Vous êtes sur la page 1sur 286

RHYPR 2/6/04 3:50 PM Page i

MOLECULAR
BIOLOGY OF
CIRCADIAN RHYTHMS
RHYPR 2/6/04 3:50 PM Page iii

MOLECULAR
BIOLOGY OF
CIRCADIAN RHYTHMS

Edited by
AMITA SEHGAL
Howard Hughes Medical Institute
Department of Neuroscience
University of Pennsylvania School of Medicine
Philadelphia, Pennsylvania

A John Wiley & Sons, Inc., Publication


RHYPR 2/6/04 3:50 PM Page iv

Copyright © 2004 by John Wiley & Sons, Inc. All rights reserved.

Published by John Wiley & Sons, Inc., Hoboken, New Jersey.


Published simultaneously in Canada.

No part of this publication may be reproduced, stored in a retrieval system, or transmitted


in any form or by any means, electronic, mechanical, photocopying, recording, scanning, or
otherwise, except as permitted under Section 107 or 108 of the 1976 United States Copy-
right Act, without either the prior written permission of the Publisher, or authorization
through payment of the appropriate per-copy fee to the Copyright Clearance Center, Inc., 222
Rosewood Drive, Danvers, MA 01923, 978-750-8400, fax 978-646-8600, or on the web at
www.copyright.com. Requests to the Publisher for permission should be addressed to the
Permissions Department, John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030,
(201) 748-6011, fax (201) 748-6008.

Limit of Liability/Disclaimer of Warranty: While the publisher and author have used their best
efforts in preparing this book, they make no representations or warranties with respect to the
accuracy or completeness of the contents of this book and specifically disclaim any implied
warranties of merchantability or fitness for a particular purpose. No warranty may be created
or extended by sales representatives or written sales materials. The advice and strategies con-
tained herein may not be suitable for your situation. You should consult with a professional
where appropriate. Neither the publisher nor author shall be liable for any loss of profit or
any other commercial damages, including but not limited to special, incidental, consequential,
or other damages.

For general information on our other products and services please contact our Customer
Care Department within the U.S. at 877-762-2974, outside the U.S. at 317-572-3993 or fax
317-572-4002.

Wiley also publishes its books in a variety of electronic formats. Some content that appears
in print, however, may not be available in electronic format.

Library of Congress Cataloging-in-Publication Data:


Molecular biology of circadian rhythms / edited by Amita Sehgal.
p. ; cm.
Includes bibliographical references and index.
ISBN 0-471-41824-2 (cloth : alk. paper)
1. Circadian rhythms. 2. Molecular biology.
[DNLM: 1. Circadian Rhythm—genetics. 2. Circadian
Rhythm—physiology. 3. Molecular Biology. QH 527 M7175 2003] I.
Sehgal, Amita.
QP84.6.M65 2003
571.7¢7—dc21 2003000610

Printed in the United States of America

10 9 8 7 6 5 4 3 2 1
RHYPR 2/6/04 3:50 PM Page v

CONTRIBUTORS

John D. Alvarez, M.D., Ph.D., Department Peter McNamara, Ph.D., Phenomix Corpo-
of Pathology, University of Pennsylvania ration, La Jolla, California
School of Medicine, Philadelphia,
Pennsylvania Nirinjini Naidoo, Ph.D., Center for Sleep
and Respiratory Neurobiology, Depart-
Juan Capel, Ph.D., Departmento de Biolo- ment of Medicine, University of Pennsylva-
gia Aplicada, Universidad de Almeria, nia School of Medicine, Philadelphia,
Almeria, Spain Pennsylvania
Anthony R. Cashmore, Ph.D., Plant Jeffrey L. Price, Ph.D., School of Biological
Science Institute, Department of Biology, Sciences, University of Missouri-Kansas
University of Pennsylvania, Philadelphia, City, Kansas City, Missouri
Pennsylvania
Amita Sehgal, Ph.D., Department of Neu-
David F. Dinges, Ph.D., Division of Sleep roscience, Howard Hughes Medical Insti-
and Chronobiology, Department of Psychi- tute, University of Pennsylvania School of
atry, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
Medicine, Philadelphia, Pennsylvania
Hans P.A. Van Dongen, Ph.D., Department
Jadwiga M. Giebultowicz, Ph.D., Depart-
of Psychiatry, University of Pennsylvania
ment of Biology, Oregon State University,
School of Medicine, Philadelphia,
Corvallis, Oregon
Pennsylvania
Jose A. Jarillo, Ph.D., Departmento de
Julie A. Williams, Ph.D., Department of
Biotechnologia, Instituto Nacional de
Neuroscience, Howard Hughes Medical
Investigion y Technologia Agraria y Ali-
Institute, University of Pennsylvania
mentaria (INIA), Madrid, Spain
School of Medicine, Philadelphia,
Gerard A. Kerkhof, Ph.D., Department Pennsylvania
of Psychology,University of Amsterdam,Ro-
etersstraat 15, Amsterdam, The Netherlands

v
RHYPR 2/6/04 3:50 PM Page vii

CONTENTS

Preface ix
Acronyms xi

䊏 PART I
INTRODUCTION TO CIRCADIAN RHYTHMS
1 General Concepts 3
Amita Sehgal

2 Genetic and Molecular Approaches Used to


Analyze Rhythms 17
Amita Sehgal and Jeffrey L. Price

䊏 PART II
MOLECULAR CONTROL OF CIRCADIAN RHYTHMS:
ANIMAL MODELS
3 Drosophila melanogaster: A Model System for
Molecular Chronobiology 33
Jeffrey L. Price

4 Molecular Analysis of Circadian Rhythms: Nonmammalian


Vertebrates 75
Julie A. Williams

5 Genetic Basis for Circadian Rhythms in Mammals 93


John D. Alvarez

䊏 PART III
MOLECULAR CONTROL OF CIRCADIAN RHYTHMS:
FROM CYANOBACTERIA TO PLANTS
6 Circadian Rhythms in Cyanobacteria 143
Nirnjini Naidoo

vii
RHYPR 2/6/04 3:50 PM Page viii

viii CONTENTS

7 Molecular Analysis of Circadian Rhythms in Neurospora 171


Amita Sehgal

8 Physiological and Molecular Characteristics of Plant


Circadian Clocks 185
Jose A. Jarillo, Juan Capel, and Anthony R. Cashmore

䊏 PART IV
CIRCADIAN ORGANIZATION IN COMPLEX ORGANISMS
9 Multiple Oscillators 213
Jadwiga M. Giebultowicz

10 Hormonal Rhythms 231


Peter McNamara

11 Human Circadian Rhythms 255


Hans P. A. Van Dongen, Gerard A. Kerkhof, and David F. Dinges

Index 271
RHYPR 2/6/04 3:50 PM Page ix

PREFACE

It appears to finally be “time”—time to and with the effort they had made to read
compile what we know about the mole- the primary literature. Another reason I
cular basis of circadian rhythms. In the thought a book like this might be useful
mid-1990s, this would have seemed a pre- is because I recently collaborated with
posterous idea; the fact that it is now being several groups whose work lead them into
done is a testament to the progress made in the area of circadian rhythms and who
the field in recent times. Up until 1994 there would have appreciated the introduction to
were only two known so-called “clock molecular circadian biology that I hope this
genes,” one in Drosophila and the other in book will provide. Finally, it appears that
Neurospora. Since then, not only have many graduate and even undergraduate
many new components been identified in institutions are now offering classes in
these two systems, but molecular circadian chronobiology, either as part of a broader
models have been rapidly developed for course or even as a course by itself. Now
other systems such as cyanobacteria and that the molecular biology of the system is
mammals. In addition, there is increasing such an integral part of circadian studies, a
awareness of the overwhelming presence of book on this topic may be beneficial. While
circadian control in normal physiology. the writing of this book was under way, I
Clearly, temporal aspects of physiology was also approached by another publisher,
must be considered for all biological pro- seeking to put a similar book together.
cesses. As a result, the circadian field has Clearly, many people now perceive the
also grown in size, populated now not only need for a textbook explaining the intri-
by diehard chronobiologists, but also by cacies of circadian function. I anticipate
many researchers who discovered that their that this book will be useful to all the
favorite molecule or process is regulated in scientists mentioned above, in particular to
a circadian fashion. undergraduate and graduate students.
My own motivation to put this book Having decided to compile this book, it
together came, at least in part, from the took me only a couple of minutes to decide
number of circadian biology queries I that it should reflect the truly interdiscipli-
started receiving from students (graduate, nary nature of this field. Molecular ap-
undergraduate, and, in a couple of cases, proaches to circadian rhythms have been
even high school students). These students used in several, very diverse species, and the
were basing their preliminary proposals or field as a whole has benefited tremendously
dissertation topics or research projects on from research done in each of these. In
the analysis of some molecular aspect of addition, the overall approaches used in all
circadian rhythms. I must add that I was these species are very similar. Thus, the
suitably impressed with the depth of the book starts with an introduction outlining
questions that these students were asking the general properties of circadian rhythms

ix
RHYPR 2/6/04 3:50 PM Page x

x PREFACE

and the description of commonly used and would like to take this opportunity to
terms (jargon) and then introduces the thank each one of them. They all cheerfully
methodology used to study the molecular honored their commitments and endured
biology of clocks. It then provides an up-to- endless messages and questions from me.
date (as much as is possible in this fast Special thanks goes to Jay Dunlap, Jennifer
moving field) account of the research done Loros, Susan Golden, and Carla Green for
in six organisms and systems commonly providing expert opinions on the Neu-
used for molecular circadian biology. The rospora and cyanobacteria chapters.
last section is devoted to the organization My secretary, Irene Stevens, scanned
of circadian systems in complex organisms many of the figures and kept track of copy-
and the overall impact on physiology. The right permits, for all of which I thank her. I
emphasis in this section is on mammals, would also like to thank members of my
although much of the discussion is also laboratory for their patience when my time
applicable to other organisms. was so heavily occupied by this book. Last,
All the chapters have been written by but definitely not least, I am indebted to my
people with expertise in the molecular family, my husband Jeff and my daughters
analysis of circadian rhythms. Where possi- Natania and Anjalie, for their endless
ble, they have been written by researchers support. They have kept me sane through-
working on the specific organism they are out my research career, including this most
writing about. In other cases, individuals recent period when I was writing and com-
working with other species have gone to piling this book.
tremendous effort to thoroughly research
the system they were assigned and provide Amita Sehgal
a comprehensive account of it. I am Philadelphia, Pennsylvania
extremely grateful to all the contributors
RHYPR 2/6/04 3:50 PM Page xi

ACRONYMS*

ACC aminocyclopropane— CLOCK circadian locomotor output


carboxylic acid cycle(s) kaput
ACTH adrenocorticotropic hormone COP constitutive photomorphogenic
ADH antidiuretic hormone CRE cyclic AMP (cAMP)-response
AMP adenosine monophosphate element (CREB = CRE
APRR arabidopsis pseudoresponse binding)
regulator CRF corticotropin releasing
ASPS advanced sleep phase factor
syndrome CRH corticotropin releasing
AVP arginine vasopressin hormone
BAC bacterial artificial chromosome CT circadian time
bHLH basic helix—loop helix DA dopamine
BMAL brain—muscle ARNT-like DBP D-element binding protein
(ARNT = arylhydrocarbon DBT double-time
receptor nuclear transcriptor) DD constant darkness (dark-dark);
BP blood pressure differential display
BRET bioluminescene resonance DEC differentiated (human) embryo
energy transfer chondrocyte
CAB chlorophyll A/B DET deetoliate(d)
CAM crassulacean acid metabolism DMH dorsal medial (nucleus of the)
CaMK calcium/calmodulin-dependent hypothalamus
kinase DREG Drosophila rhythmically
CAT catalase; chloramphenicol expressed gene
acetyltransferase DSPS delayed sleep phase syndrome
CBT core body temperature EGFR epidermal growth factor
CCG clock-controlled gene receptor
CCR carbon catabolite repression EMS ethylmethanesulfonate
CED cis-epoxycarotenoid EMSA electrophoretic mobility shift
dioxygenase assay
CiK circadian input kinase ENU N-ethyl-N-nitrosurea
CK casein kinase EPAS endothelial PAS (see PAS)
CLD cytoplasmic localization ERG electoretinogram
domain EYFP enhanced yellow fluorescent
CLIF cycle-like factor protein
ClkRE clock-responsive element FAD flavine-adenine dinucleotide

* This list does not include very common terms (e.g., DNA, IR, UV).

xi
RHYPR 2/6/04 3:50 PM Page xii

xii ACRONYMS

FASPA familial advanced sleep phase NAD nicotinamide adenine


syndrome dinucleotide
FSH follicle stimulating hormone NAT N-acetyltransferase
FLC flowering locus C NCAM neural cell adhesion molecule
FMN flavine mononucleotide NLS nuclear localization signal
FT flowering (locus) T NMDA N-methyl-D-aspartate
GA gibberellic acid NPAS neuronal PAS (see PAS)
GABA (gamma)-aminobutyric acid ORF open reading frame
GBF G-box factor PACAP pituitary adenylate cyclase
GDRDA genetically directed activating polypeptide
representational difference PAH planar aromatic hydrocarbon
analysis PAI plasminogen activator
GFP green fluorescent protein /inhibitor
GHRH growth hormone releasing PAR proline and acid residue(s)
hormone PAS PER, arylhydrocarbon
GHT geniculohypothalamic tract receptor of mammals and
GnRH gonadotropin releasing single-minded gene in
hormone Drosophila
GRP gastrin releasing peptide PCR polymerase chain reaction
GSK glycogen synthase kinase PDF pigment dispersing factor
GST glutathione-S-transferase PEPC phosphoenolpyruvate
HEK human embryonic kidney carboxylase
HIOMT hydroxyindole-O- PEX period extender
methyltransferase pl isoelectric point
HKR histidine-kinase-related PI phosphoinositide
HLF hepatic leukemia factor PIF phytochrome interacting
HR heart rate factor
IAA indole-3-acetic acid PK prokineticin; protein kinase
IGF insulinlike growth factor (PKA = protein kinase A)
IGL interogeniculate leaflet POMC proopiomelanocortin
IMLCSC intermediolateral column of PPAR peroxisome proliferator-
the spinal cord activated receptor
IRBP interophotoreceptor retinoid PRC phase-response curve
binding protein PRR pseudoresponse regulator
LD light—dark; long day PS phytochrome substrate
LDH lactate dehydrogenase PSA poly(sialic acid)
LGN lateral geniculate nucleus PT pars tuberalis
LHC light harvesting complex PTH parathyroid hormone
LL constant light (light-light) PTX pertussis toxin
LNv ventrolateral neuron PVN paraventricular nucleus
LOV light, oxygen, voltage PVT paraventricular (nucleus of)
LRE light-responsive element thalamus
MAP mean arterial pressure QTL quantitative trait locus
MAPK mitogen-activated protein RA retinoic acid
kinase RAR retinoid acid receptor
MOP member of PAS (see PAS) RCA rubisco activase
MPN medial preoptic nucleus RD retinal degeneration (RDS =
MTHF methenyltetrahydrofolate RD slow)
RHYPR 2/6/04 3:50 PM Page xiii

ACRONYMS xiii

REM rapid eye movement (sleep SHM serine


stage) hydroxymethyltransferase
REMI restriction-enzyme-mediated SON supraoptic nucleus
integration SPZ subparaventricular zone
RHT retinohypothalamic tract SSLP simple sequence length
RGC retinal ganglion cell polymorphism
ROR retinoid-related orphan SW slow wave (sleep); sleep—
receptor wake (cycle)
RPA RNase protection assay TEF thyrotroph embryonic factor
RR response regulator TGF transforming growth factor
RXR retinoid X receptor THR thyroid hormone receptor
SAMDC S-adenosylmethionine TRH thyrotropin releasing hormone
decarboxylase TSH thyrotropin secreting hormone
SCG supracervical ganglion VIP vasoactive intestinal peptide
SCN suprachiasmatic nucleus WT wild type
(mSCN, vSCN = medial, YAC yeast artificial chromosome
ventrolateral SCN) ZEP zeaxanthine epoxidase
SD short day ZT Zeitgeber time
RHY1 2/6/04 3:51 PM Page 1

Part I
INTRODUCTION TO
CIRCADIAN RHYTHMS
RHY1 2/6/04 3:51 PM Page 3

1
GENERAL CONCEPTS
Amita Sehgal

䊏 BASIC ATTRIBUTES OF CIRCADIAN occur at different times of day. As we are


RHYTHMS becoming more aware of temporal aspects
of regulation, we are realizing that there is
The word circadian derives from the Greek scarcely a physiological process that does
words circa (about) and dian (day). Circa- not involve some cycling component.
dian rhythms then are cycles that occur
with a periodicity of ~24 hours. Given that
Example of Circadian Rhythm:
we live in a cyclic environment created by
Rest–Activity Cycle
the rotation of the Earth, it is not surpris-
ing that adaptation has involved the circa- Human sleep–wake cycles constitute
dian cycling of physiological and behavioral perhaps the best known example of a
activities. What is amazing is the extent to behavior that occurs with a ~24-hour peri-
which circadian rhythms pervade life on odicity. Many other organisms display
Earth. Previously thought to be a charac- similar behavior, which is commonly
teristic of eukaryotic organisms, circadian referred to as rest–activity cycles, taking
rhythms are now known to occur in into account the fact that “rest” is not
prokaryotes such as cyanobacteria. Indeed, always “sleep.” A record depicting rest–
as we shall see in Chapter 6, much of the activity cycles in the fruitfly, Drosophila
cyanobacteria genome, and by inference melanogaster, is shown in Figure 1.1.
much of its physiology, is expressed in a Each horizontal line in this record
cyclic fashion. Clearly, almost all organisms represents a 24-hour day, and the vertical
have evolved ways of coordinating their lines correspond to the activity of a single
physiology such that different functions fly. The activity of this fly was monitored

Molecular Biology of Circadian Rhythms. Edited by Amita Sehgal


ISBN 0-471-41824-2 Copyright © 2004 John Wiley & Sons, Inc.

3
RHY1 2/6/04 3:51 PM Page 4

4 GENERAL CONCEPTS

for a total of 14 days; the first 4 days were


measured in the presence of light–dark
cycles (12 hours of light followed by 12
hours of dark each day) and the last 10
in constant darkness. Activity of this
fly shows daily consolidation, such that it
occurs at the same time each day. This is
true even when the light–dark cycles are
removed. In addition, it is clear that this
recurring pattern, or rhythm, occurs with a
DD periodicity that is close to 24 hours. This
record is referred to in the following sec-
tions to illustrate various properties of cir-
cadian rhythms.

