda, M., and apoptosis. Genes Dev. 12, 3803–3808.
Tamai, K., Iguchi-Ariga, S.M., and Ariga, H. (1998). AMY-1, a novel C-MYC Yousefi, S., Perozzo, R., Schmid, I., Ziemiecki, A., Schaffner, T., Scapozza, L.,
binding protein that stimulates transcription activity of C-MYC. Genes Cells Brunner, T., and Simon, H.U. (2006). Calpain-mediated cleavage of Atg5
3, 549–565. switches autophagy to apoptosis. Nat. Cell Biol. 8, 1124–1132.

Cell 142, 480–493, August 6, 2010 ª2010 Elsevier Inc. 493


Erratum

SIRT1 Suppresses b-Amyloid Production


by Activating the a-Secretase Gene ADAM10
Gizem Donmez, Diana Wang, Dena E. Cohen, and Leonard Guarente*
*Correspondence: leng@mit.edu
DOI 10.1016/j.cell.2010.07.034

(Cell 142, 320–332; July 23, 2010)


As a result of an error during figure preparation, the model in Figure 6D was omitted. Additionally, in Figure S1B, Tau-pSer399 was
incorrectly typed. This has been corrected to Tau-pSer396 both in the figure and in the text. Corrected versions of both figures are
shown below.
These errors in no way affect the conclusions of the paper, and the authors apologize for any confusion caused by these errors.
The online version of the article has been corrected.

Figure 6. SIRT1 Deacetylates RARb

494 Cell 142, 494–495, August 6, 2010 ª2010 Elsevier Inc.


Figure S1. Related to Figures 1 and 2

Cell 142, 494–495, August 6, 2010 ª2010 Elsevier Inc. 495


Scientific Editor Position Available
Cell is seeking a scientist to join its editorial team. The minimum qualification for this position is
a PhD in a relevant area of biomedical research, although additional post-doctoral or editorial
experience is preferred. This is a superb opportunity for a talented individual to play a critical role
in promoting science by helping researchers disseminate their findings to the wider community.

As an editor, you would be responsible for assessing submitted research papers and overseeing
the refereeing process, and you would commission and edit material for Cell’s Leading Edge.
You would also travel frequently to scientific conferences to follow developments in research and
to establish and maintain close ties with the scientific community. You would have opportunities
to pioneer and contribute to new trends in scientific publishing. The key qualities we look for
are breadth of scientific interest and the ability to think critically about a wide range of scientific
issues. The successful candidate will be highly motivated and creative and able to work inde-
pendently as well as in a team.

This is a full-time, in-house editorial position, based at the Cell Press office in Kendall Square
near MIT in Cambridge, Massachusetts. Cell Press offers an attractive salary and benefits pack-
age and a stimulating working environment that encourages innovation.

To apply, please send your CV and a letter describing your qualifications, general research inter-
ests, and motivation for pursuing a career in scientific publishing as soon as possible to our online
jobs site: http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=CEL0000R.

No phone inquiries, please. Cell Press in an equal opportunity/affirmative action employer, m/f/d/v.
The American Journal of Human Genetics
Editor Position Available

The American Society of Human Genetics is seeking an Editor for The American Journal of Human
Genetics. The Editor leads one of the world’s oldest and most prestigious journals publishing pri-
mary human genetics research.

Among the Editor’s responsibilities are determining the scope and direction of the scientific con-
tent of The Journal, overseeing manuscripts submitted for review and their publication, selecting
and supervising a staff consisting of an Editorial Assistant and doctoral-level Deputy Editor, direct-
ing interactions with the publisher (currently Cell Press), reviewing quarterly reports provided by the
publisher, evaluating the performance of the publisher, and if required, supervising the process of
the selection a new publisher. The Editor serves as a member of the Board of Directors of the Ameri-
can Society of Human Genetics (ASHG), as well as the ASHG Finance Committee, and presents
semiannual reports to the Board. All Associate Editors of The Journal are appointed by the Editor,
who also determines their duties. At the ASHG annual meeting, the Editor presides over a meeting
of the Associate Editors and presents an annual report to the ASHG membership.