Control of Circadian Rhythms by


Endogenous Clocks
The issue then is how these rhythms are
generated. The first indication that rhythms
may be generated within an organism, as
opposed to being driven by the cyclic envi-
ronment, came from the observations of the
French astronomer De Mairan in 1729. In
observing the leaf movements of a Mimosa
Figure 1.1. The circadian rhythm of
rest–activity in a fruitfly. The “activity plant, he noticed that the 24-hour periodic-
record” (also called an actogram) shown ity of the movement was maintained in
here depicts the rest:activity cycle of a single constant darkness. Despite this observa-
fly over a 14-day period. Each horizontal line tion, however, it was a long time before the
represents 24 hours or a single day. The activ-
endogenous nature of these rhythms was
ity of the fly was measured by isolating it in
a glass tube and monitoring its movement accepted.
along the tube, based on the deflection of Critics of the endogenous control idea
an invisible light beam projected into the argued that even under so-called constant
tube (see also Chapter 3). The vertical ticks conditions (e.g., constant darkness) it was
(shading) in the record correspond to activ-
not clear that all environmental influences
ity. This particular fly was monitored in
light–dark cycles for 4 days and thereafter in relating to the Earth’s rotation were absent.
constant darkness. The position of the arrow In the 1930s Kalmus and Bunning argued
indicates the transfer to constant darkness for an endogenous mechanism based on the
(DD). The light–dark bar at the top indicates finding that the periodicity of the rhythm
the timing of the light–dark cycle. As is
varied with temperature fluctuations that
evident from the record, the circadian
rhythm of rest:activity persists in constant were clearly independent of the Earth’s
darkness, but it now “freeruns” with its rotation. Although their findings of temper-
endogenous periodicity instead of being ature dependence were subsequently dis-
driven by the environment. puted by research showing that circadian
rhythms are temperature-compensated (see
discussion below), they were obviously
right about endogenous control. In the
1940s the idea that endogenous clocks
RHY1 2/6/04 3:51 PM Page 5

BASIC ATTRIBUTES OF CIRCADIAN RHYTHMS 5

control circadian rhythms gained wider It should be noted that not all rhythmic
acceptance. What helped to reinforce this functions are controlled by an endogenous
was the finding that the periodicity of circa- clock. Some are driven by the day–night
dian rhythms under noncycling environ- cycle and therefore do not persist in con-
mental conditions varies from species to stant conditions. These are referred to as
species. More importantly, the period in diurnal rhythms, to distinguish them from
most cases did not precisely match that of circadian rhythms, which, by definition,
any environmental cycle, indicating that it persist in noncycling environmental con-
was most likely endogenously generated. ditions. In humans, rhythmic processes
For instance, in the record shown in Figure include sleep–wake cycles, release of
1.2, note how, in constant darkness, activity several hormones, regulation of body tem-
each day begins and ends a little earlier than perature, blood pressure, urine production,
it did the day before. This drift occurs cholesterol metabolism, respiratory func-
because the periodicity of this rhythm, and tion, and incidence of asthma and heart
thereby that of the underlying clock, is a attacks. While some of these are clearly
little less than 24 hours. In the presence of under circadian control, others may turn
light–dark cycles, the drift does not occur out to be dependent on other factors (e.g.,
because, as discussed below, light resets the behavioral state). For instance, the release
clock. of some hormones appears to be rhythmic
Despite the general acceptance of the because it is influenced by sleep, which nor-
idea, some skeptics remained, and this mally occurs in a cyclic fashion, rather than
skepticism inspired Hamner and others to by clock function (see Chapter 12).
conduct an interesting experiment in the
early 1960s. This experiment was performed
Circadian Rhythm Properties
at the South Pole with a number of organ-
isms, all of which were maintained on a This periodicity of ~24 hours is one attrib-
rotating turntable. Although the turntable ute of circadian rhythms. A second is that
rotated with the same periodicity as the these rhythms can be synchronized or
Earth, it did so in the opposite direction. reset by environmental cues. The dominant
Under these conditions, the circadian environmental signal that affects rhythms
rhythm of eclosion in Drosophila pseud- is light, with most rhythms synchronized
oobscura persisted, as did a number of to the day–night cycle. Jurgen Aschoff
other physiological rhythms. used the term zeitgeber (“timegiver” in

Environmental Overt rhythm


signal (e.g., light) (e.g., sleep-wake cycle)
Input pathway Output pathway

Figure 1.2. A simple circadian system. The very basic circadian system is thought to contain
a clock, an input pathway that transmits environmental signals to the clock, and an output
pathway that carries signals away from the clock and results in the manifestation of overt
rhythms. In practice, there can be multiple input and output pathways to serve a single clock.
In addition, the system most likely does not work in the linear fashion depicted here. Outputs
can feedback on the clock and perhaps even on input and the clock can feedback on the
input pathway.
RHY1 2/6/04 3:51 PM Page 6

6 GENERAL CONCEPTS

German) to denote a periodic environmen- determined under freerunning conditions.


tal signal that synchronizes or sets the Again, note how in Figure 1.1, the pattern
time of a circadian rhythm. The process of of the rhythm in constant darkness
synchronization itself is called entrain- (freerun) is somewhat different from that
ment. Since most rhythms are synchronized in the presence of light–dark cycles. This is
to environmental cycles, chronobiologists because in constant darkness it runs with its
refer to time as defined by these environ- endogenous periodicity. Under these condi-
mental cycles, thereby calling it zeitgeber tions, we speak in terms of circadian time
time (ZT). When an organism is maintained (CT), which is the time of the internal
in a 24-hour cycle that consists of 12 hours clock. Since the periodicity of the internal
of light, followed by 12 hours of darkness, clock is frequently a little different from 24
zeitgeber time or ZT0 corresponds to hours, CT is not the same as ZT. However,
“lights on” and ZT12 is “lights off.” Thus, on the first and second days of constant
all timepoints between 0 and 12 refer to darkness, when the endogenous rhythm has
daytime hours, while those between 12 and not drifted significantly from the environ-
24 (which is the same as “0”) refer to night- mental cycle, CT may approximate ZT for
time hours. This terminology will be used an organism that has a periodicity close to
throughout this book. 24 hours, and researchers may use it as
Light then is a major zeitgeber that such.
entrains circadian rhythms. The time at Finally, the periods of circadian rhythms
which a rhythmic activity/process occurs are temperature compensated. This was
defines the phase of that particular rhythm. demonstrated in some classic experiments
In the record shown in Figure 1.1, the time done by Colin Pittendrigh in the 1950s. He
at which the fly is active indicates the phase determined the effects of temperature on
of its rest–activity rhythm. Likewise, the the eclosion (hatching of adult flies from
time at which it sleeps, or rests, can also be pupae) rhythm in Drosophila pseudoob-
used as a measure of phase. Typically, the scura flies. In his first series of experiments
parameter that is most constant from day he improvised “incubators”—a pressure
to day (whether wakeup time or sleep time) cooker maintained in a cold-water creek
is used to define the phase of a rhythm. It and an outhouse darkroom, to maintain
is clear that nocturnal and diurnal animals the flies at cold and warm temperatures re-
display opposite phases in their sleep–wake spectively. He found that the periodicity of
rhythms. A change in the timing of the the rhythm was relatively the same (within
zeitgeber, as might be caused by travel to a an hour) at the two temperatures. In
different time zone, results in a shift in subsequent experiments done at Princeton
the phase of the rhythm. However, as we all University, he confirmed these findings,
well know, this shift does not occur imme- showing that the periodicity was essentially
diately, thereby resulting in the phenome- the same at 26 and 16°C.
non commonly known as “jet lag.” During Thus, the circadian period remains
the process of adjustment to a new cycle a constant over a wide temperature range.
rhythm displays “transient” behavior, con- Assuming that the underlying basis of overt
sisting of erratic phase and periodicity. rhythms is some biochemical mechanism,
To reiterate an important property of as indeed must be the case, then normally
circadian rhythms, they can persist in the changes in protein function at different
the absence of cyclic environmental cues. temperatures would alter the rate of the
Under these conditions they are said to reaction and thereby the periodicity of the
be freerunning. The endogenous nature of rhythm. The rate of most physiological
a rhythm and its periodicity have to be processes doubles with a 10°C rise in
RHY1 2/6/04 3:51 PM Page 7

BASIC ATTRIBUTES OF CIRCADIAN RHYTHMS 7

temperature. The fact that this does not follows that there must be more than one
happen to circadian periodicity indicates output pathway. In fact, we now know
that there is some mechanism that com- (as described below) that many of these
pensates for these temperature-induced outputs are actually generated not just by
changes. Clearly, it is important for organ- independent output pathways, but by sepa-
isms to maintain a periodicity that approx- rate clocks within an organism.
imates that of the day–night cycle despite Together, the clock, input pathway and
changes in environmental temperature. output pathway are termed a circadian
While homeotherms can obviously do this system. Although the components of this
with no problem, organisms that do not system are typically depicted in a linear
maintain a constant body temperature had diagram, this is an oversimplification of the
to evolve mechanisms for this purpose. mechanisms involved. In many cases, the
outputs can feed back and affect the clock
(e.g., forced activity can reset the clock),
Circadian Clock Input/Output
and the clock may feedback to regulate
Circadian clocks are part of a system that input, resulting in the response to light
includes input and output components being stronger at certain times of day. The
(Fig. 2.2). If rhythms are generated by an response to light is said to be “gated” by the
endogenous clock—and, indeed, at this clock.
point we can say that they are, based not
only upon the investigations mentioned
Clock Entrainment to Input
above, but also upon a vast amount of
Components (Light)
genetic and molecular data that are the
subject of this book—then, if follows that As mentioned above, most biological
the properties of rhythms arise from in- clocks have periodicities that are not pre-
herent characteristics of the clocks that cisely 24 hours. Aschoff suggested that the
produce them. In fact, clocks are entrained periodicity of the same organism may be
by external/environmental signals through different under different conditions,
what is termed an input or entrainment depending on the functional state of the
pathway. The implication is that the clock organism or even the nature of the “con-
itself may not be in contact with the envi- stant” environment (e.g., constant light vs.
ronment, but can communicate with it constant dark). He was intrigued by the fact
through a distinct pathway(s). Since the that light-active (diurnal) animals show an
predominant entraining cue is light, the increase in spontaneous frequency, as meas-
input pathway is usually perceived as con- ured by increased bouts of activity, with
taining a photoreceptor. As we shall see in increasing intensity of light. This increased
subsequent chapters, the identification of frequency of the oscillation results in an
the photic input pathway is an intense area overall shorter period. Night-active (noc-
of research in every organism used for the turnal) animals, on the other hand, decrease
study of circadian biology. their spontaneous frequency with increas-
Time-of-day signals generated by the ing intensity of light, thereby displaying
clock must be transmitted to other parts of longer periods. On the basis of these obser-
the organism to drive physiology/behavior vations, Aschoff proposed a circadian
in a rhythmic fashion. The pathway that rule according to which diurnal animals
carries signals away from the clock and have shorter than 24-hour rhythms in LL
produces overt rhythms is called the (constant light) and longer than 24-hour
output pathway. Since outputs can vary rhythms in DD (constant darkness). The
greatly, even within the same organism, it reverse would be true in nocturnal animals.
RHY1 2/6/04 3:51 PM Page 8

8 GENERAL CONCEPTS

However, as Aschoff himself recognized, triggered by the light signal.The animal does
there are exceptions to this rule. Drosophila not become active in anticipation of the
are diurnal and yet display periodicities dark–light transition. In a nocturnal animal
that are slightly less than 24 hours (~23.8 light would have the opposite effect. This
hours) in DD. effect of light that is independent of the
Regardless of the periodicity of the endogenous clock is termed “masking,” so
endogenous rhythm all organisms are called because it can mask the manifestation
synchronized to the 24-hour environmental of the endogenous rhythm. Thus, even in an
cycle. This means that they must be reset animal that has a clock, masking effects of
every day because otherwise they would light can confound analysis of the endoge-
eventually drift out of synchrony with the nous rhythms, which are therefore best
day–night cycle.Take, for instance, a rhythm assayed under freerunning conditions (i.e.,
that has a period of 23.8 hours. If it were constant darkness and constant tempera-
to freerun, then on each successive day it ture). It should be noted that although
would be an additional 0.2 hour out of masking, as described here, denotes clock-
phase with the environment. In 2 months it independent effects of light on the activity
would have a reverse phase, relative to the rhythm, it can, in principle, apply to any
environment, from the one with which it influence that obscures a rhythmic function.
started. A rhythm such as this, which has
a shorter period than the environmental
Entrainment to Light Pulses
cycle, is delayed by 0.2 hour each day. This
is illustrated by the example shown in In addition to being entrained to day–
Figure 1.1, where the endogenous periodic- night cycles, circadian rhythms can be en-
ity of the fly is ~23.5 hours, but it synchro- trained by pulses of light. This type of
nizes to a 12–12 light–dark cycle, because it entrainment is called nonparametric, distin-
is reset each day. A longer period rhythm guishing it from parametric entrainment
must advance each day by the amount of which involves synchronizing to cycles of a
time its periodicity differs from that of stimulus. In general, a pulse of light deliv-
the day–night cycle. The manner of the ered when an organism expects to see light
resetting can differ between rhythms. A (“subjective day”, as it is called in constant
rest–activity rhythm could advance each darkness) does not reset the rhythm.
day by initiating either the rest or the active However, pulses at night reset the rhythm
phase earlier than dictated by the internal with those in the first part of the night
clock. These earlier onsets are typically having effects opposite those in the second
driven by something in the environment half of the night. A pulse in the early night
(e.g., sunrise or sunset). delays the phase of the rhythm. It can be
As will be seen in subsequent chapters, thought of as analogous to extended day-
even organisms that lack clock function, light, which has the effect of delaying the
through genetic manipulation, are driven by onset of the night function (e.g., in a diurnal
the environmental cycle to display 24-hour animal it might delay the onset of sleep or
rhythms. The difference between these rest). Conversely, a pulse in the second half
rhythms and those in clock-containing of the night advances the phase of the
organisms is that they cannot persist when rhythm. Again, the best analogy perhaps is
the rhythmic signals from the environment with a shift in the natural day–night cycle.
are removed. In addition, they cannot antic- In this case, it would be like dawn occurring
ipate the daily transitions in the environ- earlier, which would lead to earlier onset of
mental cycles. For instance, in a diurnal the next day’s activities. The idea is that a
animal that lacks a clock, activity is usually pulse in the early night resets the cycle to
RHY1 2/6/04 3:51 PM Page 9

BASIC ATTRIBUTES OF CIRCADIAN RHYTHMS 9

dusk, while one in the late night resets it to Some organisms are more sensitive to light
dawn. In practice, of course, the resetting is and display a Type 0 PRC (Fig. 1.3b). A
not quite so perfect. Delays are usually of Type 0 PRC is characterized by larger mag-
larger magnitude and easier to achieve than nitude shifts and the absence of any
advances. crossover point in the middle of the night.
A graph that plots the magnitude of the This lack of a crossover typically gives a
shift relative to the time of day that the false impression of discontinuity in the
shifting stimulus is delivered is called a PRC; in a rhythm with a 24-hour period, a
phase-response curve (PRC). A typical such 12-hour delay is indistinguishable from a
curve is shown in Figure 1.3a.As mentioned 12-hour advance, so a shift from plotting
above, there is no shift during the subjec- strong delays to weaker advances produces
tive day, which is thereby called the “dead the apparent discontinuity in the plot.
zone.” In the middle of the night, the curve Although previously regarded as very dis-
crosses over from delays to advances. In tinct entities, there is now increasing evi-
this type of PRC, also called a “Type 1 dence that with strong light pulses (either
PRC,” pulses of light at the crossover point of higher intensity or increased duration) a
do not produce any shift in the rhythm. Type I PRC can be turned into a Type 0.

(a) (b)

4
Shift (in hours)

0
12 24 12 24
-2 Circadian time

-4

Figure 1.3. Entrainment to pulses of light. Based on the response of an organism to reset-
ting stimuli delivered at different times of day, phase response curves (PRCs) can be con-
structed. This type of curve basically demonstrates the effect of time of day on the magnitude
of the phase shift produced by a specific stimulus. The best documented PRCs are those in
response to light stimuli and these are of two major kinds, shown in (a) and (b). The light–dark
cycle used to entrain the organism is indicated at the bottom of each plot. The number of
hours by which the phase of the clock shifts (y axis) in response to a light pulse delivered at
different times of day (x axis) is plotted. In a Type 1 PRC (a), pulses of light delivered during
a time when an organism would expect to see light (daytime hours) have no effect on the
phase of the rhythm. A pulse of light in the early part of the night delays the phase (indi-
cated by negative numbers), and a pulse in the second half of the night advances the rhythm
(indicated by positive numbers), with a gradual transition between phase delays and phase
advances. In a Type 0 PRC, the clock is very sensitive to light and shows large shifts, in response
to pulses, at all times of the cycle, with no gradual change from phase delays to phase
advances.
RHY1 2/6/04 3:51 PM Page 10

10 GENERAL CONCEPTS

Multiple Clocks the master or central clock (if there is one)


is usually located in the brain (Fig. 1.4).
An organism can have more than one cir- Clocks in other parts of the body are
cadian clock. It has now become clear that referred to as peripheral oscillators to dis-
most multicellular organisms have more tinguish them from the central clock. The
than one clock. It appears that with increas- degree of autonomy that these oscillators
ing complexity of physiological function, possess varies from species to species and
there is increasing need to have clocks also from oscillator to oscillator. Insects
specific for different organs/tissue systems. contain many clocks that are autonomous
The question then becomes: Is there one with respect to timekeeping as well as with
clock that controls all others in an organism respect to photoreception. However, there
—a master clock, if you will—or are all is at least one oscillator in Drosophila,
clocks equal? Among the animal systems, the insect of choice for molecular analysis

Light
Output
Peripheral
clock

Central clock
(brain) Peripheral Output
clock

Peripheral Output
clock

Light
Output

Peripheral
clock
Central clock
(brain) Peripheral Output
clock

Peripheral
clock
Output

Figure 1.4. Organization of clocks in complex organisms. In mammals, a light-responsive


central clock in the brain regulates clock activity in clocks found in peripheral tissues (top).
The peripheral clocks are not directly photosensitive although they can entrained by other
signals (e.g., hormones). In other organisms, peripheral clocks can be photosensitive and vary
in their dependence upon the central clock.
RHY1 2/6/04 3:51 PM Page 11

CLOCK MECHANISMS 11

of clock function, that is controlled by 䊏 CLOCK MECHANISMS


the central clock although it may also be
capable of direct photoreception. Likewise, Since the general acceptance of the idea
most oscillators in the nonmammalian that clocks are endogenous, a major ques-
vertebrate, zebrafish, are autonomous tion has been: How, within an organism,
and have their own photoreceptors, do you make a clock? Over the years, a
although interactions with the central number of mathematical models have been
clock likely occur. Other nonmamalian developed to explain how this might occur.
vertebrates, that do not have the transpar- One aspect that these models have in
ent skin of the zebrafish, may not show common is the presence of a feedback loop.
this extent of autonomy in peripheral Overt cycles must have their basis in an
oscillators. endogenous cycle or loop that can sustain
In mammals, there is a master clock that oscillations indefinitely in the absence of
is housed in the hypothalamus and is called environmental cues. The question then is
the suprachiasmatic nucleus (SCN). Thus, how these loops are generated. Do they
clock functions appear to have become cen- operate at the level of the organism, the
tralized over the course of evolution as tissue, or the cell? As outlined below and
have other body functions (Fig. 1.4). The described in detail in other chapters in this
SCN can sustain clock activity for extended book, in all organisms examined, the basic
periods of time in culture. It is also the only clock mechanism (i.e., the oscillatory loop)
clock in the body, with the exception of is, in fact, intracellular. While an intracellu-
the eye clock, which receives photic infor- lar clock might be expected of a cyanobac-
mation. Details of this can be found in terium or of the bread mold Neurospora, it
Chapter 4. comes as a surprise when it turns out to be
Although peripheral oscillators in the case in a mammal. This is not to say that
mammals cannot perceive light, they can timekeeping is performed by a single cell.
be entrained by other physiological stimuli. In multicellular organisms, a clock tissue
Most notable are hormones, which might (central or peripheral) is likely to be com-
be controlled by the SCN, but may also act posed of a cluster of cells, each of which
independently on peripheral oscillators. In contains a loop that oscillates in synchrony
addition, retinoids will reset a clock in the with the loops in neighboring cells. An SCN
vasculature, and restricted feeding can is an example of such a cluster. Rhythmic
entrain the clock in the liver without affect- signals generated by these clusters drive
ing the SCN. Clearly, the organization of output rhythms.
circadian systems in terms of central and As one might have gathered from the
peripheral oscillators, the interactions discussion above, the general nature of
between the different components and the the clock mechanism is conserved from
mechanisms that entrain them are all cyanobacteria to mammals. However,
important aspects of physiology. These since circadian rhythms are present in
issues are discussed in the chapters on widely separated phyla and the actual
peripheral oscillators and hormonal molecules involved are different in the
rhythms. Specific details that pertain to diverse phyla, the prevailing notion is that
individual organisms can be found in the clocks evolved several times. The fact that
chapters on these respective organisms. the same mechanism evolved in each case
attests to the efficacy of this particular
mechanism. In addition, the availability of
multiple, biological systems that use the
same mechanism has allowed a powerful,
RHY1 2/6/04 3:51 PM Page 12

12 GENERAL CONCEPTS

multidisciplinary approach to the study of cific genes regulate their own expression.
clocks. Particularly important have been These genes are called clock genes, and the
studies in model organisms, which offer autoregulation consists of the proteins
powerful genetic approaches and simpler negatively regulating synthesis of their own
systems than more complex vertebrates. mRNAs (Fig. 1.5). Each complete turn of
A general model for the clock mechanism the loop takes ~24 hours to complete,
was first worked out in Drosophila and resulting in circadian oscillations of RNA
then in Neurospora. Mammalian studies of and protein levels. Rhythmic expression
clock function built largely on the infor- of the proteins ensures that the negative
mation that was already available from feedback on transcription is rhythmic,
these other systems, in particular on the which, in turn, ensures rhythmic expression
framework provided by the Drosophila of the mRNA. The precise molecules that
model. This book highlights this multiple- function in this fashion may differ from
species approach to the molecular analysis one species to another although certain
of rhythms by describing the salient fea- structural features are conserved from
tures of the clock, input and output path- cyanobacteria to mammals. Between Dro-
ways in different species and comparing, sophila and mammals, the molecules are
wherever applicable, the similarities in the also conserved.
systems. Given that this is a cyclic process, it is,
of course, difficult to ascertain what comes
first—the RNA cycling or the protein
Clock Mechanism at a Molecular Level
cycling. It is clear that the mRNAs do not
The basic clock mechanism is a feedback cycle in the absence of rhythmic feedback
loop in which oscillating products of spe- from the proteins. However, the most

Gene X

RNA
Negative
feedback

Protein

Figure 1.5. The basic model for a 24-hour oscillator. The model is based on work done
in many different species ranging from cyanobacteria to mammals and consists of an autor-
egulatory feedback loop in which a clock gene encodes cycling RNA and protein, and the
protein negatively regulates synthesis of its own mRNA. The loop takes ~24 hours to com-
plete (shown within a 24-hour clock).
RHY1 2/6/04 3:51 PM Page 13

CLOCK MECHANISMS 13

recent data indicate that cycling of specific those that drive a rhythm in cholesterol
clock proteins can be maintained under metabolism in the liver.
conditions where their RNAs do not cycle. What is clear, however, is that one of the
Posttranslational mechanisms, such as cyclic ways clock genes transmit output signals
phosphorylation, appear to contribute to is by driving rhythms of gene expression.
rhythmic expression of clock proteins and Although the number varies from species
may account for oscillations of these pro- to species, we now know that a significant
teins under conditions where cycling of the proportion of the genome is expressed
corresponding mRNAs is blocked. It is rhythmically in all organisms that possess
important to note also that these experi- clocks. The number as well as the nature of
ments have not eliminated the cycling of all the genes expressed in a rhythmic fashion
clock gene mRNAs in an organism, so the vary not only from species to species but
cycling of one may be able to compensate also from one organ or tissue to another
for loss of cycling of the others. in the same organism. A gene expressed
Although we have refrained from the rhythmically in one tissue may not be
use of chronobiology jargon in this book, it expressed rhythmically in another tissue,
is worth introducing some of the terminol- because it serves a different function in the
ogy used in the field here because it may other tissue or because the process with
appear in other readings of the circadian lit- which it is involved does not cycle in
erature. A molecule that is part of the clock the other tissue. The rhythms in gene
is frequently referred to as a clock compo- expression are translated into rhythms in
nent. A clock component can be a state protein expression and function that ulti-
variable, which is a molecule that imparts mately give rise to rhythms in physiology
time-of-day cues through rhythmic changes and behavior.
in its levels or activity, or a state parameter, The most direct mechanism by which the
which does not itself cycle, but is required clock can drive a rhythm in gene expression
for the appropriate regulation of the state is by rhythmically regulating promoter activ-
variable. Researchers have developed crite- ity. Promoters of many genes contain ele-
ria for state variables, which include a ments recognized by transcription factors
requirement that its levels or activity cycle, that are part of the clock. Rhythmic activity
that it regulate itself through a feedback of these factors results in rhythmic expres-
mechanism and that mutations in it sion not only of clock genes but also of genes
produce effects on overt rhythms. that are downstream of the clock (Fig. 1.6).
These downstream genes may encode other
transcription factors or other proteins that
Gene Expression
affect a cell property or function.
We are starting to also gain an understand- However, it is important to note that
ing of how it is that clocks communicate clocks can also regulate downstream com-
their temporal signals to the rest of the ponents through transcription-independent
organism, although these “output pathway” mechanisms. Although the mechanisms
mechanisms represent the least understood are not known yet, there are examples of
aspect of circadian rhythms. The diversity circadian components that cycle at the
of overt rhythms, and thereby that of the protein, but not the RNA level. In addition,
output pathways that lead to these rhythms, in Drosophila, a neuropeptide that is
may account, in part, for the dearth of released specifically by brain clock cells
knowledge in this area. Clearly, the mole- cycles at axon terminals. Since expression
cules and mechanisms that produce rhyth- of this protein does not cycle in the cell
mic rest–activity will be very different from body, the interpretation is that the levels do
RHY1 2/6/04 3:51 PM Page 14

14 GENERAL CONCEPTS

Clock gene Clock-controlled Clock-controlled


gene gene

Transcription factors,
hormones,
ion channels

Figure 1.6. Control of downstream genes by the clock. A molecular clock (such as the one
shown in Fig. 1.7) can regulate the output pathway by driving cyclic expression of down-
stream genes. One mechanism by which this occurs is shown here. While regulating its own
transcription, the clock protein also regulates the transcription of downstream genes at a spe-
cific time of day. This leads to rhythmic expression of those genes.

not cycle, but the release from axon termi- field and so the material presented here
nals does. A simplistic model for circadian provides the basis, but by no means the
control of a complex function, such as complete picture, of molecular clock
rest–activity behavior, is that oscillations in function.
clock genes result in oscillations in some
cellular (neuronal) property. This oscilla-
tion in turn drives rhythmic expression of 䊏 ADAPTIVE SIGNIFICANCE OF
an output (perhaps a hormone) from the CIRCADIAN RHYTHMS
cell, and that output mechanism then acts
rhythmically on cells controlling rest–activ- An introduction to circadian rhythms
ity (Fig. 1.7). This results in the manifesta- would, of course, be incomplete without
tion of rest–activity rhythms. In a simple some discussion of their adaptive signifi-
circadian system the final output rhythm cance. Several possible explanations have
may be produced directly by the clock cell. been proposed. In 1987 Rapp argued that
Thus the process is more complicated biological systems are periodic because
than would appear at first glance. These temporal organization allows for better
complexities, most of which are not com- synchrony or coordination between differ-
pletely understood yet, are acknowledged ent processes in the same organism, it
here and elsewhere in this book. However, allows incompatible processes to be sepa-
at the same time, it is clear that we have rated from each other in time (this is
learned a lot about how clocks operate in particularly advantageous in unicellular
many different species. Amazingly, most of organisms; see Chapter 6) and it allows for
this progress has occurred very recently (as a more precise, efficient system.
of 2003). In the late 1990s a book focused Perhaps it is easy to see how adaptation
on molecular mechanisms underlying clock to a cyclic environment would be facilitated
function might have seemed premature. by cyclic physiology or behavior. However,
Now it is perceived as something that is most rhythms can be driven by the envi-
needed. It is important to note, though, that ronmental cycle. As mentioned above, even
this is still a rapidly moving and growing organisms that lack clocks, or contain
RHY1 2/6/04 3:51 PM Page 15

ADAPTIVE SIGNIFICANCE OF CIRCADIAN RHYTHMS 15

Rhythmic rest-activity

Cyclic molecule Cyclic membrane Cyclic hormone Cyclic behavior


potential release

Molecular Cellular Physiological Behavioral

Figure 1.7. Control of an overt, behavioral rhythm by the clock. A simple model is shown in
which the clock drives rhythmic expression of a gene that regulates membrane potential.
Cycling membrane potential results in rhythmic release of a secreted factor that has an effect
on levels of rest and activity.

mutant clocks with aberrant periodicity, can of animals that lack one or more clock
be driven to display rhythmic activity in a genes in all tissues and so are arrhythmic.
12-hour light–12-hour dark cycle. Given Interestingly, early attempts to identify via-
this, why do we then need endogenous bility or fertility deficits in these animals
clocks? The most commonly cited explana- failed. The advantage conferred by clocks
tion for endogenous rhythmicity is that may not be evident under laboratory con-
it enables organisms to anticipate daily ditions where the animals are supplied with
transitions in the environment. It is stated ample food and water and do not have to
above that changes in the activity patterns fend for themselves. However, more
of clockless animals are triggered by the detailed analysis is now revealing deficits
environmental stimulus (say, light). An associated with loss of clock function. Fur-
animal with a clock becomes active in antic- thermore, researchers found a small reduc-
ipation of the dark–light transition, which tion in lifespan in some of the Drosophila
may confer a selective advantage on it. It is mutants that affect circadian period.
a case of the early bird getting the worm. The differences in lifespan between wild
In addition, a clock allows finer temporal type and mutants were altered somewhat
control over physiological processes. in light–dark cycles of different lengths.
The advent of cellular and genetic tools However, the reduction in lifespan was
allowed researchers to experimentally test seen even when the mutants were main-
the importance of clocks. The basic experi- tained in a light–dark cycle that approxi-
ment is to remove the clock and assay sur- mated the periodicity of their internal
vival or fitness. Cellularly, this has been clock. Thus, the advantage of having a ~24-
done by lesioning the SCN. Although we hour rhythm in Drosophila may extend
now know that clocks are found in many beyond the need to match the periodicity of
tissues outside the SCN, the master clock in the environmental cycle. In cyanobacteria,
mammals is in the SCN. In addition, synchrony with the environmental cycle
genetic tools have allowed the generation confers a selective advantage. When cyan-
RHY1 2/6/04 3:51 PM Page 16

16 GENERAL CONCEPTS

obacterial strains of different periodicities chipmunks: Essential for survival? J Comp


are grown in the same culture, the ones that Physiol [A] 186: 169–180.
grow best, and thereby outcompete their Dunlap, JC, Loros, JJ, DeCoursey, P (eds)
neighbors, have periodicities that match (2003): Chronobiology: Biological Timekeep-
ing, Sinauer Assoc.
that of the environment. Finally, studies of
Klarsfeld, A, Rouyer, F (1998): Effect of circa-
SCN-lesioned mammals released into the
dian rhythms and LD periodicity on the
wild have also revealed advantages of lifespan of Drosophila melanogaster. J Biol
having clocks. The lack of a clock appears Rhythms 13: 471–478.
to make these animals more susceptible to Mori, T, Johnson, CH (2001): Circadian pro-
predators. gramming in cyanobacteria. Semin Cell Dev
Biol 12: 271–280.
Pittendrigh, CS (1954): On temperature inde-
pendence in the clock system controlling
FURTHER READING emergence time in Drosophila. Proc Natl
Acad Sci (USA) 40: 1018–1029.
Aschoff, J (1960): Exogenous and endogenous Pittendrigh, CS (1993): Temporal organization:
components in circadian rhythms. Cold Spr Reflections of a Darwinian clock-watcher.
Harb SympQuant Biol 25: 11–28. Ann Rev Physiol 55: 17–54.
Bunning, E (1973): The Physiological Rapp, PE (1987): Why are so many biological
Clock. Circadian Rhythms and Biological systems periodic? Prog Neurobiol 29: 261–
Chronometry, The English Universities Press. 273.
DeCoursey, PJ, Walker, JK, Smith, SA (2000): Saunders, DS (1982): Insect Clocks, Pergamon
A circadian pacemaker in free-living Press, Oxford (Oxfordshire); New York.
RHY2 2/6/04 3:51 PM Page 17

2
GENETIC AND MOLECULAR
APPROACHES USED TO
ANALYZE RHYTHMS
Amita Sehgal and Jeffrey L. Price

Since the approaches used to study the ally part of the clock, has led to a standard
molecular basis of circadian rhythms are, to set of questions that are now asked about
a great extent, the same in different species, every new candidate circadian gene. These
it is worth discussing the general nature of questions include
these approaches. Genetic approaches have
been by far the most successful at unravel- • Do levels of the RNA and protein cycle?
ing the intricate mechanisms that underlie • Is it expressed in clock cells?
clock function. Whether in cyanobacte- • Is it part of the clock?
ria, Arabidopsis, Neurospora, Drosophila, • Does it show an acute response to light?
or mouse, genetic screens, in which organ- • Does it interact with other clock
isms subjected to random mutagenesis are proteins?
screened for circadian phenotypes, have
provided a wealth of information. They As a result, regardless of the organism under
have identified genes involved in every study, these are the questions that must be
aspect of the circadian system. What we addressed. Although the precise methodol-
have since learned about some of these ogy may differ,the overall approaches are the
genes, in particular the genes that are actu- same and are discussed below.

Molecular Biology of Circadian Rhythms. Edited by Amita Sehgal


ISBN 0-471-41824-2 Copyright © 2004 John Wiley & Sons, Inc.

17
RHY2 2/6/04 3:51 PM Page 18

18 GENETIC AND MOLECULAR APPROACHES USED TO ANALYZE RHYTHMS

䊏 USE OF GENETIC SCREENS TO from species to species. Classical methods


IDENTIFY CIRCADIAN GENES use chemical mutagens, the concentrations
of which are titrated to yield maximal
Random Mutagenesis Screens mutagenesis without overly compromising
the viability or fertility of the organism.
In the absence of any initial knowledge Another popular method for mutagenesis,
about the molecules that participate in a particularly in Drosophila, is transposon
process, a genetic analysis is frequently the mobilization. Transposons are small seg-
best way to initiate an analysis of the ments of extraneous DNA that can insert
process (see Fig. 2.1). A genetic analysis into chromosomes largely, although not
requires no a priori assumptions about the entirely, in a random fashion. They can be
mechanism of the process. Instead, it iden- mobilized by introducing an enzymatic
tifies mutants in which the process (e.g., a activity (transposase), and they frequently
circadian rhythm such as locomotor activ- affect gene expression at sites of insertion.
ity) is altered. The identity of the affected In order to select for germline mutations,
genes and the elucidation of the pathways it is the progeny of mutagenized indivi-
with which they are involved may reveal duals that are screened in the assay of
the mechanism. Conceptually, genetic interest. The assay historically consists of
analysis is analogous to figuring out how an measurement of an overt rhythm, although
automobile works by removing or altering researchers have also developed methods
parts, and then determining the effects to screen for disrupted molecular rhythms.
of this manipulation on the functioning of Also, to date, the emphasis has been on
other parts and of the car as a whole. freerunning phenotypes, and so the rhythm
The difference is that one does not choose is assayed under constant conditions.
the part to remove as the mutants are Candidates that show aberrant phenotypes
generated through random mutagenesis. are bred or propagated and subsequent
To maximize the chances of detecting a generations are further analyzed.
handful of circadian genes from the vast,
total number of genes in the genome, large
Mutant Phenotypes
numbers of mutagenized individuals must
be screened. The mutant phenotype may provide some
The methods used for mutagenesis vary clues regarding the role of the gene in the


Figure 2.1. Isolation and analysis of circadian genes through forward mutagenesis. A
schematic representation of the standard protocol is shown. In a forward mutagenesis screen
the organism is mutagenized and the progeny are screened for circadian phenotypes. Mutants
with interesting phenotypes are rescreened, and the relevant mutations are then mapped
and subjected to molecular cloning. Once the gene is in hand, cDNAs (complementary to the
mRNA) for it can be obtained, and these provide probes for RNA detection and reagents for
the production and analysis of protein. The gene itself, with its promoter, exons, and introns,
provides DNA elements that regulate expression of the gene (e.g., the promoter and elements
that mediate RNA stability or splicing). The cDNA and genomic clones (either wild-type
or mutant forms constructed by the experimenter) can also be expressed in transgenic cells
or organisms to test specific hypotheses for the circadian mechanism. For instance, as noted
in the text, the promoter could be fused to a reporter like luciferase and expressed in the
organism to determine if it drives cyclic expression. It could also be expressed in cultured cells
and tested as a target for candidate transcription factors.
RHY2 2/6/04 3:51 PM Page 19

19
RHY2 2/6/04 3:51 PM Page 20

20 GENETIC AND MOLECULAR APPROACHES USED TO ANALYZE RHYTHMS

circadian system. Genetic analysis may elu- all its outputs with the rhythmic release of
cidate input and output pathways, as well as a neurotransmitter, a mutation that elimi-
the mechanism of the circadian oscillator. nated the synthesis of the neurotransmitter
As discussed in Chapter 1, the circadian could produce arrhythmia in all of the
paradigm consists of input pathways, which known outputs without an effect on the
keep the clock in phase with environmen- oscillator mechanism per se. Eventually,
tal cues such as light and temperature molecular assays (discussed below) can be
cycles (a process termed entrainment); a used to determine whether the oscillator
circadian oscillator, which is entrained by mechanism is affected.
the input pathway but capable of sustaining If a gene can be mutated to alter the
an oscillation in the absence of environ- period length of circadian outputs, it cannot
mental cues; and output pathways, which be involved with just an output pathway. Its
convey the temporal information of the effect on the period length would indicate
oscillator to the diverse processes it regu- that it is affecting the basic timekeeping
lates (Fig. 1.2). While this paradigm may be mechanism. This could be through a direct
somewhat simpler than the system is in effect on the clock (i.e., the mutation is in a
reality, it has been of enormous heuristic clock component), through an effect on an
value. If a gene is necessary for any of these input component that, by virtue of being
pathways, a mutation producing a nonfunc- upstream of the clock, can affect clock func-
tional gene product can disrupt overt circa- tion, or through modulation of an output
dian rhythms such as those of rest–activity that feeds back to affect clock function.
(Fig. 1.1). For instance, mutations of a It can be difficult to distinguish between
particular gene that is necessary for a clock- these possibilities. Mutations in the input
controlled process, or for passing temporal pathway may be delineated through assays
information from the clock to the overtly for entrainment—for example, either
rhythmic process (the “output’), would entrainment to light–dark cycles or to acute
produce arrhythmia in the output process, pulses of light. The prediction for a clock
while leaving the circadian oscillator intact. component is that a null mutation, which by
In an analogous manner, removal of the definition eliminates all function of the
clutch will produce a car that cannot move, encoded protein, should eliminate circa-
but the motor will still work. Just as one dian rhythms. The standard way of deter-
cannot conclude that a clutch is a compo- mining whether a mutation is a null is by
nent of the motor because the car will not determining whether it produces the
move when it is taken out, one cannot con- same result in complementation tests as a
clude that a gene is essential for the central deletion of the gene. For example, the
circadian oscillator because it can produce per0 mutation in Drosophila has been
an arrhythmic output when mutated. In determined to be a null because it uncovers
fact, some of the circadian mutants affect a short-period, long-period or arrhythmic
just one rhythmic output of the clock and phenotypes when heterozygous with a
can therefore be assigned to a specific short-period, long-period, or arrhythmic
output pathway. per allele, just as a deletion of per
Mutations that produce arrhythmia in all does when heterozygous with the same
known circadian outputs are more likely to alleles. Although a null mutation in a clock
affect the central oscillator mechanism, but gene is expected to produce arrhythmia, it
they could also affect an output pathway should be noted that in mammals, redun-
that is common to all known circadian dant or compensatory mechanisms may
outputs. For instance, if a circadian oscilla- result in less severe phenotypes (e.g., period
tor communicated temporal information to alterations).
RHY2 2/6/04 3:51 PM Page 21