The term of the appointment is five years and includes a yearly stipend. The new Editor will be
selected by the end of 2010 and will begin receiving manuscripts approximately in September 2011;
there will be partial overlap with the Boston office. Applicants should be accomplished scientists
in the field of human genetics and should have a broad knowledge and appreciation of the field.
Nominations, as well as applications consisting of a letter of interest and curriculum vitae, should be
sent to:

AJHG Editorial Search Committee


American Society of Human Genetics
9650 Rockville Pike
Bethesda, MD 20814
Positions Available

The University of Vermont College of Medicine is seeking applications from, or nominations


of, outstanding candidates for the Chair of Biochemistry. This position is part of a strategic
initiative in the College to build on areas of strength and increase interactions among the
basic and clinical sciences. The College is particularly interested in candidates with substan-
tive credentials in molecular cancer biology especially in areas related to environmental
carcinogenesis. It is expected that candidates would have credentials appropriate for a
tenured position. The Chair will be responsible for the development and oversight of the
educational and research programs of the Department and have the opportunity and
resources to implement a visionary approach to the discipline while promoting translational
research. Current strengths of the Department are in Solution and Structural Studies of
Macromolecules, and the Cell and Molecular Biology of cancer and of thrombosis.

The successful candidate will be an internationally recognized scientist with a Ph.D. or


equivalent degree who has demonstrable skills as a leader, and substantial experience in the
administrative, teaching and research activities of an academic department. The College
seeks an innovative Chair who will position the Department as a national academic and
research leader.

The University of Vermont is located in Burlington, a vibrant community with year-round


recreational opportunities, safe communities and excellent schools. This progressive
community has been frequently cited as one of the most livable cities in the U.S.

Women and those from diverse racial, ethnic and cultural backgrounds are encouraged to
apply. Applications will be accepted until the position is filled. A curriculum vitae and letter
of interest should be sent electronically to Linda Thatcher at the address below.

Susan S. Wallace, Ph.D.


Chair, Biochemistry Search Committee
c/o Linda Thatcher (Linda.thatcher@UVM.edu)
E-126 Given Building
University of Vermont College of Medicine
Burlington, VT 05405-0068
Or apply online at http://www.uvmjobs.com

The University of Vermont is an Equal Opportunity/Affirmative Action Employer.


Positions Available

Faculty Position
Department of Molecular Biology
Princeton University
The Molecular Biology Department at Princeton University invites appli-
cations for a tenure-track faculty position at the Assistant Professor level
in Developmental Biology. We are seeking an outstanding investigator
using multicellular model systems to address fundamental questions in
cell signaling, gene regulation, pattern formation, morphogenesis and/
or pathogenesis. The University has a strong commitment to interdis-
ciplinary studies, especially in the areas of systems biology, imaging,
genomics, biophysics and neuroscience. The department has high level
computing and microscope facilities, DNA array and high through-put
sequencing technologies, mass spectrometry, modern transgenic mouse
facilities and state of the art vivarium.
Applicants must have an excellent record of research productivity and
demonstrate the ability to develop a rigorous research program. All
applicants must have a Ph.D. or equivalent degree and a commitment
to teaching at the undergraduate and graduate levels.
Applications must be submitted online at http://jobs.princeton.edu,
requisition # 1000527, and should include a cover letter, curriculum
vitae, a two-page research description, and contact information for
three references. All materials must be submitted as PDF files. For full
consideration, applications should be received by November 1, 2010.
Princeton University is an equal opportunity employer and complies
with applicable EEO and affirmative action regulations.

Bacteriology Search:
Tenure Track or Tenured Faculty
Department of Microbiology
University of Washington
The Department of Microbiology at the University of Washington in Seattle is conducting a search for an Assistant or Associate
Professor in the fields of bacterial-host interactions and/or bacterial physiology. We are looking for an innovative investigator
who currently has or will develop an independent research program studying bacterial pathogenesis, bacterial interactions with
the immune system, mutually beneficial relationships between microbes and hosts, or bacterial physiology relevant to a variety of
microbial systems. The position is a 12-month, full-time, tenure track position in the School of Medicine. In addition to research,
the new faculty member will support the department’s teaching mission, including teaching at the undergraduate or graduate
level. All University of Washington Faculty engage in teaching, research and service.
Salary and benefits are competitive and will be commensurate with the qualifications and experience of the applicant. The Uni-
versity of Washington is an affirmative action, equal opportunity employer and has built a culturally diverse faculty. Applications
from female and minority candidates are especially encouraged. Expanding opportunities for women and other underrepresented
groups in the faculty is an important goal of the department.
Applicants with a Ph.D., 1-2 years postdoctoral experience and a strong publication record should send their CV, a one or two
page statement of research interests and the names and contact information for three references to:
Chair, Bacteriology Search Committee
Department of Microbiology
Box 357242, Room K357B, University of Washington
1705 N.E. Pacific Street, Seattle WA 98195
Application materials may also be sent by email c/o Bonnie Hightower at bbh@uw.edu. Applications should be submitted by
September 15, 2010, but search will remain open until the position is filled.
Positions Available