MOLECULAR ANALYSIS OF CIRCADIAN GENES 21

The final determination of how a muta- short sequence run can be compared with
tion affects circadian rhythms must await existing databases to extract the sequence
isolation of the gene affected. At that point, of the full-length gene. While it is still
the spatiotemporal expression profile, pos- important to sequence the clones isolated,
sible interactions with known clock pro- because there are frequently errors in the
teins, and the biochemical function of the database sequence, the availability of this
protein all contribute toward determining information allows researchers to rapidly
its precise role in the circadian system. start designing experiments based on the
sequence. The sequence may predict a
biochemical function—For instance, it may
䊏 MOLECULAR ANALYSIS OF contain domains found in kinases or tran-
CIRCADIAN GENES scription factors, and/or it may reveal the
presence of a PAS (PER, aryl hydrocarbon
The methods used to isolate the gene receptor of mammals, and single-minded
corresponding to a mutant phenotype gene in Drosophila) domain, which is a
vary somewhat from species to species. The protein–protein interaction domain found
chromosomal location of the mutation, in many clock proteins. Data obtained from
and the proximity of genetic/molecular other assays (described below) can be
markers, can also influence the methodol- coupled with the sequence information to
ogy and ease of mapping it and isolating the address the role of the candidate gene. For
relevant gene. Thus, these methods are not instance, if the original mutant affects
discussed at length here, although details expression of per and the gene encodes a
for individual genes can be found in subse- transcription factor, then it is worth deter-
quent chapters. Once the gene is isolated, mining if per is a direct transcriptional
it is analyzed through a standard series target. The DNA clones can be used to
of procedures outlined in Figure 2.1 and express the proteins in bacteria or tissue
described below. It is important to note, culture cells, so that large quantities of the
though, that the approaches discussed here proteins can be isolated and used for bio-
are based on what we know of existing chemical studies (see text below).
clock genes. When the first circadian gene, The recombinant proteins produced in
which was the Drosophila period (per) bacteria or cultured cells are also injected
gene, was being analyzed, there was no into mice, rats, or rabbits to produce anti-
clear course of action. It was the analysis of bodies that specifically bind to the proteins.
per that paved the way for future molecu- The DNA clones provide probes to analyze
lar studies of circadian rhythms. Expression RNA, while the antibodies can be used to
studies of per pinpointed the clock cells in detect protein. Using the cloned genes and
Drosophila, and the discovery of per cycling antibodies as probes, one can ask when and
and feedback regulation led to the current where the genes are expressed, and how the
model for clock function. functions of the multiple clock genes inter-
act to produce circadian rhythms.
Gene Sequencing
Cell Culture
The sequence may predict the biochemical
function of the protein. Once a gene is iso- Cell culture systems can be used to study
lated, it is sequenced—these days quite protein function and are becoming
rapidly by automated sequencing facilities. increasingly popular for the study of circa-
Now that entire genomes of many organ- dian rhythms. As will be discussed in
isms have been sequenced, the results of a Chapter 4, mammalian cell lines can be
RHY2 2/6/04 3:51 PM Page 22

22 GENETIC AND MOLECULAR APPROACHES USED TO ANALYZE RHYTHMS

induced to display a cycling circadian gene observed, even if the gene is a direct target
expression, which can also be reset by of the transcription factor, because another
chemical/humoral signals. Although this is factor is also required.
obviously a very useful and easily manipu-
lated system, some caution is required
Temporal Analysis
because the physiological effects as well as
some of the interactions in these cells may Knowing, as we do now, that levels of many
not be relevant for clock function in the clock and clock-controlled genes cycle, it
intact organism. However, regardless of follows that every new potential circadian
their limitations for understanding clock gene is assayed for temporal regulation.
physiology in whole organisms, cell culture Tissues are collected at different times of
systems are valuable for assaying biochem- day, and RNA levels are assayed through
ical properties of candidate proteins. For northern blot analysis, RNase protection
instance, phosphorylation assays and tran- assays (Fig. 2.3), or quantitative polymerase
scription assays are effectively performed chain reaction (PCR) experiments. Like-
in cultured cells. wise protein expression is assayed in tissue
If the RNA levels of the candidate gene obtained at different times of day, usually
are found to cycle (see text below), it is through western blot analysis (Fig. 2.4). The
worth investigating the promoter region western blots will frequently also reveal
of the gene in question. Transcription changes in the mobility (i.e., the size of the
factors involved in clock function have protein) over the course of the day. For
been identified, and the promoter sites many clock proteins these mobility changes
recognized by many of these factors are are known to be due to cyclic phosphoryla-
known. The presence of such a recognition tion events, which are part of a mechanism
site in the promoter region of the gene of regulating clock protein levels and function.
interest would suggest that is likely to be a A key point to remember with all of
direct target of a clock transcription factor. these studies is that while oscillations
Alternatively, as mentioned above, the of RNA, protein, or phosphorylation levels
candidate gene may encode a transcription may be first assayed in the presence of
factor that affects expression of a known light–dark cycles, they are circadian-
clock gene. Transcriptional effects can relevant only if they also occur in freerun-
be tested experimentally by fusing the ning conditions (e.g., constant darkness and
relevant promoter to a reporter gene constant temperature). The reason for also
such as bacterial b-galactosidase or firefly assaying them in the presence of light–dark
luciferase. Neither of these genes is present cycles is because molecular oscillations
endogenously in the cell culture system, frequently dampen in freerun. Thus, if the
and neither would show any regulation by assays were done only in freerun, the low
circadian transcription factors unless a cir- amplitude of some oscillations could result
cadian promoter is fused to the reporter in a failure to detect any cycling.
gene. Activation/repression of the reporter
by the candidate transcription factor then
Spatial Analysis
establishes the role of the transcription
factor and the target promoter sequence. In situ experiments using either labeled
Typically, these experiments are done in RNA probes or antibodies can be used to
cultured cells transfected with the relevant identify the sites of expression. In these
constructs (Fig. 2.2). It should be noted, experiments, the RNA or protein is
though, that sometimes activation is not detected in tissue sections or whole mounts
RHY2 2/6/04 3:51 PM Page 23

MOLECULAR ANALYSIS OF CIRCADIAN GENES 23

Transfected cells

(a) prom X is an
luciferase Luciferase activity
activator of
(as compared to baseline) prom

(b) prom X is a
luciferase Luciferase activity
repressor of
(as compared to activator prom
alone)

Activator
X
Prom- target promoter
X-candidate transcription factor

Figure 2.2. Use of cell culture assays to determine the effects of candidate genes on tran-
scription. As described in the text, the promoter of the putative target gene is fused to a
reporter and transfected into cultured cells. The reporter shown here is the firefly luciferase
protein whose activity can be reliably and easily quantified in a luminometer. Luciferase activ-
ity is assayed in cells cotransfected with the candidate transcription factor as well as in control
cells (these may be cotransfected with a transcription factor that is not relevant for this
process). The level of luciferase activity in control cells serves as a baseline. If the candidate
factor is suspected of being a repressor, its effects are best studied under conditions where
the target promoter is actively transcribed. Multiple transcription factors may be transfected
to determine how the presence of one affects the activity of the other.

of biological specimens, such that the gene circuits, that are part of the input or output
product is visualized along with the cellular pathways of the circadian system.
and subcellular structure of the specimen.
Thus, these types of experiments tell us not
Subcellular Localization
only whether and when a gene is expressed,
but precisely where it is expressed. If a gene The subcellular localization may cycle over
is expressed in clock cells it becomes a can- the course of a day. It has become clear
didate for a clock gene, although it could through analysis of several clock genes that
turn out to be a component of the input or the subcellular localization—in particular
the output pathway. Conversely, if it is not the expression in nuclei—is an important,
expressed in clock cells, then it almost cer- regulated process. Although temporal
tainly is not a clock gene. Its localization control of nuclear expression has not been
may then serve to identify the cells, or the demonstrated in all cases, the fact that
RHY2 2/6/04 3:51 PM Page 24

24 GENETIC AND MOLECULAR APPROACHES USED TO ANALYZE RHYTHMS

Zeitgeber
z time zeitgeber time
8 12 20 2
4 8 12 16 20 24
mPert 1
PER

Tubulin
Figure 2.4. Temporal analysis of protein
expression by western blot analysis. The data
Figure 2.3. Temporal analysis of RNA shown are for the Drosophila period protein
expression by RNase protection assay (RPA). (PER). Adult Drosophila were entrained to a
The data shown here are for the mPer1 gene light–dark cycle for several days, after which
in the mouse kidney. Mice were entrained to they were collected at the timepoints
light–dark cycles for several days and then indicated [in zeitgeber time (ZT)]. Heads
collected and sacrificed at the times indicated were isolated and homogenized to prepare
(recall that ZT0 = lights on and ZT12 = lights protein extracts. The extracts were run on
off). Kidneys were isolated and homoge- SDS polyacrylamide gels, which separate
nized. RNA was extracted for each timepoint denatured proteins based on their size. The
and subjected to the analysis shown here. proteins were then transferred to mem-
The analysis involves the synthesis of a radi- branes (“western-blotted”) and probed with
olabeled antisense RNA probe from the gene an anti-PER antibody. The antibody binds
being studied and then the hybridization of to the membrane only where PER antigen
this probe to total RNA from the relevant is present, and the presence of the bound
tissue. The mix is then treated with RNases antibody is detected indirectly by coupling
that digest all the single-stranded RNAs an enzyme, which emits a luminescent
present. RNA that corresponds to the gene signal assayed through autoradiography. As
under study is hybridized to the probe, and evident from the figure, levels of PER cycle
thus the radiolabeled probe is “protected” over the course of the day. The phosphory-
from digestion. Protected fragments are visu- lation state of PER also cycles. The high-
alized on a denaturing gel. Typically, the molecular-weight bands seen at ZT2
same RNA samples are also assayed for an represent phosphorylated forms of PER.
RNA that is expected to be constant in
all samples. This serves to control for the
amount of RNA in each sample. The control
RNA assayed in the experiment shown here Assaying Expression in
is that of the tubulin gene. A radiolabeled Mutant Backgrounds
antisense probe complementary to the
tubulin mRNA was added to the sample at Now that multiple clock genes and other
the same time as the mPer1 probe. As is components of the circadian system are
evident, levels of tubulin are constant at all known, there is a framework in which new
times of day while levels of mPer1 cycle. components can be placed. In order to iden-
tify their relative placement or role, it is
important to determine their relationship
multiple mechanisms are employed to reg- to known components. One way of doing
ulate the expression of some clock proteins this is to look for physical interactions,
in nuclei makes it likely that nuclear either through yeast two-hybrid systems or
import/export occurs in a temporal fashion. coimmunoprecipitation assays (explained
Thus, when determining the spatial distri- below). However, it is sometimes difficult to
bution of a new protein, it is also desirable predict the putative partners, and in many
to determine its subcellular localization cases the interactions are weak and unde-
and, if possible, determine if the protein tectable in these assays. The other way to go
moves from one compartment to another about addressing this question is to use the
within the cell over the course of a day. genetic tools available, namely, the existing
RHY2 2/6/04 3:51 PM Page 25

MOLECULAR ANALYSIS OF CIRCADIAN GENES 25

clock mutants. By assaying the expression scription of clock gene B, then a null mutant
of the new gene in different genetic back- of A will express high levels of the mRNA
grounds, one can determine the order of for B, while overexpression of A from a
gene action in the mechanism. For instance, transgene will produce low levels of the
if a mutation in A alters the expression of mRNA for B. Overexpression of a gene
B, but a mutation in B does not alter the that encodes cycling RNA and/or protein
expression of A, then B is downstream of A will sometimes eliminate cyclic expression
(i.e., B is regulated by A, and B does not of the gene product(s). This approach can
regulate A). Expression of known clock thus be used to test the importance of the
genes should also be assayed in the new cycling of that gene product to the overt
mutant—obviously this can be done even rhythm. If output rhythms are disrupted
before the affected gene is cloned. Effects under conditions that eliminate RNA/
on expression can range from altered levels protein cycling of a specific gene, then most
and disrupted cycling to changes in the likely cyclic expression of the gene in ques-
subcellular distribution. Interpretations of tion is required for a rhythmic output.
these findings must take the cyclic nature of Tissue specific expression of a transgene
the circadian process into account. For can be achieved by fusing the gene to a
instance, low levels of a clock protein that promoter that is expressed exclusively in
negatively regulates its own transcription specific cell types in an organism that is
could result from a regulatory mutation null for the endogenous copy of the gene.
that affects gene expression, but they could This allows one to assesses the effect of the
also arise from a mutation that reduces sta- clock gene in that specific tissue, without
bility of the clock protein. In the latter the confounding effects of expression in
event, RNA levels would be expected to be other tissues. For instance, expression of the
high due to reduced negative feedback. Drosophila per gene exclusively in several
central brain neurons produces circadian
activity rhythms in a per null mutant fly,
Transgenic Clock Gene Expression
which has arrhythmic activity without
Expression of transgenic clock genes in expression of the transgene. This experi-
organisms allows analysis of circadian clock ment supports the view that these cells are
mechanisms at the molecular and whole the site of the clock controlling circadian
organism level. In all the model organisms activity. In several organisms (e.g., Neu-
employed by chronobiologists, techniques rospora and mice), it is possible to replace
for the introduction of exogenous genes the endogenous gene with a nonfunctional
(i.e., transgenes) have been developed. transgenic copy, thereby producing a null
These techniques allow the insertion of mutant. This is an important accomplish-
“designer” genes created by the experi- ment if classical forward genetics has not
menter into the genomes of the organism. produced a null mutation for the gene.
The types of experiments that transgenic Another important approach is the
approaches allow are too numerous to com- analysis of transgenes with genetically
pletely describe here, but a few general engineered mutations in particular regions
approaches are worth mentioning. Overex- of the protein coding region or the pro-
pression of a clock protein can be accom- moter. These mutations may abolish some
plished by fusing the gene to a promoter particular aspect of the gene’s proper func-
that is highly expressed in a particular tion, such as circadian regulation of its
tissue. This produces a genotype that is expression or its protein–protein interac-
complementary to the null genotype. For tion with another clock protein. Such a
instance, if clock gene A represses the tran- result establishes the importance of differ-
RHY2 2/6/04 3:51 PM Page 26

26 GENETIC AND MOLECULAR APPROACHES USED TO ANALYZE RHYTHMS

ent protein and promoter domains for new proteins that interact with a protein
specific biochemical functions. In the case of interest. Typically, a cDNA encoding
of promoter analysis, reporter genes are the protein of interest (“bait”) is fused to
usually employed, for the reasons described sequences encoding the DNA binding
above. Some reporter genes (such as domain of a transcription factor and
luciferase) can be imaged in living organ- expressed in yeast. The protein with
isms and allow circadian regulation of pro- which interaction is being tested (“prey”) is
moters to be followed for many days in a fused to an activation domain that cannot
single organism. While analysis of mutant by itself associate with the DNA binding
promoters and genes can be pursued in protein fused to the bait. A functional tran-
vitro or cell culture, transgenic organisms scription factor is generated only when the
allow the effects on whole organism circa- two protein domains (the bait and the prey)
dian physiology to be assessed. interact and bring the DNA binding and
activation domains together. The functional
transcription factor is detected on the basis
䊏 OTHER METHODS OF ISOLATING of its ability to bind and activate its target
CIRCADIAN GENES promoter sequence, which is fused to a
reporter gene. The reporter typically pro-
Homology to Known Clock Genes in duces a colored product or a product nec-
Other Organisms essary for survival of the yeast. For a
The mammalian period (per) genes were random screen, a library is generated in
isolated or identified on the basis of their which each cDNA is fused to the activation
homology to Drosophila per. Conversely, domain of the transcription factor. Identifi-
the isolation of the Drosophila Clock gene cation of interacting proteins is based on
was facilitated by the previous existence the color assay or the ability of the yeast to
of the mammalian Clock gene. Thus, this grow on a particular nutrient-deficient
cross-species approach is clearly a powerful medium (the nutrient is produced only
method to identify circadian-relevant when a functional transcription factor is
genes. Until recently, polymerase chain generated).
reaction (PCR) technology was used to It is also possible to analyze protein–
amplify homologous sequences and protein interactions more directly with bio-
thereby isolate a gene from another species. chemical assays that can be applied to pro-
However, as the genomes of an increasing teins expressed in vitro, in bacteria, in cell
number of organisms are being sequenced, culture or in vivo. For instance, an antibody
the most efficient method is to scan com- which recognizes one clock protein can
puter databases of genomic sequence be used to “immunoprecipitate” that clock
for the sequence of interest. This area of protein by linking the antibody to a
biology, termed bioinformatics, is now as bead, which settles out of solution; the
important to molecular biologists as are bead brings with it the clock protein recog-
standard “bench” experiments. nized by the antibody, and (importantly)
any other proteins that form stable
protein/protein interactions with the clock
Interaction with Known Clock Proteins
protein. The recovery of these other pro-
The most common method for this purpose teins with the beads shows that these
is the yeast two-hybrid assay (Fig. 2.5). proteins interact with the clock protein.
Using this assay, one can either test for an Another approach requires the fusion of a
interaction between two known proteins or short ligand binding domain to one clock
else conduct a random screen to identify protein. For instance, the protein glu-
RHY2 2/6/04 3:51 PM Page 27

OTHER METHODS OF ISOLATING CIRCADIAN GENES 27

Inter. Activating

DNA
binding
b-galactosidase Blue color

DNA
binding
b-galactosidase

Figure 2.5. Identification of interacting proteins through the yeast two-hybrid assay. The
protein of interest (protein X) is fused to the DNA binding domain of a transcription factor.
An entire library of cDNA sequences is fused to a domain that is capable of activating tran-
scription (“Activating”). Transcription of the reporter occurs only when the DNA binding and
activating domains are brought together through the interaction of protein X with a protein
in the library (“Inter.”). Transcription of the reporter produces a protein that either allows
growth of the yeast on specific media or renders a blue color to the yeast colony. In the
example shown here, the reporter is b-galactosidase, which confers a blue color.

tathione-S-transferase (GST), which binds genomics technology (described below),


glutathione tightly, can be fused to a clock the most popular methods for such screens
protein. The clock protein will now be were subtractive hybdridization and differ-
retained on beads which are coated with ential display. Both methods involve the
glutathione, thereby allowing the recovery isolation of RNA at different times of day
of the clock protein along with the beads. If and the synthesis of the corresponding
another protein that is not fused to GST cDNAs from these RNAs. In subtractive
is also recovered on the beads, a hybridization, the cDNAs from one time of
protein–protein interaction with the clock day are hybridized to the cDNAs from
protein is demonstrated. another time of day and those that fail
to hybridize, because they are not present
at the other time of day, are cloned. These
Assays for Circadian Regulation represent RNAs that are expressed pre-
dominantly at a specific time(s) of day.
Differential Display/Subtractive
Differential display experiments are based
Hybridization Experiments
on the same principle, but consist of “dis-
The finding that expression of many circa- playing” all the RNA products expressed at
dian genes cycles with a ~24-hour period a specific time of day and comparing those
has prompted researchers to use this prop- with ones expressed at a different time of
erty to identify new circadian genes. In day. In practice, radiolabeled cDNAs are
principle, the approach consists of search- synthesized from each RNA population
ing for RNAs whose levels vary at different using random primers, and these are then
times of day. Prior to the development of separated on a denaturing gel. Radio-
RHY2 2/6/04 3:51 PM Page 28

28 GENETIC AND MOLECULAR APPROACHES USED TO ANALYZE RHYTHMS

labeled “bands” that are found in RNA is altered in the course of a biological/
samples extracted at some times of day, but pathological process. Thus, it can be used to
not at others are cut out of the gel and identify genes that are up- or downregu-
cloned. This technology has been refined in lated when cells become cancerous, when a
different ways to reduce the frequency of particular behavioral state is induced or
false positives and has been quite success- when a specific stimulus is introduced.
ful in identifying cycling genes. From the circadian point of view, it is the
variation of gene expression over the
course of a day–night cycle that is of inter-
Presence of Circadian Elements
est, as well as the effects of various clock
in the Promoter
mutations on gene expression. Conse-
As noted in Chapter 1 and elsewhere in this quently, this technology has been used to
book, some of the known clock proteins identify cycling genes in several systems.
are transcription factors that bind to spe- The example described below (see also Fig.
cific DNA sequences. These sequences, 2.6) describes the isolation of circadian
commonly termed “consensus sites,” can be regulated genes from Drosophila heads.
used to indicate whether the gene is likely On the basis of the known genome
to be clock-controlled. Now that the sequence, it is possible to photochemically
genomes of many different organisms have
been sequenced, one can, in theory, scan
databases of genomic sequence to identify Entrain organism to light-dark cycles
putative clock-controlled genes. The caveat
is these consensus sites are usually quite
frequent and, in many cases, nonfunctional. Transfer to constant darkness
Sometimes they are functional, but not
under control of the clock, as there are non-
clock transcription factors that recognize
Collect samples at different times of day
the same sequences. It has become clear
that regions flanking the consensus sites
can play a role in determining specificity
and functionality. However, until specific
sequences or structures are known for these Extract RNA
flanking regions, it is difficult to distinguish
a clock-controlled consensus site from a
non-clock-controlled one.
Subtractive Differential Microarrays
hybridization display
Microarrays
This is yet another example of the applica- Figure 2.6. Isolation of circadian genes
tions of genomics technology. As men- through molecular screens for time-of-day-
specific expression. Three different ap-
tioned above, there has been considerable proaches that have been successfully used to
emphasis on sequencing entire genomes as isolate genes whose expression changes over
well as large numbers of randomly selected the course of a day are shown. With the
cDNAs from several organisms. As a result, advent of microarrays, the other two
there is a wealth of sequence information methods have largely been phased out,
although they may still be useful in organ-
that can be exploited for high-throughput isms where microarrays have not been gen-
analysis. The goal of this type of analysis is erated because of lack of sufficient genomic
to identify all the genes whose regulation information.
RHY2 2/6/04 3:51 PM Page 29

FURTHER READING 29

synthesize short oligonucleotides comple- FURTHER READING


mentary to unique regions of all the pre-
dicted 13,600 genes in the Drosophila Ewer, J, Frisch, B, Hamblen-Coyle, MJ, Rosbash,
genome.The DNA for all of these genes can M, Hall, JC (1992): Expression of the period
be fixed as spots on glass slides; the small clock gene within different cell types in the
size of the spots and the precision of the brain of Drosophila adults and mosaic analy-
arraying process allow spots for all of the sis of these cells’ influence on circadian
predicted Drosophila genes to be arrayed behavioral rhythms. J Neurosci 12:
on a small glass slide (a “microarray” or 3321–3349.
Frisch, B, Hardin, PE, Hamblen-Coyle, MJ,
“gene chip”). These slides are hybridized
Rosbash, M, Hall, JC (1994): A promoterless
with fluorescently labeled nucleic acids period gene mediates behavioral rhythmicity
synthesized from mRNAs collected at and cyclical per expression in a restricted
different timepoints. The amount of subset of the Drosophila nervous system.
fluorescently labeled nucleic acid that Neuron 12: 555–570.
hybridizes to a particular spot is propor- Gekakis, N, Saez, L, Delahaye-Brown, AM,
tional to the amount of mRNA for that Myers, MP, Sehgal, A, Young, MW, Weitz, CJ
gene in the total mRNA, and is determined (1995): Isolation of timeless by PER protein
by measuring the fluorescent intensity of interaction: Defective interaction between
the spot after hybridization. Hybridizations timeless protein and long-period mutant
of fluorescently labeled nucleic acids pre- PERL. Science 270: 811–815.
Harmer, SL, Hogenesch, JB, Straume, M, Chang,
pared from different circadian timepoints
HS, Han, B, Zhu, T, Wang, X, Kreps, JA, Kay,
track the changing patterns of gene expres- SA (2000): Orchestrated transcription of key
sion on a genomic scale. pathways in Arabidopsis by the circadian
Its is clear that the molecular analysis of clock. Science 290: 2110–2113.
circadian rhythms has advanced to a point Konopka, RJ, Benzer, S (1971): Clock mutants of
where it lends itself to the most sophisti- Drosophila melanogaster. Proc Natl Acad Sci
cated, cutting-edge technology. In addition (USA) 68: 2112–2116.
to the approaches described here, which are Kornmann, B, Preitner, N, Rifat, D, Fleury-Olela,
common to most organisms, other elegant F, Schibler, U (2001): Analysis of circadian
methods have been used in specific organ- liver gene expression by ADDER, a highly
isms. For instance, in both Drosophila sensitive method for the display of differen-
tially expressed mRNAs. Nucleic Acids Rese.
and mammals, chimeric animals (mosaics)
29: E51–51.
have been made in which some cells of Loros, JJ, Denome, SA, Dunlap, JC (1989):
the animal are mutant for a particular Molecular cloning of genes under control of
clock gene and others are wild type. In the circadian clock in Neurospora. Science
Drosophila, this approach helped to iden- 243: 385–388.
tify the central clock cells in the Drosophila Low-Zeddies, SS, Takahashi, JS (2001): Chimera
brain, because circadian activity rhythms analysis of the Clock mutation in mice shows
are only altered by the mutation if it is that complex cellular integration determines
present in these central clock cells. In circadian behavior. Cell 105: 25–42.
mammals, the study of chimeric animals Siwicki, KK, Eastman, C, Petersen, G, Rosbash,
showed that properties of circadian M, Hall, JC (1988): Antibodies to the period
gene product of Drosophila reveal diverse
rhythms such as periodicity, rhythm
tissue distribution and rhythmic changes in
strength, and phase are conferred by dif- the visual system. Neuron 1: 141–150.
ferent, overlapping subsets of cells in the Welsh, S, Kay, SA (1997): Reporter gene expres-
SCN. Thus, a combination of approaches is sion for monitoring gene transfer. Curr Opin
being successfully used to further our Biotechnol 8: 617–622.
understanding of clock function.
RHY3 2/6/04 3:52 PM Page 31

Part II
MOLECULAR CONTROL
OF CIRCADIAN RHYTHMS:
ANIMAL MODELS
RHY3 2/6/04 3:52 PM Page 33

3
DROSOPHILA MELANOGASTER:
A MODEL SYSTEM FOR
MOLECULAR CHRONOBIOLOGY
Jeffrey L. Price

䊏 AN INTRODUCTION AND the genes affected by Drosophila clock


HISTORICAL PERSPECTIVE mutations. This combination of phenomeno-
logical, genetic, and molecular genetic
The demonstration of a circadian rhythm in approaches in Drosophila has led to a virtual
Drosophila eclosion by Kalmus and Bunning explosion in our knowledge of clock mecha-
in the 1930s initiated the development of nisms. Fortunately, the mammalian clock
Drosophila into one of the primary model employs many of the same genes as the
systems for chronobiologists. Drosophila Drosophila clock, so the models derived for
pseudoobscura was employed by Colin Pit- the Drosophila clock have been readily
tendrigh to explore the formal properties of applied to the mammalian clock. Studies of
phase resetting and to formulate the general the mammalian clock have become a potent
circadian principle of temperature compen- force in their own right, allowing a synergy
sation in the 1950s (see Chapter 1). In 1971, that has facilitated work in both Drosophila
Ronald Konopka and Seymour Benzer iso- and mammals.
lated the first single gene “clock” mutants in
Drosophila melanogaster, thereby initiating
Circadian Rhythms of Drosophila
genetic analysis of the clock mechanism.With
the advent of Drosophila molecular biology The circadian rhythm demonstrated by
in the 1970s, it became possible to isolate Kalmus and Bunning in the 1930s is called

Molecular Biology of Circadian Rhythms. Edited by Amita Sehgal


ISBN 0-471-41824-2 Copyright © 2004 John Wiley & Sons, Inc.

33
RHY3 2/6/04 3:52 PM Page 34

34 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

the “eclosion rhythm.” The term “eclosion” cycles. Drosophila rest is associated with
refers to the emergence of the fly from the prolonged immobility, a characteristic
pupal case at the end of its metamorphosis posture, and unresponsiveness to sensory
from the larval to adult stage. Eclosion is stimuli, as is mammalian sleep. Another
said to be “gated” by the clock, because a similarity is that flies respond homeostati-
fly that is developmentally ready to eclose cally to disrupted rest with an increased
during the late day or night nevertheless tendency to rest. This response is affected
refrains from eclosing until the next circa- by several clock mutations. Drosophila rest
dian “gate,” which occurs during an 8–10- is suppressed by exposure to caffeine and
hour interval beginning 1–2 hr before dawn. enhanced by cyclohexyladenosine and
While an individual fly ecloses only once hyrdoxyzine, which have similar effects on
in its lifetime, a population of flies with a sleep in mammals. Finally, wakefulness
broad distribution of developmental stages in Drosophila is associated with elevated
produces recurrent eclosion activity around levels of several mRNAs that are also ele-
the time of dawn in a light–dark cycle (LD), vated in the rat cerebral cortex during
or at the time of subjective dawn in con- wakefulness in the rat, and the capacity to
stant darkness (DD). Eclosion activity is tolerate sleep deprivation is increased by
defined as the number of flies emerging in expression of heat shock genes, which also
fixed intervals (e.g., hourly intervals), and confer resistance to other stresses such as
the numbers for successive intervals are heat shock. Hence, a genetic and molecular
plotted as a function of collection time. analysis of “sleep–wake” in Drosophila
Such plots show a circadian rhythm, in may yield insights into the mechanisms and
which the peaks of eclosion are approxi- function of human sleep, in addition to the
mately 24 hours apart in wild-type flies role of circadian rhythms in the regulation
entrained to light–dark cycles (LD) and of this process.
then transferred to constant darkness (DD)
(Fig. 3.1a). Circadian control of eclosion
Drosophila Clock Proteins
is mediated by at least four hormones
(ecdysone, ecdysis triggering hormone, These proteins are expressed in the central
eclosion hormone, and crustacean car- brain and in peripheral oscillators. A group
dioactive peptide). of adult brain neurons contains a circadian
With the advent of electronic data oscillator that controls locomotor activity
collection systems, locomotor behavior and eclosion. However, there is evidence
became the most widely assayed circadian for circadian clock control of olfaction,
behavior in Drosophila. In this assay, indi- retinal physiology, response to starvation,
vidual flies are placed in glass tubes, and responsiveness to dopamine, sensitization
their activity is detected as deflections of an to cocaine, sensitivity to ethanol, ovarian
infrared light beam that is passed through diapause, Malpighian tubules (the fruitfly
the tube. Actograms, in which activity is kidney), and the gut in Drosophila
plotted as a function of time, show a circa- melanogaster. Initially, the broad tissue dis-
dian rhythm.The flies are active at the times tribution of clock gene expression was sur-
of lights on and off in LD, and during prising, as the head had been shown to be
the subjective day in DD (Fig. 3.1b). Super- the relevant site for regulation of loco-
ficially, these records resemble the motor activity, and it was thought that
sleep/wake records of mammals. clocks would be found exclusively in the
It has been shown that the Drosophila nervous systems of higher organisms. We
activity rhythm has more than just a super- now know that the circadian oscillator is
ficial resemblance to human sleep–wake largely cell autonomous, and that many of
RHY3 2/6/04 3:52 PM Page 35

Figure 3.1. Examples of eclosion (a) and locomotor activity (b) in wild-type and per mutant flies. The flies had previ-
ously been entrained to L : D 12 hours : 12 hours but are monitored here in DD. For A, the number of flies emerging from
pupae every hour is plotted as a function of time, while in B an activity event for an individual fly is recorded as a deflec-
tion of a chart recorder pen. The activity in B is double-plotted on a timescale that approximates the period of the
endogenous clock, so that active periods occur at approximately the same time in successive circadian days. In the arrhyth-
mic mutant fly, there are no organized bouts of activity. [Parts (a) and (b) both reprinted with permission from Konopka

35
and Benzer (1971).]
RHY3 2/6/04 3:52 PM Page 36

36 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

the physiological processes listed above Single-Gene Clock Mutations


are controlled by peripheral and nonneural
In 1971 Konopka and Benzer were the first
clocks, which are dispersed throughout the
to isolate single-gene clock mutations in any
body. These peripheral clocks are discussed
organism.The original three mutations were
in more detail in Chapter 9.
isolated by testing fly lines, each of which
There are no documented circadian
contained a mutagenized sex (X) chromo-
behaviors or physiological responses in
some, for aberrant eclosion. Three lines
Drosophila larvae, but circadian rhythms in
eclosed abnormally. When these three lines
gene expression do occur in several small
were entrained to LD and then subjected to
groups of neurons, and are affected by the
hourly collections in DD, one exhibited a
various clock mutations, as are the circa-
short-period profile, one a long-period
dian rhythms of adults. One of these groups
profile, and one an arrhythmic eclosion
of neurons is thought to survive metamor-
profile (Fig. 3.1a). Their locomotor activity
phosis and ultimately to form some of
rhythms were affected in the same way (Fig.
the adult neurons that control circadian
3.1b). Genetic tests showed that all of these
rhythms of locomotor activity and eclosion.
mutations affected the same gene, which
This group is likely to mediate larval “time
was dubbed period (or per, for brevity). In
memory,” in which locomotor and eclosion
accordance with the considerations outlined
rhythms of adults kept in DD can be
in Chapter 2, the centrality of the per gene
entrained by prior exposure to LD during
to the clock oscillator mechanism was sug-
the larval stage.
gested by the effects of the mutants on both
behavioral outputs of the clock (eclosion
and locomotor activity) and the isolation of
䊏 GENETIC ANALYSIS OF CIRCADIAN
long-period, short-period, and arrhythmic
RHYTHMS IN DROSOPHILA alleles. The short-period allele was dubbed
perS, the long-period allele perL, and the
We begin our study of the Drosophila clock
arrhythmic allele per0.
mechanism by considering the classi-
cal genetic analysis of the period (per)
Locomotor Activity Rhythms Produced
gene. This work illuminated features of the
in the Head
tissue and cellular locus of the circadian
clock controlling locomotor and eclosion The per gene functions primarily in a group
behavior. Then, we consider the molecular of neurons within the head to produce
genetic studies of per and other genes locomotor activity rhythms. In the first
constituting the core oscillator mechanism. experiment, the head of a perS fly was trans-
Next, we consider the mechanism of the planted into the abdomen of an arrhythmic
input or entrainment pathways, and finally per0 fly (Fig. 3.2a). The transplantation
we consider the mechanisms for coupling resulted in a short-period circadian rhythm of
the clock to outputs such as locomotor locomotor activity, corresponding with the
activity. genotype of the transplanted head, in a fly


Figure 3.2. Experiments that established that per expression in the lateral neurons is neces-
sary and sufficient for locomotor activity rhythms: (a) transplantation of a perS head into the
abdomen of a per0 fly produces perS locomotor activity; (b) gynandromorph flies with perS
male heads(s) exhibit a fully mutant (19-hour) rhythm, while flies with perS/per+ (s/+) female
heads and perS male bodies exhibit a perS/per+ rhythm (21.5 hours); (c) in mutant and trans-
genic flies with altered per expression in the head, rhythmic locomotor activity results only if
per is expressed in the lateral neurons.
RHY3 2/6/04 3:52 PM Page 37

GENETIC ANALYSIS OF CIRCADIAN RHYTHMS IN DROSOPHILA 37


RHY3 2/6/04 3:52 PM Page 38

38 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

that would otherwise have been arrhythmic. brain locus that did not require per expres-
These results established that expression of sion in the eyes or optic lobes.
the per mutant gene in the head was sufficient Later, it became possible to produce
to produce the phenotype. In addition, they transgenic flies in which genetically engi-
suggested that the head produced a diffusible neered per genes were placed in the
factor that could drive rhythmic locomotor genome of flies. Some of these allowed
activity, since the transplanted head was the expression of per only in cells that
unlikely to generate axonal projections to have been termed the “lateral neurons,”
other neurons or the thoracic muscles con- because they are found at the side of the
trolling the legs. central brain, where it is joined to the optic
Complementary experiments demon- lobe medulla. Expression of per in these
strated the expression of the per mutation cells alone rescued the locomotor arrhyth-
in the head was necessary and sufficient micity of flies that were otherwise per0,
for the mutant phenotype (Fig. 3.2b). In because their endogenous gene was per0
females that were heterozygous for perS or (Fig. 3.2c). Therefore, expression of per in
per0 (per+/per0 or per+/perS), loss of the X the lateral neurons is sufficient for circadian
chromosome carrying the per+ allele was rhythms of locomotor activity, and expres-
induced in some of the embryonic cells, sion of per is not required in the eyes and
which then carried only the mutant allele optic lobes.
(perS or per0) and became male cells (X0 is In addition, analyses of various muta-
male in Drosophila). A cell that had lost the tions affecting adult eye development
per+ X divided to produce a patch of male showed that the lateral neurons are neces-
tissue, while patches in which the per+ X had sary for circadian rhythms of locomotor
not been lost remained female. Flies gener- activity, while the eyes and optic lobes are
ated in this way are part male and part dispensable (Fig. 3.2c). Most such muta-
female and are termed gynandromorphs. tions have no effects or only minor effects
In these cases, the female patches were on circadian locomotor activity. Even
per+/perS or per+/per0, while the male mutants that are missing the compound
patches were perS or per0. Since the per eyes entirely can still be entrained by light
mutant chromosome also carried another and have wild-type periods. However, the
mutation (e.g., a recessive body color disconnected mutation (disco), which dis-
mutation), male patches could be identified rupts the connection of the compound eye
in the adult fly because they exhibited to the optic lobes, also produces a high fre-
this mutant phenotype (e.g., mutant body quency of locomotor arrhythmia. In these
color). Tests of locomotor activity revealed flies, identifiable lateral neurons are almost
whether a particular gynandromorph had a always absent, but per expression persists in
fully mutant circadian rhythm (19 hours the eyes and other sites of expression.
or arrhythmic) or one consistent with a Therefore, per must be expressed in the
heterozygous genotype (21.5 hours for lateral neurons for circadian locomotor
perS/per+ or 24 hours for per0/per+). The vast activity.
majority of flies with fully mutant rhythms
had male patches in the head, demon-
Molecular Cloning
strating that the per mutant allele must be
expressed in the head to affect period Classical genetic analysis of per localized
length or rhythmicity. Furthermore, it was the gene for molecular cloning. With the
shown that small groups of cells in the brain advent of molecular genetic techniques, it
were sufficient to confer the fully mutant became possible to clone genes affected by
phenotype, suggesting a discrete central mutations, thereby leading to a quantum
RHY3 2/6/04 3:52 PM Page 39

CIRCADIAN OSCILLATIONS OF period AND timeless GENE PRODUCTS 39

leap in our understanding of many genes. mulate in per0 flies. Presumably, the per0
The intense genetic analysis of Drosophila translation product is not stable enough
during the twentieth century had produced to accumulate, and the per0 mutation is
high-resolution genetic maps of all the described as a null mutation, meaning
Drosophila chromosomes, and per was that it lacks all gene activity. By contrast,
located in particularly well mapped region the perS and perL genes are still functional
between the y and w genes on the X chro- enough to support rhythmicity, albeit with
mosome. Two groups successfully cloned altered period length.
the per gene. The laboratories of Jeffrey
Hall and Michael Rosbash accomplished
this by cloning microdissected DNA from 䊏 CIRCADIAN OSCILLATIONS OF
the region of the X chromosome that con- period AND timeless GENE
tained per, while the laboratory of Michael PRODUCTS
Young “walked” to per from a previously
cloned region by successive isolation of Circadian oscillations of these genes are
overlapping clones. generated by feedback of period and time-
Ultimately, introduction of per trans- less proteins on their gene products.
genes into per0 flies identified the per tran-
scription unit. In Drosophila, genes that are
flanked by sequences from a class of trans- PER Protein Versus per mRNA
posable elements known as “P elements” The circadian oscillation of per protein
can integrate into the genomic DNA of the (PER) lags the oscillation of per mRNA by
fly, thereby producing a transgenic fly car- about 6 hours. Since per was the first clock
rying a gene chosen by the experimenter. It gene to be cloned, it was the first clock
was shown that introduction of one gene gene for which circadian oscillations were
from the genetic interval known to contain demonstrated. per mRNA and protein
per could produce wild-type circadian were shown to oscillate with a circadian
behavior in a fly that contained only the rhythm (Fig. 3.3) in all tissues except the
per0 mutation at the endogenous gene ovary. These oscillations continued in DD
locus. Therefore, this gene was in fact per. and were affected by the various per muta-
Transgenes with different amounts of DNA tions in the same way that circadian behav-
upstream of the coding region of per ior was affected. perS flies had short-period
expressed different amounts of per mRNA. molecular oscillations, perL flies had long-
Those transgenes expressing higher levels period molecular oscillations, and per0 flies
of per had shorter periods, while longer did not produce the molecular oscillations.
periods resulted from lower levels of per A transgene expressing wild-type per
expression. protein (PER) could rescue rhythmic oscil-
The mutant perS, perL, and per0 genes lations of the endogenous per0 mRNA,
were sequenced and found to differ at since per0 mRNA, which is constitutively
single nucleotides from the wild-type gene. expressed in the per0 mutant without a
In the perS and perL mutant genes, the transgene, exhibited circadian oscillations
nucleotide changes produced a single in a per0 fly that also contained a wild-type
amino acid change, while the per0 mutation per transgene. Thus, PER protein was
produced a translational stop codon in the required for a feedback regulation of its
amino-terminal part of the reading frame. own gene products. Because of their per-
At best, the per0 gene could encode only a sistence in DD and the effects of the
truncated per protein, but even a truncated mutants on the period of both behavior and
per protein has never been seen to accu- the molecular oscillations, the resulting
RHY3 2/6/04 3:52 PM Page 40