FACULTY POSITION IN AGING AND


AGE-RELATED DISEASE
Assistant Professor Level
The Buck Institute invites applications for the Faculty position at the

60
Assistant Professor level. The Buck Institute is the first independent
research facility in the country focused solely on aging and age-related
disease. The candidate cannot have held a previous faculty position.
The successful candidate is expected to develop and maintain an extra-
murally funded research program in basic mechanisms of aging or
age-related disease. Applicants with expertise in molecular, cellular,
animal models, imaging, and/or computational experimental approaches
related to understanding the mechanisms of aging or disease in human
health are encouraged to apply. Areas of interest include but are not
limited to inflammation or metabolism. Candidates must have a Ph.D., years of leadership in
M.D., or equivalent degree and at least two years of relevant postdoc-
toral research experience. The position will be supported with five human genetics research,
years’ salary support, startup funds and excellent laboratory space in a
first class facility.
education and service.
Please electronically submit curriculum vitae, a brief statement of
current and future research interests, and contact information for three
1948–2008
references to facultyrecruitmentir@buckinstitute.org. Applicants are
encouraged to apply immediately for full consideration. The position www.ashg.org
could start immediately. This recruitment is through the NIH Recovery
and Reinvestment Act.

Look Again. Discover More.


• Access to the 14 Cell Press
primary research journals and
14 Trends reviews title, all on
the same platform
• Improved, more robust article and
author search

• Easy to navigate home page,


articles pages and archive

www.cell.com
.%7

Find Your Ideal Job!

så3EARCHåJOBSåBYåKEYWORD å
LOCATIONååTYPEå

så0OSTåYOURåRESUMEåå
ANONYMOUSLYå

så#REATEåAå*OBå!LERTåå
ANDåLETåYOURåIDEALåå
JOBålNDåYOU

careers.cell.com
,OOKING¬FOR¬WHAT¬#ELL¬
HAS¬PUBLISHED¬IN¬¬
)MMUNOLOGY¬LATELY


PHOIDå#ELLS å å åPPån

ONåINå,YM Då-OTIF
Så!CTIVATI L å  å*AN OSINE "ASE
CTERIZEå2A SE å* 0 å# EL IC å4 YR
Så#HARA OO LA SM
å(YSTERESI KRABORTYå!
+ å2 Nå#YTOP n
NALINGåAND EISSå! å#HA å#$EPSILO  åPPå
$IGITALå3IG OV ER Nå # å9ANGå- å7 N E å" IN D INGåOFåTHE L å   å. OVå å
ANå* å' ICå-EMB RA å# EL
- å:IKHERM åBYå$YNAM RPFENNIGå+7 LOPMENT
$ASå* å(Oå !CTIVATION ** å7UCHE Cå#ELLå$EVE HUANGå9 å2
EIZISå
Rå å#6 å#HOUå RI TI
Få4 C E LLå2 E C E P TO
IE TE RS å# $ å# AR M AN
A C YT O ID å$ E N D
OL TE Rå7 å2AUCHå! å:
2EGULATIO
NåO LLå*2 å3CH W RåOFå0LASM 3 å+ANTå3'
å(
NONå% å#AL
Lå-% å3CHNE Cå2EGULATO å(OLLANDERå.
8Uå# å'AG E N TI A LåA NDå3PECIl RG å$ å: WEIERå# åDEN
åISåANå%SS LEYå2 å(OLM
BE ISEASE
RI P TI O N å& ACTORå%  å4 å3 CH EUå3 å,OCKS A M M A TO RYå"OWELå$
4RANSC AE DA UMANå)N m
EHNERå- å- å2ISKåFORå( BERGå23åå
ATONå-, å, 
RSå'ENETIC ERå,( å"LUM
#ISSEå" å# å å   åPPån N D å# O N FE å3 CH RE IB ER å3 å'LIMCH
"å#ELL å
å/CT ATIONåA å(IGGINSå$
%
ALå)NmAMM ENHUISå%%
2 å3 TR E SS åTOå)NTESTIN SS IG å3 å4 ILGå( å.IEUW
Så% KMANå*. å:
EI
8"0å,INK NKEå! å'LIC
EEå!( å&RA 
+ASERå! å, åPPån
EPå å MUNITY n
#EL L å   å3 åOFå!UTOIM  åPPå EOSTASIS
)N TR IN SI C å)NITIATION   å! UGå å MUNEå(OM
VE N TS å# E LL Vå 2 å# EL L å
IN TA IN Så )M
4REXå0RE EIDMANNå4
å-EDZH ITO THATå-A å#HENå9(åå
å+Oå*3 å( å)MMUNITYå ( å9ANGå8
3TETSONå$" N A TE åA N Då!DAPTIVE å(ILLIARDå" å(Uå3 å3HENå
Få)N Gå, å8Uå, RACTIONS
EGULATORåO å3UNå* å+ON OMICå)NTE
EGATIVEå2 ILLIARDå! å,Iå, ANGEå'EN
4)0% åAå. OD Yå 2 * å( Rå , O N G 2
Gå3 å#ARM TIONSåFO REå#åå
3UNå( å'ON åPPån
 Så)MPLICA CHå4! å-UR
  å- AYå å å( E A VY # HAINå,OCU å' RO SV ELDå&' å+NO
L å GLOBULIN **
#EL
Eå)MMUNO ANå$ONGENå
å2IBLETå2 åV JURY
U CTUREåOFåTH å#UTCHINå3 TEå,UNGå)N
4H E å $ å3 TR
å: EL M å- # å0 EA Kå - -
E Yå 0 A TH W AYåOFå!CU Gå ( å, IUå( å3UNå9
åå
ALAå3 åVAN n GåASåAå+ NGå9( å7AN NINGERå*-åå
*HUNJHUNW å PP å  å3 IG N A LIN ZE Nå 2 å, EU Rå# * å0 EN
å!PRå å
 ECEPTORå VAå- å6ELDH
UI
IANGå# å"IN
DE
#ELL å å4OLL ,IKEå2 å' å%RMOLAE ICHOLLSå* å*
D A TI VE å3 TRESSåAND ER KM AN Nå 4 åVANå,OO ST å- å( OL MDAHLå2 å.
TIONåOFå/XI Iå2 å0 VI
AN -ALHAM KIRAå3 å(ULTQ
)DENTIlCA ' å9AGHUBI EIRISå*3 å3LU
TSKYå!3 å!
å9 å+ UB Aå + å.EELYå' AV AR Iå3 å0
)MAI å-ECHå# å"
- å+OPFå-
0ASPARAKISå å   åPPå
n
 å! PR å 
#EL L å 

,OOKå!GAIN
$ISCOVERå-ORE
WWWCELLCOM
4HEå"IOPHYSICALå3OCIETYå
ANDå#ELLå0RESS
!åNEWåPARTNERSHIP

3EEåWHATSåNEWåAT
WWWBIOPHYSJORG
SnapShot: Nonhomologous DNA
End Joining (NHEJ)
Michael R. Lieber1 and Thomas E. Wilson2
1
University of Southern California, Los Angeles, CA 90089, USA and 2University of Michigan, Ann Arbor, MI 48109, USA

496 Cell 142, August 6, 2010 ©2010 Elsevier Inc. DOI 10.1016/j.cell.2010.07.035 See online version for legend and references.
G1 (SPEG)
1 (ARK RK)
2 (SNA

STK11

EG_D2)

FES (FPS

J
MATK (H
MYLK

3
JA
MERT
TSSK4

DAPK3

NTR
A XL
FER (T Y

NTRK
DAPK1

B1)
OBSCN_D

2)
A)
CSK
5)

ERBB (Erb

EPH A4
(LKB1)

2 (HER
STK17B A (DRAK

(SMMLC

OBSCN

RET

1 (TRK

EPH
STK17

A5
(LMR2 )
PIM2

LMTK2 (LMR1
)

EGFR
MET (Dtk)
ALK
2

(RON)
MST1R(cMET)

NTRK
IGF1R
LTK
(DRAK2 1)

K)

INSR

AATK

A3
TYRO3

EPH
K106)
DAPK2
)

4)
TEK (TIE2)