40 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

molecular oscillations of the per gene prod- 2. Why does per mRNA begin to fall
ucts were thought likely to be part of when high levels of PER protein have
the long-sought circadian oscillator accumulated?
mechanism.
Intriguingly, the phases of the per mRNA We will address the second question first,
and protein rhythms were quite distinct (Fig. and return to the first question in a later
3.3); PER protein reaches its peak about 6 section.
hours after the peak in per mRNA, and per
mRNA levels are already decreasing at this
PER Transcription
time. PER protein levels are lowest when
per mRNA levels are starting to accumulate. PER negatively regulates its own transcrip-
The interesting phase relationship posed tion. In all tissues except the ovary, in which
two immediate questions: per expression is not rhythmic, PER accu-
mulates in nuclei with a circadian rhythm.
1. What posttranscriptional control per mRNA levels begin to fall when PER
mechanism results in the long lag becomes strongly nuclear, and begin to
between per mRNA accumulation rise after nuclear PER levels have declined
and PER protein accumulation? during the day. This phase relationship of
Relative abundance

Eye RNA

Brain RNA

Eye protein

Brain protein

Zeitgeber time
Figure 3.3. per mRNA and protein levels exhibit circadian oscillations in both the eyes and
brain. The mRNA oscillation was detected with an RNAse protection assay, in which a radioac-
tively labeled per probe is hybridized to Drosophila RNA. The protein oscillation was detected
with an immunoblot analysis. [Reprinted from Zeng et al. (1994): Constitutive overexpression
of the Drosophila period protein inhibits period mRNA cycling. EMBO J 13(15): 3590–3598 by
permission of Oxford University Press.]
RHY3 2/6/04 3:52 PM Page 41

CIRCADIAN OSCILLATIONS OF period AND timeless GENE PRODUCTS 41

per mRNA and nuclear PER protein sug- per mRNA stability both contribute to per
gested that PER protein might negatively mRNA oscillations, in addition to the tran-
regulate transcription of its own mRNA. scriptional effects that are mediated by the
It was possible to test the hypothesis that upstream promoter.
PER negatively regulates its own expres-
sion by generating a line of flies in which
The PAS Domain
PER was constitutively expressed at high
levels in the eye (Fig. 3.4a). The coding PER contains a protein–protein interaction
region for the per gene was placed down- region called the PAS domain. Initially, the
stream of the rhodopsin promoter, which conceptual translation of per’s nucleotide
produced constitutively high levels of eye- sequence did not provide much informa-
specific per mRNA and protein. As a result, tion about the function of PER protein.
the oscillation of per mRNA expressed Subsequent to the cloning of per, the con-
from the endogenous gene in the eye was ceptual translations of several genes cloned
suppressed (Fig. 3.4a, lower panel), and the from Drosophila and mammals showed
level of expression was similar to the levels weak homology to a region of per that was
expressed at the trough of the wild-type termed the “PAS” region, after the three
cycle. These results were consistent with an proteins in which the homology was first
intracellular negative feedback mechanism, observed (PER, the aryl hydrocarbon
since the endogenous per gene was receptor of mammals, and the single-
repressed only in the eyes. Molecular oscil- minded gene in Drosophila). Most of these
lations persisted in other tissues (e.g., the other genes with a PAS region also have a
brain; Fig. 3.4a, top panel), as did circadian basic helix–loop-helix (bHLH) domain,
rhythms of locomotor activity, which do not which identifies them as members of a
derive from the eye (see discussion above). bHLH/PAS transcription factor family,
The effects of PER on its mRNA were several of whose members are molecular
shown to be mostly transcriptional because components of the circadian oscillator in
transgenes containing the per promoter Drosophila and mammals (Fig. 3.5a). PER
fused with the bacterial b-galactosidase, does not contain a bHLH domain, which is
chloramphenicol acetyl transferase (CAT), necessary to bind DNA (Fig. 3.5b). There-
or luciferase (luc) produced circadian oscil- fore, it cannot bind DNA directly. However,
lation of the reporter gene, which lacked biochemical experiments have established
any part of per except the regions that the PAS region as a protein–protein inter-
could bind transcription factors (Fig. 3.4b, action domain, suggesting that PER may
top panel). Rhythmic transcription of the bind to other proteins. Further genetic
reporter gene required PER, because the analysis in Drosophila identified other
oscillation of the reporter gene did not factors that do bind PER to produce post-
occur in a per0 fly. The phase of the oscilla- translational control of PER protein and
tion produced by the per promoter did not the transcriptional control of per mRNA.
precisely match the phase of endogenous
per (Fig. 3.4b, top), while a reporter gene
tim Gene Products
that contained enough per mRNA
sequence as well as the reporter sequence The timeless (tim) gene products also oscil-
did oscillate in phase with endogenous per late. Like the per gene, tim, the second
mRNA (Fig. 3.4b, bottom). An extensive Drosophila clock gene shown to be part of
analysis of this difference has shown that the circadian oscillator was identified in a
regulatory sequences within the transcribed screen for recessive mutants that affected
part of per and a circadian regulation of eclosion. The first tim mutant produced
RHY3 2/6/04 3:52 PM Page 42

42 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

Figure 3.4. The production of per mRNA oscillations involves intracellular negative feedback
by PER protein, transcriptional regulation of the per promoter, and posttranscriptional effects
on per mRNA. (a) per mRNA oscillations are observed in both the eyes and brains of wild-
type flies (CS), while flies in which PER is overexpressed from the eye-specific rhodopsin pro-
moter exhibit per mRNA oscillations in the brain but not the eye. In the eyes of these
overexpressing flies, the endogenous per mRNA, which is quantitated here instead of the
transgenic mRNA, is expressed at constitutively low levels. [Reprinted from Zeng et al. (1994):
Constitutive overexpression of the Drosophila period protein inhibits period mRNA cycling.
EMBO J 13(15): 3590–3598 by permission of Oxford University Press]. (b) Transgenic reporter
genes driven by the per promoter were introduced into flies expressing wild-type PER. The
levels of the BS-CAT reporter mRNA, which does not contain any per mRNA sequences but
does contain the promoter regions that are responsive to transcriptional regulation, oscillate
with a circadian rhythm that differs from the oscillation of per mRNA, while a BG-luc reporter
mRNA containing the 5¢ part of the per mRNA oscillates in phase with per mRNA, presum-
ably because it is regulated both transcriptionally and posttranscriptionally. [Reprinted from
Stanewsky et al. (1997): Multiple circadian-regulated elements contribute to cycling period
gene expression in Drosophila. EMBO J 16(16): 5006–5018 by permission of Oxford University
Press.]
RHY3 2/6/04 3:52 PM Page 43

CIRCADIAN OSCILLATIONS OF period AND timeless GENE PRODUCTS 43

Figure 3.5. Several clock proteins found in both Drosophila and mammals contain PAS
domains. (a) Alignment of similar PAS proteins from mammals, fruitflies, and silkmoths. The
PAS region, which can mediate protein/protein interactions, contains short regions that are
almost direct copies of each other [(a) and (b)]. PER also contains other regions that are con-
served in mouse (m) and silkmoth (ap) PER (% identities shown). CLOCK and CYCLE contain
basic helix–loop-helix (bHLH) regions that bind to DNA. CLOCK also contains a polyglutamine-
rich (polyQ) C terminus, which is likely to activate RNA polymerase at the promoter to which
the CLOCK/CYC heterodimer binds. The mutations in the Drosophila and mouse Clk genes are
found in a polyglutamine region, while the Drosophila cyc0 mutation occurs in the PAS region.
[Top, reprinted from Shearman et al. (1997): Two period homologs: circadian expression and
photic regulation in the suprachiasmatic nuclei. Neuron 19: 1261–1269, copyright (1997) with
permission from Elsevier Science. Middle, reprinted from Allada et al. (1998), copyright (1998)
with permission from Elsevier Science. Bottom, reprinted from Rutila et al. (1998): CYCLE is
a second bHLH-PAS clock protein essential for circadian rhythmicity and transcription
of Drosophila period and timeless. Cell 93: 805–813, copyright (1998) with permission from
Elsevier Science.] (b) bHLH transcription factors generally bind as dimers consisting of two
different peptides (e.g., CLK and CYC). The dimers are formed by protein–protein interac-
tions involving the two pairs of alpha helices, while E-box elements in the DNA are bound
by the basic regions of the heterodimer.

arrhythmic locomotor activity and eclosion. have been identified. tim mRNA and
This mutant (tim0) is the result of a deletion protein undergo circadian oscillations with
in the middle of the tim coding region. phases quite similar to those of per gene
The deletion apparently produces a “null” products (Fig. 3.3). In particular, there is
mutant with no evidence of residual func- also the same lag between the tim mRNA
tion. In addition, tim mutants that shorten and protein oscillations. tim and per
circadian period, lengthen circadian period, mutants affect these oscillations just as they
and suppress some of the perL phenotype affect behavior.
RHY3 2/6/04 3:52 PM Page 44

44 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

PER/TIM Collaboration be necessary for stabilizing PER in the


cytoplasm and producing its nuclear accu-
The timeless protein (TIM) associates with mulation and oscillations.
PER to produce post-translational control of A complementary analysis of the effects
PER levels and nuclear accumulation of a of per0 mutants on the tim gene products
PER/TIM complex. With the isolation of the (Fig. 3.6b) showed that per was important
tim0 mutant, it was now possible to investi- for the circadian regulation of tim. tim
gate the effects of one mutation (tim0 or per0) RNA does not oscillate in a per0 mutant in
on the expression of the other gene products either LD or DD. While TIM protein levels
(per or tim, respectively). The results were oscillate in LD in the per0 mutant, TIM is
quite striking. In the tim0 mutant (Fig. 3.6a), expressed at constitutively high levels in
per mRNA levels did not oscillate in a circa- DD in the per0 mutant, in contrast to the
dian manner and were expressed at levels persistent oscillation observed in wild-type
approximating the median of the wild-type flies. These high levels of TIM accumulate
cycle. Even more striking were the effects on only in the cytoplasm, so PER is required
PER protein. The levels of PER detected by for nuclear accumulation and circadian
immunoblot analysis were constitutively oscillations of TIM, just as TIM is required
low—equivalent to the levels found at the for these features of PER’s regulation.
trough of the wild-type cycle. So the presence However, since TIM can accumulate to
of TIM somehow stabilized PER in the wild- high levels in the dark in the per0 mutant,
type flies. PER is not required to stabilize TIM, while
Additional insight was gained by exam- TIM is required to stabilize PER.
ining the effects of the tim0 mutation on The findings suggested that both the sta-
reporter transgenes, in which bacterial bility and nuclear translocation of PER and
b-galactosidase was fused to the first half of TIM might be regulated by the circadian
per and expressed under the control of the clock. Because the oscillations of TIM
per promoter. This protein was expressed at levels could be driven by LD in the absence
high and equivalent levels in both wild of a tim mRNA oscillations, the stability of
type and tim0 flies, because the fusion of TIM was also suggested to be regulated
b-galactosidase to PER somehow stabilized by light (as we shall see, the regulation of
the remaining parts of PER to the proteol- TIM by light is part of the entrainment
ysis that acted on PER itself in the tim0 pathway). Consistent with a regulation of
mutant. More importantly, the subcellular nuclear translocation, PER and TIM have
localization of the reporter protein (PER- been seen to accumulate in the cytoplasm
bGAL) was altered in the tim0 mutants. of the lateral neurons prior to nuclear accu-
PER-bGAL accumulated in the nuclei of mulation during the circadian cycle.
wild-type flies but was exclusively cytoplas- PER and TIM interact directly through
mic in the tim0 mutant. The effect of tim0 domains that include nuclear and cytoplas-
was not a general one for all nuclear pro- mic localization signals. In addition to its
teins, because nuclear accumulation of isolation through positional cloning, the tim
other nuclear proteins was normal in tim0 gene was identified on the basis of its inter-
flies. In fact, the deletion of a region of PER action with PER. This was in the course of
containing the PAS region of homology and a yeast two-hybrid screen designed to look
another interaction sequence [the cytoplas- for PER-interacting proteins. Consistent
mic localization domain (CLD); see text with the idea of a direct intracellular inter-
below] produced a b-galactosidase reporter action of the two proteins, the tim gene
fusion protein that localized to nuclei even products were expressed in the head in the
in tim0 flies. In summary, TIM was shown to same places where per was seen. In fact, two
RHY3 2/6/04 3:52 PM Page 45

CIRCADIAN OSCILLATIONS OF period AND timeless GENE PRODUCTS 45

(a)
o

per mRNA expression


in LD

PER protein expression


in LD
tim o
Wild type

PER-b GAL
subcellular localization
Figure 3.6. PER and TIM regulate each other transcriptionally and posttranscriptionally. (a)
per expression in wild-type (left) and tim0 mutant (right) heads. per mRNA (analyzed by RNAse
protection assay) did not exhibit circadian oscillations in tim0 flies. PER proten (analyzed by
immunoblot analysis, in which the bound anti-PER antibody results in the emission of a lumi-
nescent signal) was expressed at constitutively low levels in tim0 flies (nonsp—nonspecific
crossreacting band). The low levels of PER preclude its detection in situ, so a PER-bGAL
reporter, which accumulates to high levels in tim0 flies, was detected instead. It is expressed
in the same tissues as in wild-type flies but fails to localize to nuclei (arrowheads). The nuclei
are best visialized in the retina where they appear as a layer. [Top, reprinted with permission
from Sehgal et al. (1994): Loss of circadian behavioral rhythms and per RNA oscillations in the
Drosophila mutant timeless. Science 263: 1603–1606, copyright (1994) American Association
for the Advancement of Science. Middle, reprinted from Price et al. (1995) with permission
of Oxford University Press. Bottom, reprinted with permission from Vosshall et al. (1994), copy-
right (1994) American Association for the Advancement of Science.]
RHY3 2/6/04 3:52 PM Page 46

46 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

(b) Wild type per o

tim mRNA expression


in LD

TIM protein expression


in LD

TIM protein expression


in DD

TIM subcellular
localization
Figure 3.6. Continued. (b) tim expression in wild-type (left) and per0 mutant (right) heads.
Detection was accomplished as in (a). The per0 mutation eliminates the circadian oscillations of
tim gene products and the nuclear accumulation of TIM, but it does not eliminate LD-driven
cycles of TIM protein or generally depress TIM protein levels. [Top, reprinted with permission
from Sehgal et al. (1995): Rhythmic expression of timeless: A basis for promoting circadian cycles
in period gene autoregulation. Science 270: 808–810, copyright (1995) American Association
for the Advancement of Science. Middle, reprinted by permission from Zeng et al. (1996): A
light-entrainment mechanism for the Drosophila circadian clock. Nature 380: 129–135, copy-
right (1996) Macmillan Publishers Ltd. Bottom, reprinted from Myers et al. (1996): Light-induced
degradation of TIMELESS and entrainment of the Drosophila circadian clock. Science 271:
1736–1740, copyright (1996) American Association for the Advancement of Science.]
RHY3 2/6/04 3:52 PM Page 47

dCLOCK AND CYCLE 47

that TIM and PER coregulate their subcel-


lular localization by direct binding of
regions that regulate subcellular localiza-
tion, thereby masking the cytoplasmic
localization domains and unmasking
nuclear localization signals in both proteins.
This interaction is also likely to stabilize
PER by protecting it against proteolytic
Figure 3.6. Continued. (c) The mutual regu- degradation in the cytoplasm. Once in the
lation of per and tim involves protein/protein nucleus, PER and TIM negatively regulate
interactions between PER and TIM. Both pro- their own promoters, in a manner that we
teins contain putative nuclear localization
signals (NLSs) thought to mediate nuclear shall examine in the next section.
import. In addition, both proteins contain
cytoplasmic localization domains (CLDs),
which retain PER (or TIM) in the cytoplasm 䊏 dCLOCK AND CYCLE
unless the other is coexpressed. PER also con-
tains the previously described PAS region
[black bars denote (a) and (b) repeats]. Inter- These circadian oscillator elements form
actions between PER and TIM occur as shown. a transcription factor that positively regu-
[Reprinted from Saez and Young (1996), lates the per and tim promoters, negatively
copyright (1996) with permission from Else- regulates dClock, and is inhibited by bind-
vier Science.] ing of PER and TIM.

distinct interaction domains exist within


The E Box
both PER and TIM (Fig. 3.6c). Within PER,
one of these was the PAS region, and the dClock (dClk) and cycle (Cyc) encode basic
other was termed a cytoplasmic localization helix-loop helix (bHLH)/PAS transcription
domain (CLD) because it also functions to factors that bind to an “E” box within the
retain PER in the cytoplasm in the absence per and tim promoters. Several research
of TIM. Deletion of this CLD causes PER directions converged to elucidate a role for
to move to the nucleus of Drosophila tissue these two genes in the Drosophila circadian
culture cells even without TIM, while PER clock. The Clk gene was identified first in
which includes the CLD will move into the mice from studies of the first circadian
nucleus only if TIM is coexpressed. Fur- mouse mutant to be isolated (also called
thermore, the perL mutation, which lies in Clk; see Chapter 4). The conceptual trans-
the PAS region of per, reduces the interac- lation of the mouse Clk gene showed that
tion of PER and TIM and delays the it encoded a basic helix–loop helix tran-
nuclear localization of PERL at high tem- scription (bHLH) factor that also con-
perature, thereby further establishing the tained a PAS region of homology (Fig. 3.5).
link between PER/TIM association and As discussed above, the PAS region also is
nuclear localization. found in PER and has been shown to
Within TIM, one of the interaction mediate protein–protein associations. The
domains includes its nuclear localization bHLH domain exists in a large number
signal, which interacts with the PER PAS of transcription factors, in which the basic
domain. The other interaction domain in (b) region binds to a short six-nucleotide
TIM interacts with the CLD of PER. TIM sequence (CACGTG; the E box) within the
also has a cytoplasmic localization domain, promoters of certain genes. Binding of
which retains TIM in the cytoplasm unless the basic regions usually requires bHLH
PER binds to TIM. All these data argue factors to dimerize with each other. The
RHY3 2/6/04 3:52 PM Page 48