4 (HER
ROS1 (ROS)
CHEK

RYK

DDR1
(SURT

DDR2
(IRR)
PASK

K
)
TIE1

MUS
MGC

3 (LMR3
1 (CHK1

ERBB
TRAD
TRIO

STYK1
INSRR

B1
4210
B2

EPH
JAK1
TNK1 (ACK)

LMTK
5 (NIM

TYK2
)
CAM EPH

JAK 2
TTN

A6
3)

JAK
MEL K

3
CAM

EPH
TNK2

3 (HER
K1

1)
SNR
K
K1D (CA
(CA MKI

ERBB
MKI alph CAM CAM B3

ROR1
2
CAM delt a) EPH

ROR
CAM K2G K2B

A7
K1G a)

EPH
(CA CAM K2D (CA (CA
MKI PNC K2A (CA MK II MK II
gam K (CAM (CA MKI gam bet B4

)
(CCK4
ma) KI MKI I del ma) a) EPH
beta I alph ta)

PTK7

70
SYK
PHK
)

ZAP
a)

TRIB
G2 G1 (PH
PHK

(PH K

TRIB
2 (TRB(TRB
K gam gam

A2
1

(PYK )
2)
2)

PTK 2 (FAK

EPH
ma ma 1)

TRIB
1)
2)

2B
PTK
3 (TRB

HUN33
STK
A8

A1
K
EPH

3)

EPH
CAM
CAM K4
KV

STK
RPS6 (VAC

40
RPS6 KA1_ CAS

(SG
KA3_ D2 AMK K

K49
L) R2)
D2 (RSK (ACT R2B)

5)
TK
(RSK 1_D2 PSKH R2 (ACT
2_D2 ) DCA PSKH 1 ACV R2B
RPS6 ) DCA MKL 2 A10 ACV
KA2_ MKL 1 EPH B6 R2)
D2 RPS6 2 EPH beta 4) R1)
(RSK (ALK beta
3_D2 RPS6 KA4_ (TGF R2)
R1B (TGF
RPS6 ) KA5_ D2 DCA BR2 (MIS ACV R1
KA6_ D2 (MSK MKL
TGF R2 1) R1A TGFB
D2 (MSK 2_D2 AMH R2 (ALK 2) BMP R1B
(RSK4 1_D2 ) 3 BMP RL1 (ALK BMP 7)
_D2) ) ACV R1 (ALK
ACV 1C
ACVR

K
MKN
MKN K1 1
(MNK
K2 (MNK IRAK 3
1) IRAK

AM
2)
MAP MAP
MAPK KAPK KAPK
APK2 3 5 (PRA
K) IRAK2
HIPK2 D3)
HIPK1 PRKD 4 (ANKR 88)
IRAK )

C
PRKD 1 (PKCM ILK RIPK1 RIPK4 (SGK2
HIPK3 3 (EPK2 ) RIPK3 (RICK ANKK1
(DYRK6 CHEK RIPK2
) )
DYRK3 2 (CHK
PRKD MLKL
DYRK2 2 (PKD2 2)
HIPK4 )
DYRK4 LRRK2
LRRK1

Webinar Announcement
DYRK1A BRAF (cRAF)
DYRK1B ARAF RAF1
CLK1

TK
CLK4 KSR (KSR1)
CLK3 KSR2
CLK2
SRPK1
SRPK2

L
STK23
(MSSK1)
ZAK
(TAK1)
MAP3K7
GSK3B
(GSK3 PRPF4B
GSK3A (GSK3 beta) (PRP4) (HH498)
alpha) TNNI3K
MAK
LIMK2
ICK LIMK1
RAGE (MOK) TESK1
TESK2
CDKL4
CDKL1
CDKL2
CDKL3
MAPK14 (p38 alpha)
CDKL5
MAPK11 (p38 beta)
MAPK13 (p38 delta)
MAPK12 (p38 gamma)
MAPK8 (JNK1)
MAPK10 (JNK3)

CMGC
MAPK9 (JNK2)
MAPK1 (ERK2)
MAPK3 (ERK1)
MAPK7 (ERK5)
NLK

STE
ERK8 (ERK7)
MAPK4 (ERK4)
MAPK6 (ERK3)

CDK4
CDK6

CDK3
CDK2
CDC2 (CDK1) CDK5 TAOK2
TAOK3 (JIK) TAOK1 (

PCTK2 (PCTAIRE2)
PCTK1 (PCTAIRE1)PCTK3 (PCTAIRE3) STK24 (M
STK25 (YSK1) MASK (MS
PFTK1 (PFTAIRE1) STK3 (MST2)
(PFTAIRE2) CDK8 STK4 (MST1)
ALS2CR7
CDK7
(CDK11)
CDC2L6
NRK (ZC4) MINK1
TNIK MAP4K4(ZC3/M