48 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

dimerization is mediated through a region dCLK proteins were shown to associate


containing two alpha helices connected by with each other both in vitro and in vivo,
a loop (the HLH region; Fig. 3.5). Because thereby forming a heterodimer (Fig. 3.7a).
mouse Clk protein (CLK) also contained a Although all bHLH transcription factors
PAS region, it seemed possible that it might bind to an E-box-containing DNA se-
interact with a mouse PER via the PAS quence, a particular bHLH heterodimer
region, and that the Drosophila circadian will recognize only the subset of E-box
clock might also utilize a dCLK protein. sequences that have a particular DNA
Both predictions would be validated, but sequence in the flanking region. Both
here we will concentrate on the Drosophila the per and tim promoters have E-box
Clk protein (dCLK; see Chapter 4 for more sequences in them, and an E-box sequence
on the mammalian clock genes). in the per promoter has been shown to be
Attempts to clone a Drosophila Clk necessary for normal circadian oscillations
gene on the basis of homology to the mouse of per mRNA.
gene coincided with attempts to clone the So an obvious hypothesis to test was
genes affected by two clock mutations in whether this particular E box was a target
Drosophila. Both of these mutations cause for the dCLK/CYC heterodimer. This was
arrhythmic circadian behavior when tested by introducing a luciferase reporter
homozygous. The Jrk mutation turned out gene under the control of the per or tim E
to affect the Drosophila version (ortholog) box into a Drosophila cell line, along with
of the mouse Clk gene, and so it was DNA that expressed dCLK. Luciferase is
renamed dClkJrk. The other mutation (cyc0) the enzyme that produces flashes of light in
also affected a bHLH/PAS transcription fireflies, and it can produce a glow in trans-
factor protein (CYC; Fig. 3.5a). CYC and genic fruit fly cells, too. Since CYC was


Figure 3.7. dCLOCK and CYCLE associate to form a transcription factor that positively regu-
lates the per and tim promoters by binding to them, and this binding is negatively regulated
by PER and TIM. (a) A yeast two-hybrid assay, in which dCLOCK and CYC (or dBMAL1) are
shown to associate. dCLOCK is fused to a DNA binding domain (LEXA), while dBMAL1 is fused
to a domain that activates RNA polymerase (VP16). Transcription of a b-galactosidase (b-Gal)
gene can only occur if a protein/protein association between dCLK and dBMAL occurs. Because
b-GAL cleaves a substance in the food to produce a blue color, transcription of the b-Gal gene
(and therefore a protein–protein interaction) is demonstrated in yeast colonies that turn blue
(seen as darker colored colonies here). (b) dCLK/CYC activates transcription of per through
the per E box, and PER and TIM inhibit this activation. Introduction of a dCLK expressing trans-
gene into a cell line that already expresses CYC activates expression of a reporter gene driven
by the per E box. Mutation of the E box or introduction of both PER and TIM expressing trans-
genes reduces the activation. Activation is measured by the light emission that is catalyzed
by luciferase. [Parts (a) and (b) reprinted with permission from Darlington et al. (1998), copy-
right (1998) American Association for the Advancement of Science.] (c) dCLK is required for
activation of the per promoter in vivo. A luciferase reporter gene is rhythmically expressed
from the per promoter in wild-type or +/dClkJrk flies but it is constitutively expressed at low
levels in a homozyogus dClkJrk mutant. [Reprinted permission from Allada et al. (1998), copy-
right (1998) with permission from Elsevier Science.] (d) dCLK/CYC binds to the E box in vitro,
and this binding is reduced by PER, TIM, or PER/TIM. The indicated proteins are added to the
radioactively labeled E-box sequence. The addition of dCLK and CYC reduces the mobility of
the radioactively labeled DNA on a gel, thereby producing a band. Although PER and TIM do
not produce novel mobility shifts (E1 and E2 are produced by contaminating proteins in the
assay), they do reduce the amount of dCLK/CYC-dependent complex. [Reprinted from Lee et
al. (1999) with permission from the American Society for Microbiology.]
(a) (b)
dCLOCK dBMAL VP16

LEXA
RHY3 2/6/04 3:52 PM Page 49

promoter b-Gal TIM PER

dBMAL dCLOCK
light

E box promoter luciferase

(c) light (d)


dBMAL dCLOCK (d)

PER TIM
E box promoter luciferase dCLOCK dBMAL

E box
dCLOCK dBMAL

E box E box

49
RHY3 2/6/04 3:52 PM Page 50

50 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

expressed from the endogenous cyc gene of binding to dCLK/CYC and preventing
the cells, the dClk transgene resulted in a them from binding to the per and tim genes.
dCLK/CYC heterodimer, which caused a However, other data do not support this
significant increase in the luciferase “glow” mechanism in its entirety, as incubation of
of the cell line (Fig. 3.7b). Since single Drosophila head extracts with DNA con-
nucleotide changes in the per E-box taining a tim E box produces comparable
sequence eliminated the stimulation of amounts of a bound dCLK/CYC at all
luciferase by dCLK, the effect of dCLK was times of day. The high levels of PER and
shown to be mediated by the E box. TIM bound with dCLK/CYC in head
Another line of evidence showing that extracts isolated from night timepoints
dCLK and CYC regulated per and tim tran- would be predicted to reduce the amount
scription comes from assays of endogenous of complex formed during the night.
per and tim levels and transcription rates in Since both PER and TIM bind to
the dClkJrk and cyc0 mutants. These are dCLK/CYC in vivo, it is not clear what the
constitutively low in the mutants (e.g., Fig. relative contributions of PER, TIM, and
3.7c), as expected for mutations that elimi- PER/TIM are to negative feedback in vivo.
nate positively acting transcription factors In vitro, either PER or TIM alone can
for the per and tim promoters. Finally, inhibit binding of dCLK/CYC to the E box
when purified dCLK and CYC proteins are (Fig. 3.7d). In vivo, nuclear PER persists
mixed together and added to E-box DNA after TIM is largely eliminated by light, and
in vitro, they can be shown to bind to the negative feedback also persists for much of
DNA (Fig. 3.7d). this time. Moreover, in the timUL mutant,
elimination of nuclear TIMUL protein by
light enhances repression of per and tim
PER/TIM–dCLK/CYC Binding
mRNA, demonstrating that PER alone is
PER and TIM bind to the dCLK/CYC het- a more effective negative regulator than is
erodimer and abrogate its E-box binding. PER/TIMUL. In tissue culture cells, mutant
If PER and TIM are expressed in the PER missing a CLD can transport to nuclei
Drosophila cell line along with dCLK, and repress the per promoter in the absence
endogenously expressed CYC and the per of TIM.
E-box-driven luciferase, the levels of
luciferase are reduced relative to its expres-
Interlocked Feedback Loops
sion in the absence of PER and TIM (Fig.
3.7b). Coexpression of both PER and TIM The Drosophila clock consists of inter-
is required for this negative feedback. One locked feedback loops. The levels of dClk
reason for this requirement is that neither mRNA oscillate with a circadian rhythm
PER nor TIM is nuclear unless both are that has a phase opposite that of per and tim
expressed. In vivo, the binding of PER and mRNA (Fig. 3.8a). There is almost no phase
TIM with dCLK occurs during the night lag between dClk mRNA and protein, both
but not during the latter part of the day— of which peak in abundance around the
consistent with the times at which per and time of dawn, while per and tim mRNA
tim transcription are inhibited. In vitro, levels peak at the time of dusk. The time at
this binding abrogates the binding of which dClk mRNA peaks is also the time at
dCLK/CYC to the per promoter (Fig. 3.7d) which PER and TIM are most strongly
without disrupting the dCLK/CYC inter- localized to nuclei, suggesting that PER
action. Therefore, these data argue that and TIM might positively regulate dClk
PER and TIM exert negative feedback by mRNA. Consistent with this notion, dClk
RHY3 2/6/04 3:52 PM Page 51

dCLOCK AND CYCLE 51

(a)

per
(b)

tim

Wild type
Midday Late night
(c)

CYC dCLK
Zeltgeber (hours)

Figure 3.8. per and tim positively regulate dClk mRNA levels and negatively regulate per
and tim mRNAs—most likely in both cases through a negative regulation of dCLK/CYC. (a)
The dClock mRNA oscillates with a phase opposite to the per and tim mRNA oscillations. The
upper panel is an autoradiograph of a gel demonstrating these oscillations. The radioactive
band is produced by hybridization of the indicated mRNA to a complementary radioactively
labeled RNA produced from the cloned gene. The hybridization event protects the radioac-
tive “probe” from digestion by RNAse, and the amount of protected probe is therefore a
measure of the complementary mRNA present. In the lower panel, the amount of radioac-
tivity for each band has been normalized to the amount of radioactivity for a constitutively
expressed mRNA (RP49, also visualized on the gel). [Reprinted from Bae et al. (1998): Circa-
dian regulation of a Drosophila homolog of the mammalian Clock gene: PER and TIM func-
tion as positive regulators. Mol Cell Biol 18: 6142–6151 with permission from the American
Society for Microbiology.] (b) dClk mRNA is constitutively expressed at low levels in the per0
mutant and at high levels in the dClkJrk, cyc0. per0; dClkJrk, and per0; cyc0 mutants. dClk mRNA
levels were quantitated as in (a) from early evening time points (black bars) and early sub-
jective day timepoints (light bars). (c) Model suggesting how negative regulation of dCLK/CYC
by PER/TIM can produce oppositely phased oscillations by interlocked feedback loops. The
large circles represent the nucleus. [Parts (b) and (c) reprinted with permission from Glossop
et al. (1999), copyright (1999) American Association for the Advancement of Science.]

51
RHY3 2/6/04 3:52 PM Page 52

52 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

mRNA and protein are expressed at con- but the function of this phosphorylation
stitutively low levels in per0 and tim0 flies, is still unknown. In the case of PER and
which do not express any functional PER TIM, maximal levels of phosphorylation
or TIM (respectively; e.g., Fig. 3.8b). are observed before the proteins disappear,
How might PER and TIM produce such suggesting that phosphorylation targets
“positive feedback?” An important insight them for degradation. Genetic analysis has
comes from the observation that dClk identified kinases that are involved in the
mRNA levels are constitutively high in phosphorylation of both these proteins, and
dClkjrk or cyc0 single mutants, and also in has elucidated some of the roles for this
double mutants such as per0; dClkJrk and phosphorylation.
per0; cyc0 (Fig. 3.8b). Taken together with
the opposite effects of per0 or tim0 mutants
on dClk mRNA, these results argue that
The dbt Gene Product
dCLK/CYC negatively regulates the
mRNA levels of dClk, and that PER and The double-time (dbt) gene product regu-
TIM operate upstream of dCLK/CYC to lates the stability and nuclear accumulation
eliminate this negative feedback. The elim- of PER protein. PER becomes progres-
ination of the two sequentially acting sively phosphorylated after its synthesis
negative feedbacks produces a positive during late night, and it becomes maximally
effect on dClk in the double mutants (Fig. phosphorylated during morning, when high
3.8b). It is not known how PER and TIM levels of nuclear PER are also detected.
abrogate negative feedback by dCLK/ Then, levels of PER begin to decline.
CYC, but they are likely to do so in the However, the role of this phosphorylation
same way they abrogate positive feedback was not known until the isolation of the dbt
by CLK/CYC—by binding to the mutants allowed the Drosophila clock to be
CLK/CYC heterodimer (Fig. 3.8c). It is now studied under conditions in which the phos-
known how dCLK/CYC negatively regu- phorylation of PER is altered. dbt muta-
lates dClk mRNA (Fig. 3.8c). It does so tions that shorten, lengthen, or eliminate
indirectly by activating a repressor of the circadian rhythmicity have all been identi-
dClk promoter—vrille (to be discussed in a fied. The mutation which shortens circadian
later section). period expedites both the phosphorylation
of PER and its subsequent disappearance.
The mutations that lengthen the circadian
䊏 POSTTRANSLATIONAL period delay or prolong PER phosphoryla-
REGULATION OF CIRCADIAN tion, and result in persistently high levels of
OSCILLATOR PROTEINS BY PER after TIM disappears in the morning.
PHOSPHORYLATION Two dbt mutations produce molecular
and/or behavioral arrhythmicity. One of
The function of several circadian oscillator these is also a pupal lethal. Both produce
proteins is regulated posttranslationally by constitutively high levels of PER whose
phosphorylation. In addition to rhythms of phosphorylation state is constitutive (either
level and nuclear accumulation, PER and constitutively low or constitutively hetero-
TIM are phosphorylated rhythmically. This geneous). So there is a correlation between
rhythm is observed as a change in the the timing or amount of PER phosphoryla-
mobility of PER and TIM on SDS (sodium tion and the turnover of PER in the dbt
dodecyl sulfate)–polyacrylamide gels (e.g., mutants. This correlation is consistent with
Figs. 3.6a and 3.6b, middle panels). In addi- a role for phosphorylation in triggering the
tion, the dCLK protein is phosphorylated, degradation of PER.
RHY3 2/6/04 3:52 PM Page 53

POSTTRANSLATIONAL REGULATION OF CIRCADIAN OSCILLATOR PROTEINS BY PHOSPHORYLATION 53

Casein Kinase I bility of mammalian PER in tissue culture


cells (see Chapter 4). Moreover, there are
dbt encodes an ortholog of the vertebrate many potential casein kinase I phosphory-
e/d casein kinase I isoforms. This type of lation sites in both mammalian and
protein kinase is thought to phosphorylate Drosophila PER, and sequential phospho-
threonines and serines. While many protein rylation of these may explain the progres-
kinases have to be activated by the binding sive phosphorylation of PER that is
of a ligand to a receptor or by a second observed during the circadian cycle.
messenger [e.g., Ca2+, diacylglycerol or In the case of two of the three long-
cyclic adenosine monophosphate (cAMP)], period dbt mutants and the arrhythmic
casein kinase I has not been shown to be mutant that also produces pupal lethality,
directly activated by this type of mecha- it is clear that the mutations reduce the
nism. Instead, it prefers to phosphorylate kinase activity of DBT. The two long-
substrates that have already been phospho- period dbt mutations, which change two
rylated on a threonine, serine, or tyrosine amino acids in DBT, have been introduced
one to two amino acids upstream from the into the yeast homologue, where they
target serine or threonine. So dbt protein reduce the activity of the kinase in vitro.
(DBT) may be activated to phosphorylate The lethal dbt allele (dbtP) arises from
PER by its own prior kinase activity on insertion of a P transposable element,
PER, or by the activity of other, messenger- which strongly reduces the level of dbt
dependent kinases on PER. It has been mRNA and produces hypophosphorylated
shown that DBT can bind with PER in vitro PER in vivo. Therefore, lowered levels of
and in cell culture, and this interaction is DBT activity can reduce or delay phospho-
likely to enhance the targeting of PER as rylation of PER and lead to elevated or
a substrate. dbt mRNA and protein are persistent expression of PER.
expressed at the same sites as per mRNA in
the adult head, so DBT is expressed in a
PER in Cytoplasm
manner that allows it to undergo direct
interactions with PER. Wild-type DBT acts to destabilize PER in
It is certain that the DBT kinase has the cytoplasm. The effects of altering PER
many other substrates in addition to PER, phosphorylation are seen most clearly in the
since many mutations in this gene are P-element-induced mutant of dbt, which
lethal, with effects on cell survival and strongly depresses DBT expression and
growth in imaginal discs (these have been causes pupal lethality. As mentioned above,
dubbed “discs overgrown” or “dco” muta- PER is constitutively hypophosphorylated
tions). In vertebrates and yeast, casein in this mutant (Fig. 3.9a). Since this mutant
kinase I has been shown to be involved in also results in pupal lethality, adult circadian
processes as diverse as DNA repair, cell behaviors cannot be assayed. However, it is
cycle progression, vesicle trafficking, Wnt possible to assay the rhythms of per in the
signaling (a pathway important for devel- larval lateral neurons, which mediate
opment), and circadian rhythms. the larval time memory discussed earlier. In
So far, it has not been possible to the lateral neurons of this mutant, PER
produce enzymatically active recombinant protein is expressed at constitutively high
DBT protein, so a demonstration that DBT levels in both DD and LD, rather than with
directly phosphorylates PER has not been the oscillations that are found in wild-type
possible in flies. However, casein kinase I larvae (Fig. 3.9a). Moreover, PER is seen in
has been shown to bind and phosphorylate parts of the larval central nervous system
mammalian PER, and to decrease the sta- where it is not normally detected (Fig. 3.9b).
RHY3 2/6/04 3:52 PM Page 54

54 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

The overexpression is not the result of high


levels or otherwise aberrant per mRNA
expression, because the levels and sites of
per mRNA expression appear to be normal
in the mutant. For instance, a reporter gene
under the control of the per promoter
expresses its mRNA in tissues where per
PER protein levels are higher in dbt P larvae mRNA is also expressed. The resulting
than in wild type larvae, and PER is not highly
phosphorylated in dbt P larvae
PER-bGAL fusion protein, which is more
(a) stable than PER itself, accumulates in
many sites where PER is not observed in
wild type, as does PER in dbtP (Fig. 3.9b).
Therefore, the dramatic accumulation of
PER in dbtP larvae is most likely to arise
posttranscriptionally.
In fact, it seems most likely that the
observed hypophosphorylation of PER
produces the high levels of PER in this dbt
Like PER-bGAL (SG3) but unlike PER (WT) in wild type mutant, in sites where TIM expression is
larvae, PER in dbt P larvae is expressed in parts of the not readily detected. In the tim0 mutant,
central nervous system where TIM and PER do not
accumulate in wild type larvae recall that PER is cytoplasmic and
(b) expressed at very low levels. Apparently,
PER does not require high levels of TIM
Figure 3.9. Low levels of dbt expression for its stability in the absence of DBT and
lead to high levels of constitutively expressed in the absence of wild-type phosphoryla-
PER that is hypophosphorylated. (a) Extracts tion of PER. This hypothesis was confirmed
of larvae or adults collected at the indicated by subjecting larvae to constant light (LL),
timepoints were immunoblotted with an which eliminates PER in wild-type larvae
antibody for PER, which generated a lumi-
nescent band. PER is much more highly by destabilizing TIM. In the dbt mutant,
expressed in dbtP larvae than in wild-type PER levels were unaffected by LL, thereby
larvae; per0 larvae do not express PER and establishing that PER stability is largely
therefore serve as a negative control. The TIM-independent. It seems that wild-type
mobility of PER in dbtP is comparable to that DBT protein is likely to destabilize PER in
of PER in adults at ZT14, when PER is not
extensively phosphorylated. (b) Immunolo- the cytoplasm unless high levels of TIM
calization of PER in the central nervous have accumulated. Thus, the cytoplasmic
systems of wild-type and dbtP larvae, and of destabilization of PER seems to be one
a PER-bGAL fusion protein (SG3) expressed result of DBT-dependent phosphorylation
from a per promoter in wild-type larvae. The of PER.
extensive sites of PER expression in dbtP
larvae represent bonafide sites of per tran-
scription. The absence of most of these PER PER in Nuclei
sites in wild type most likely results from
post-transcriptional destabilization. In dbtP DBT also regulates nuclear accumulation
this regulation is lost. [Reprinted from Price of PER. In the dbtS mutant, the accumula-
et al. (1998), copyright (1998) with permis-
sion from Elsevier Science.] tion of total PER levels is slightly advanced
relative to wild type in LD, and yet the
accumulation of nuclear PER is delayed by
3 hours. In addition, the time course of per
mRNA is altered in the dbtS mutant, sug-
RHY3 2/6/04 3:52 PM Page 55