AT Y
(PITSLRE)CDC2L2 CDK10 CDK9
CDC2L1 MYO3A (ZC1/
MYO3B
CCRK
(CHED) CRK7
CDC2L5
MAP4K MAP4K3
MAP4K 2 (GCK) MAP4K (KHS2)
STK10 1 (HPK1) 5 (KHS1)
SLK (LOK)

PIC
SGM1

AL
RIOK2
(mTOR) ATR
TRRAP FRAP1 ATM EEF2K
K)
(DNA-P RIOK1
PRKDC RIOK3
)
4 (TIF1A )
TRIM2 3 (TIF1G )
TRIM3 8 (TIF1B
TRIM2
AK3)
1 (ALPH (PTK9)

Webinar series: Applications


ALPK L)
TWF1(PTK9 PIK3R

R
TWF2 TAF1 SCYL 4
) LOC4 2

HE
1_D2 TEX1 0068
L (TAF BRDT PINK 4 (SGK 7 (SGK
TAF1 BRD4 1 307)

AG OT
EIF2 EIF2A
BCR AK1
(HRI K4 DKFZ
(GCN P

C
)
) 2) KIAA200
9 (G11 EIF2A
3 STK1 EIF2 K2
BRD 2 K1) AK3 (PKR
NRB
7 (CHA K2)

CK1
BRD (PEK
) P (NR
TRPM M6 (CHA (H11 FASTK BP1
)
TRP B8 )
HSP WN
K4
K5
BUB

1 (RSK L2)
ADC

L1)
CDC 1

6KC (RSK
WN

SBK
K2 GSG 7

1 (SBK
RPS 6KL1
2 (HA FLJ2

K1
ADC

RPS
WN 3356

)
SPIN
WN K3 ) (SGK
K1

HK1 )
196)

HK3
1)

LOC 390 (IKK bet


1)
PDK 3 (PD

LOC K (IKK
HAK 2)

1 (PDK

649 975 alph a)


1 (PD

CHU KB
(ALP HAK P

PDK

023 (LO a)
IKB 1 (IKK
K3 SCY

PDPK
ALP K2 (ALP

(SG C64
TBK E
L

IKBK

K11 664
K2

MOS
ALP

0) 3)
ADC

PRKY X G1 (PKG 2)
1)

epsi
PRK PRK (PKG

PKM 1 (WEE1B)
STK16 GAK

WEE 2

lon)
YT1
G2

WEE
PRK

(MY
(MPSK1)

RIPK (TOP
CSNK1G3 (CK1

TTK

ERN 2 (IRE
T1)
PBK
)
HK2

ERN
5 (SGK K)
STK35 (CLIK1)
CSNK1A1 (CK1 alpha) 2)
CSNK1D (CK1 delta)
CSNK1E (CK1 epsilon)
CSNK1A1L (CK1 alpha

1 (IRE
PDIK1L (SGK071)
C9ORF96(KIS)
2 (PD

UHMK1
) K4

CAM K1
CSNK1G1 (CK1 gamma 2)
CSNK1G2

1)
CK3 ADC

496)
CAMK
PDK

VRK3
STK32A (YANK1) STK32C (YANK3) MASTL

TTBK1

2)
TTBK2
(AD

(CLIK1L)

KK2
CI

FLJ3268(FUSED)
ma)

STK36
gamma 3)
CAB

MAST3

ULK4
gam

(CK1 gamma

AAK1 (BIKE)
(SGK494)

BMP2K

5 (NEK10)

ULK3
alph ) (PKA

AUR
(PKA beta CG

CSNK 2A2
VRK1

KA
VRK2

CSNK
ACA (PKA PRKA

FLJ25006

TLK2
LATS2
LATS1
DK

(AUR
TLK1
)

PKN3

PLK4
2A1 (CK2
HK4

1)
a)

MAST1
BCK

ULK1
CIT (CRIK)

AUR KB (AUR
4 (PD

ULK2

A)
(CK2 alpha
STK32B (YANK2)

AUR
KC
PDK

PRK PRKACB

(AUR B)
alpha 2)

PLK1

C)
NEK6
NEK7

1)
PKN2

ADRBK1 (GRK3, BARK2)


(GPRK7)(GRK2, BARK1)
PKN1

MAST2
MAST4

NEK2
A6 (RSK4)
SGK2

PLK3 (SNK
PLK2
I (PKC zeta)

NEK9
iota)

NEK8

(FNK )
GRK1 (RHOK)
RPS6K

ADRBK2

GRK6 (GPRK6)
PRKC (PKC

a)

)
gamm
Z

SGK3
PRKC

AKT3 SGK

2 (P70S6 1 (p70S6(RSk1)

GRK7

STK38 (NDR1)
AKT1 (PKB beta) (PKB

STK38L (NDR2)

NEK11
K beta) K)
RPS6K A5 (MSK1)
A4 (MSK2)
A1
delta )
PRKC E (PKC (PKC eta) D (PKC theta

RPS6K
)

alpha)

GRK5 (GPRK5)
GRK4 (GPRK4)
of a kinase binding assay

ROCK1
RPS6KB RPS6KB

RPS6K

ROCK2
RPS6K A2 (RSK3)
(PKC

A3 (RSK2)
(PKB
PRKC Q

NEK4
DMPK (DMPK1)
PRKC

AKT2

RPS6K
gamm on)

NEK3
a)
epsil
H
PRKC PRKC

beta) G (PKC

CDC42BPG (DMPK2)

NEK1
NEK5
)
alpha
PRKC B (PKC
A (PKC
PRKC

CDC42BPB (MRCK beta)


CDC42BPA (MRCK alpha)
Solutions for advancing your workflow from challenging
targets to downstream applications
Join us for this free two-part webinar series designed to highlight the versatility of the LanthaScreen® Eu Kinase
Binding Assay to help solve your assay development challenges and provide a simplified solution for multiple
downstream applications.

Webinar Part I: Assay development for broad kinome coverage, including difficult targets
This introductory webinar will help you:

→ Understand the basic principles of assay design and where the LanthaScreen® Eu Kinase Binding Assay may fit into your workflow

→ Discover how this assay platform can alleviate the assay development challenges of difficult kinase targets

→ Learn how easy this assay is to use for kinases with low purity, non-activated kinases, or kinases without a known substrate

Webinar Part II: Advanced applications of the LanthaScreen® Eu Kinase Binding Assay Platform
Learn how the LanthaScreen® Eu Kinase Binding Assay:

→ Can augment the characterization of compounds by simplifying on- and off-rate experiments

→ Can detect multiple classes of kinase inhibitors

→ Can be utilized for cell lysate–based applications

Each webinar series is being offered in North American and European time zones.

View the dates and times of each webinar and register to attend at www.invitrogen.com/kbawebinar.

www.invitrogen.com
For research use only. Not intended for any animal or human therapeutic or diagnostic use, unless otherwise stated.
© 2010 Life Technologies Corporation. All rights reserved. The trademarks mentioned herein are the property of Life Technologies Corporation or their respective owners. These products may be covered by one or more Limited Use Label
Licenses (see Invitrogen catalog or www.invitrogen.com). By use of these products you accept the terms and conditions of all applicable Limited Use Label Licenses. CO21534 0710
SAN TA CR UZ B I OT ECH N O LO GY, I N C . SANTA CRUZ
®

Leading (#1 worldwide) supplier BIOTECHNOLOGY

of specialty biochemicals for


The Power to Question

proteomics research.

Products include over 71,300 unique biochemicals


sorted alphabetically, by CAS number, by formula
and on the basis of functional activity.

See our web site for product details


and ordering information.

www.scbt.com

HEADQUARTERS
TOLL FREE: 800.457.3801 TEL: 831.457.3800
FAX: 831.457.3801 E-MAIL: scbt@scbt.com

EUROPEAN SUPPORT
TOLL FREE: +00800.4573.8000 PHONE: +49.6221.4503.0
FAX: +49.6221.4503.45 E-MAIL: europe@scbt.com

ASIAN SUPPORT
JAPAN TOLL FREE: (010) 800.40402026 S. KOREA TOLL FREE: 00798.1.1.002.0297
N. CHINA TOLL FREE: 10.800.711.0752 S. CHINA TOLL FREE: 10.800.110.0694
FAX: 831.457.3801 E-MAIL: asia@scbt.com

ChemCruz ™

Vous aimerez peut-être aussi