POSTTRANSLATIONAL REGULATION OF CIRCADIAN OSCILLATOR PROTEINS BY PHOSPHORYLATION 55

gesting an alteration in the timing of nuclear, while mostly cytoplasmic localiza-


nuclear feedback. The delay in accumula- tion of DBT is found during the early
tion of nuclear PER does not occur in the evening, when PER is cytoplasmic. There-
perS mutant, which has an endogenous fore, DBT may localize to the nucleus or
period quite similar to the dbtS period (19 cytoplasm at different times in part because
and 18 hours, respectively). These results of its association with the PER/TIM
suggest that DBT regulates the nuclear complex. Accordingly, its associations with
accumulation of PER independently of its PER could mediate both effects on cyto-
regulation of cytoplasmic stability. It may plasmic and nuclear regulation of PER
do so by preferentially altering the stability (Fig. 3.10).
of nuclear PER relative to cytoplasmic
PER. Since PER declines to lower levels in
DBT Regulation of PER-Driven Feedback
the nuclei of dbtS flies, it is likely that the
in Nucleus
stability of nuclear PER is reduced by the
dbtS mutation during this time. As well, DBT regulates both the initiation and ter-
analysis of the long-period dbt mutants and mination of feedback exerted by PER in
a perS; dbt double mutant have argued that the nucleus. Destabilization of cytoplasmic
these mutants alter the stability of PER PER and delay of its nuclear accumulation
during the time of its nuclear decline. are predicted to delay initiation of the neg-
However, the effect of the dbtS mutation ative and positive feedback that PER and
on delayed accumulation of nuclear PER TIM exert in the nucleus (Fig. 3.10). Delays
may also result from an effect on the nuclear in negative feedback are thought to be
transport of PER. Mammalian casein kinase essential for circadian rhythmicity, because
I has this effect on mammalian PERs mathematical modeling of the negative
expressed in cultured cells. It binds to PER feedback loop has argued that immediate
and phosphorylates it, thereby masking or negative feedback would produce a consti-
unmasking a nuclear localization signal, and tutive equilibrium; the level of PER protein
either antagonizing or promoting nuclear that accumulates would maintain the equi-
transport of PER (depending on the isoform librium level of per mRNA by repression,
of PER). Likewise, Drosophila DBT could while in turn that level of per mRNA would
antagonize or promote the nuclear trans- maintain a constant level of PER. This is
port of PER, and DBTS would then be a exactly the type of phenotype observed in
stronger antagonist or weaker promoter the larval lateral neurons of the P-element-
than wild-type DBT. The combined analysis induced dbt mutant, in which the high levels
of casein kinase I and PER in mammals and of PER that accumulate are thought to
Drosophila has shown than multiple aspects exert immediate feedback in the nucleus
of PER’s temporal program are regulated (Fig. 3.9). In wild-type flies, the delay in neg-
by its phosphorylation. ative feedback allows high levels of per and
tim mRNA to be expressed before they
are repressed, thereby producing molecular
DBT/PER Association In Vivo and oscillations. Presumably, associations be-
Nuclear Localization tween PER and TIM may eventually stabi-
lize PER and promote nuclear localization
While the DBT protein and mRNA levels of the PER/TIM complex, thereby termi-
do not oscillate, the amount of nuclear nating the delay in initiation of negative
DBT does oscillate (Fig. 3.10). High levels feedback. Alternatively, extensive phos-
of nuclear DBT are found during the late phorylation of PER (or TIM; see
night and morning, at times when PER is discussion below), or some other signal,
RHY3 2/6/04 3:52 PM Page 56

56 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

Figure 3.10. Model for regulation of the timing of nuclear feedback processes by DBT. DBT
binds with PER (P) and phosphorylates it in the cytoplasm, thereby causing it to be degraded
and possibly antagonizing the nuclear import of PER. TIM stabilizes PER and causes it to be
translocated into the nucleus. When light or the circadian clock eliminates TIM, DBT, which
is transported into the nucleus with PER, is able to phosphorylate nuclear PER and target it
for degradation, thereby terminating the repression of CLK/CYC by PER. [Reprinted from Kloss
et al. (2001): Phosphorylation of PERIOD is influenced by cycling physical associations of
DOUBLE-TIME, PERIOD, and TIMELESS in the Drosophila clock. Neuron 30: 699–706, copyright
(2001) with permission from Elsevier Science.]

ultimately triggers nuclear accumulation of duces a TIMUL protein that persists much
PER and TIM. longer than does wild-type TIM in DD.
Since TIM is light-sensitive, light reduces Light eliminates the TIMUL protein and
the levels of nuclear TIM before nuclear produces more rapid phosphorylation of
PER levels are reduced, and the termina- PER. So TIM may antagonize DBT-
tion of feedback in the nucleus is likely to dependent phosphorylation and turnover
result when PER levels are subsequently of PER, while DBT antagonizes the stabi-
reduced to some threshold level. Indeed, lization of PER (and possibly its nuclear
per and tim mRNA do not begin to accu- translocation) effected by TIM in both the
mulate until several hours after nuclear cytoplasm and the nucleus (Fig. 3.10).
TIM is no longer detectable. Since DBT It is predicted that a dbt mutation that
regulates the time course of the decline in increases the cytoplasmic and nuclear sta-
nuclear PER, it also regulates the time at bility of PER should tend to expedite the ini-
which both positive and negative nuclear tiation of nuclear feedback, but delay the
feedback are terminated. However, the termination of the same. Depending on
final stages of PER phosphorylation may whether the effects on initiation or termina-
require the elimination of TIM from the tion are larger, the mutant could either
PER/TIM complex (Fig. 3.10), because produce a shorter- or longer-period clock.
maximal phosphorylation of PER is Perhaps this is why both short- and long-
delayed in the timUL mutant, which pro- period dbt mutants can be isolated. In the
RHY3 2/6/04 3:52 PM Page 57

ENTRAINMENT OF DROSOPHILA CIRCADIAN RHYTHM BY LIGHT 57

case of the dbtS mutant, analysis of resetting this promoter landed was achieved by
of the clock by light shows a shortening of crossing many of these random insertion
the subjective day, when PER is disappear- lines to a tim-GAL4 driver, which expresses
ing from the nucleus and negative feedback GAL4 from the tim promoter and therefore
is terminated. A shortening of this part of specifically causes the UAS-driven genes
the circadian program evidently leads to a to be expressed in clock tissues. In one of
shorter overall period for the circadian these lines, the UAS sequence had inserted
program, despite the delayed accumulation within the sgg gene, thereby causing its
of PER in the nucleus that is observed in LD. overexpression in tim-expressing cells. This
overexpression causes a shortening of cir-
cadian period, an accelerated accumulation
The sgg Protein Kinase
of PER/TIM in nuclei, and an accumulation
The shaggy (sgg) protein kinase regulates of a hyperphosphorylated form of TIM.
the timing of nuclear feedback by phos- Conversely, genotypes in which SGG activ-
phorylating TIM. sgg, which has a role in ity can be specifically lowered in the adult,
the determination of segment polarity, was so that embryonic lethality is bypassed,
originally identified in screens for muta- lengthen circadian period and lead to
tions that affect the segmentation of the hypophosphorylation of TIM and over-
Drosophila embryo. Loss of function muta- expression of PER and TIM. In vitro, a
tions lead to a loss of segment polarity (i.e., vertebrate ortholog of SGG (glycogen syn-
there is no difference between the anterior thase kinase-3b) has been shown to phos-
and posterior parts of the segment). It is phorylate TIM, so the effects on TIM
now known that sgg functions as part of the phosphorylation and nuclear accumulation
wingless (wg) pathway (Fig. 3.11a). WG appear to be direct. TIM is apparently a
protein binds to the Dfrizzled2 receptor on target for SGG just as is b-catenin in the wg
the cell surface, and the signal transduction pathway, except that SGG facilitates the
pathway that is activated by this interaction nuclear accumulation of TIM instead of
downregulates the activity of the sgg antagonizing it, as it does for b-catenin.
kinase (also known as glycogen synthase
kinase 3) on armadillo protein (a b-
catenin). When active, sgg protein (SGG) 䊏 ENTRAINMENT OF DROSOPHILA
causes the phosphorylation of armadillo, CIRCADIAN RHYTHM BY LIGHT
which leads to its degradation. When SGG
activity is downregulated in response to Light-Induced TIM Degradation
the WG signal, armadillo protein is able to
accumulate and move to the nucleus, where Intriguingly, the onset of light leads to a
it activates transcription of a battery of rapid reduction in the levels of TIM, so TIM
genes that confer the anterior/posterior is unstable in the presence of light (Fig.
fate of the segment. 3.12a). This instability is present in both
sgg was identified as a clock gene in wild-type flies and in per0 flies. The response
a screen for genes whose overexpression of TIM to light in both per0 and wild-type
caused aberrant circadian locomotor activ- flies demonstrates that the effect of light on
ity rhythms (Fig. 3.11b). A promoter with a TIM requires neither PER nor a functional
UAS sequence, which binds the yeast tran- clock. Therefore, the response of TIM to
scription factor GAL4, was moved around light can be considered a part of an input
the Drosophila genome, so that it landed at pathway that mediates entrainment to light.
many different random places. Then, over- In wild-type flies, constant light leads to
expression of genes in the regions where arrhythmicity, which is correlated with the
RHY3 2/6/04 3:52 PM Page 58

58 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

Figure 3.11. The dbt and sgg protein kinases have roles in both the Wnt (wg) signaling
pathway and the circadian clock. (a) Model for the role of casein kinase I and sgg in the wg
pathway. The role for casein kinase I has been shown in Xenopus, while the role for sgg has
been shown in many systems (including Drosophila). Casein kinase I activates the wg pathway
by repressing SGG, which otherwise causes b-catenin to be degraded. (b) A role for sgg in the
Drosophila circadian clock has been demonstrated by overexpressing SGG in tim expressing
cells (top), or by lowering SGG protein levels in the adult (bottom). The UAS-GAL4 system was
used to overexpress SGG specifically in tim cells. tim is the promoter of the timeless gene,
GAL4 is a yeast transcription factor, and UAS is the promoter that GAL4 binds and activates.
Bottom, the lethality associated with the sgg mutant was rescued by expressing a sgg trans-
gene, under control of a heat shock promoter, during development. By decreasing the tem-
perature in adults, the effects of reduced sgg expression on circadian rhythms were
determined. The heat shock promoter (hs) activates sgg at elevated temperatures and is
turned off at lower temperatures.

rapid degradation of TIM in response to not contributing to nuclear feedback. The


light and a more gradual decline of PER presence of high levels of tim mRNA allows
levels as well. Recall that PER requires TIM the resynthesis of TIM, stabilization of
for its stability, so constant light produces PER by TIM and transport of PER/TIM
low levels of PER as a consequence of the complex to the nucleus; phase delays are
low levels of TIM. produced in the process. By contrast, during
It is possible to explain the wild- the second half of the night, most PER and
type Drosophila PRC on the basis of the TIM are nuclear and their mRNA levels are
observed per and tim oscillations, the sensi- low, so degradation of TIM in response to
tivity of TIM to light, and the model for the light expedites the termination of nuclear
molecular mechanism of per and tim oscil- feedback, under conditions in which resyn-
lations (Fig. 3.12b). During the first half of thesis of TIM and PER at high rates is not
the night, when phase delays are obtained possible; early termination of nuclear feed-
in response to short light pulses, light back leads to a phase advance. During the
degrades mostly cytoplasmic TIM, which is subjective day, the circadian clock has
RHY3 2/6/04 3:52 PM Page 59

ENTRAINMENT OF DROSOPHILA CIRCADIAN RHYTHM BY LIGHT 59

(a) (b) TIM cannot be retranslated


after its degradation, and
phase advance termination of feedback
is accelerated

24

18 6 no TIM target

12

p ha se delay

TIM is retranslated PER


from high levels of TIM
tim mRNA
TIM level

Phase shift (hours)

Delay Advance
Circadian time (hours)
Minutes after pulse

Figure 3.12. The resetting of circadian phase in response to timed light pulses entails rapid
degradation of TIM. (a) TIM protein was assayed by immunoblot analysis in the heads of wild-
type or tim0 flies collected at the indicated circadian time in the dark, or after a light pulse.
In the top panel, flies were collected after a light pulse at ZT15 (produces a phase delay)
or at ZT19 (produces a phase advance), whereas in the middle panel flies were collected at
the indicated times after initiation of a light pulse at ZT19.5. In the bottom panel, the signal
intensity for TIM was quantitated for several experiments like the one in the middle
panel. [Reprinted from Hunter-Ensor et al. (1996), copyright (1996) with permission from
Elsevier Science.] (b) The phase response curve for wild-type flies in response to 10-minute
light pulses is explained by the response of TIM and per/tim cycling. Flies were entrained to
LD: 12 hours : 12 hours and then released into DD. The average phase of flies that received
a light pulse at the indicated circadian time was subtracted from the average phase of flies
that had not received the pulse to calculate phase shifts. The hatched bars denote the times
when lights would have been illuminated if the LD cycle had continued. [Reprinted from
Hunter-Ensor et al. (1996), copyright (1996) with permission from Elsevier Science. Also,
reprinted with permission from Myers et al. (1996): Light-induced degradation of TIMELESS
and entrainment of the Drosophila circadian clock. Science 271: 1736–1740, copyright (1996)
American Association for the Advancement of Science.] Delays are thought to be produced
when PER/TIM are cytoplasmic and RNA levels are high. Advances result when the proteins
are nuclear and RNA levels are low.

lowered TIM levels, so there can be little or As the amount of light delivered in the light
no further effect of light. The amount of pulse is reduced by shortening the light
phase resetting is therefore little or none. pulse or reducing the intensity of light, the
The degradation of TIM in response to magnitude of phase resetting and TIM
light has been investigated under various degradation are reduced in parallel. In addi-
conditions that address its relevance to tion, the sensitivity of the phase-resetting
resetting of the behavioral rhythms by light. response to the wavelength of the light pulse
RHY3 2/6/04 3:52 PM Page 60

60 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

parallels the sensitivity of TIM degradation Genetic screens and genome-scale analy-
to the wavelength. These parallels are con- sis identified a circadian cryptochrome
sistent with a role for the degradation of photoreceptor. Identification of this pho-
TIM in the resetting of circadian phase. toreceptor benefited from revolutionary
advances that have been occurring in
molecular genetic analyses of many model
Cryptochrome: A Dedicated Circadian
organisms.The genetic screen made use of a
Photoreceptor
clock-controlled reporter gene, consisting of
Since TIM is not homologous to any known firefly luciferase fused to the N-terminal
photoreceptor and is not directly sensitive two-thirds of per and transcribed from a per
to light, it was thought to be the down- promoter (Fig. 3.13). Firefly luciferase
stream target of a clock photoreceptor metabolizes the organic molecule luciferin
pathway. Mutant adults lacking eyes or to produce light emission. When expressed
visual transduction pathways have rhythms under the control of the per promoter,
that can be entrained to light and can luciferase mRNA and protein oscillate with
degrade TIM in response to light, so the a circadian rhythm. Fruitflies fed luciferin
photoreceptors and biochemistry used for produce a rhythmic glow resulting from the
vision are not necessary for entrainment of oscillating levels of luciferase,which thereby
circadian rhythms by light (Fig. 3.2). “reports” the timing of the circadian clock.
Instead, it seemed likely that at least one Mutations in other, endogenous genes of the
nonvisual photoreceptor pigment and fruitfly are expected to alter the rhythmicity
pathway were involved in entrainment of and period of this glow if the affected genes
the Drosophila clock by light. contribute to the circadian oscillator or

Inputs Circadian oscillator

Rhythmic glow

Light x x
LUCIFERASE

Luciferin

per promoter per luciferase

Figure 3.13. Design of a genetic screen for mutants that affect the circadian oscillation of
per transcription. Circadian oscillations of luciferase transcription are driven by the per pro-
moter, which responds to clock-driven alterations of dCLK/CYC-dependent activation. The
resulting circadian oscillations of luciferase activity produce a rhythmic glow in flies fed
luciferin (the substrate for luciferase). The glow can be measured and analyzed in single flies,
just as locomotor activity can be (Fig. 3.1). A mutation (X) that affects the input pathway or
the oscillator mechanism is predicted to alter the circadian glow rhythm.
RHY3 2/6/04 3:52 PM Page 61

ENTRAINMENT OF DROSOPHILA CIRCADIAN RHYTHM BY LIGHT 61

input pathways that regulate the per blue light, so a blue-light photoreceptor had
promoter (Fig. 3.13). In such a screen, a long been implicated in circadian photore-
mutation affecting a clock photoreceptor ception. The Drosophila gene was dubbed
was identified because it produced an cry in recognition of its homology to the
arrhythmic glow of luciferase. plant blue-light photoreceptors, which were
The affected gene also turned up as called cryptochromes. Plant cryptochromes
an expressed sequence in the Berkeley and photolyases bind to two chromophores:
Drosophila Genome Project, which was pterins (MTHF or 8-HDF) and flavins. The
actively engaged in sequencing the entire conceptual translation of the Drosophila
Drosophila genome at the time (likewise, cry gene contains domains with sequence
the Human Genome Project was undertak- homology to the domains that bind these
ing the same task for the human genome). chromophores in photolyases and plant
The sequence attracted the interest of clock cryptochromes (Fig. 3.14).
researchers because it was homologous to a The circadian phenotype of the mutant
class of blue-light photoreceptors in plants, (cryb) was consistent with a role for the
and to a class of enzymes called photo- gene in circadian photoreception (Fig.
lyases, which are activated by blue light to 3.14). The cryb mutation is predicted to
repair DNA damaged by ultraviolet light change an amino acid in the domain that
(Fig. 3.14). Resetting of the Drosophila cir- binds the flavin chromophore, and so it is
cadian rhythm is maximally responsive to likely to disrupt the detection of light that

Figure 3.14. Drosophila cry is homologous to photolyases and plant cryptochromes and
has the properties of a circadian photoreceptor. All of these proteins have binding sites for
two kinds of chromophores (FAD and MTHF/8-HDF). Human and Drosophila CRYs are more
homologous to the 6–4 photolyases than to the other family members. Note that not all
photoreceptor functions are subserved by Drosophila cry.
RHY3 2/6/04 3:52 PM Page 62

62 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

is mediated by this chromophore. In the dark period. Moreover, it associates directly


cryb mutant, TIM is no longer degraded in with TIM in yeast only in the presence of
response to light in most tissues, and both light (Fig. 3.15a). The CRYb protein, which
PER and TIM are constitutively expressed is predicted to be unresponsive to light
at moderately high levels in most expres- because it cannot bind the flavin chro-
sion sites. One exceptional site is a subset mophore, does not associate with TIM in
of the lateral neurons, which, as discussed either light or dark. Hence, CRY is capable
above, are the cells that control rhythmic of a light-influenced association with TIM
locomotor activity. Here, PER and TIM in the absence of any other Drosophila-
continue to oscillate and are entrained by specific component besides TIM. In a
light. Likewise, locomotor activity contin- Drosophila cell line, coexpression of CRY
ues to be rhythmic in the cryb mutant, and with dCLK, PER, and TIM abrogates the
it is entrained by LD cycles. However, the repression of a dCLK-activated reporter
capacity of short light pulses to reset the gene, but only if the cells are exposed to
clock is virtually eliminated, so the cryb light. In the dark, PER/TIM can repress
PRC is quite flat. In complementary exper- dCLK-activated transcription even if CRY
iments, CRY overexpression in clock cells is coexpressed. Moreover, coexpression of
alters the sensitivity of the phase response the CRYb protein does not eliminate
to pulses of light relative to wild-type flies. repression by PER/TIM even in the
Finally, the arrhythmic locomotor activity presence of light (Fig. 3.15b).
that normally results from LL is not In addition, a light-dependent association
obtained in cryb mutants (i.e., the rhythms between PER and CRY has been detected
resemble those obtained in DD). in yeast and a Drosophila cell line. Deletion
These results argue that cry is not an of the C-terminal end of CRY allows this
essential component of the molecular oscil- association with PER, as well as the one with
lator in lateral neurons, but rather that it is TIM, to occur in both the light and the dark.
a component of a somewhat redundant These results have been interpreted to show
input pathway that responds to light. While that the C-terminal end of CRY mediates
some effects of light on lateral neurons are the light-dependent interactions with PER
entirely dependent on cry, others are also and TIM. It has been proposed that the C
responsive to other photoreceptors. The terminus binds a nuclear repressor of the
constant levels of TIM that are observed in PER and TIM interactions in the dark, while
most clock tissues could result from an light eliminates the CRY/repressor interac-
absence of photic entrainment and light- tion and produces an interaction with PER
mediated TIM degradation in these tissues. and TIM instead. Deletion of the C-terminal
However, evidence presented in a later region would also eliminate the interaction
chapter will argue that cry is likely to be a with this repressor and produce interactions
component of the oscillator mechanism in with PER or TIM in both light and dark.The
some peripheral oscillator tissues outside role of the PER/CRY interaction in the
the lateral neurons (see Chapter 9). entrainment or function of the clock is not
yet known.
CRY/TIM Association and Degradation
Phosphorylation and Ubiquitination
In response to light, CRY associates with
TIM and both are degraded. In wild-type The response of TIM to light involves phos-
flies, CRY protein is light-sensitive, such phorylation and degradation by the ubiqui-
that it degrades in response to light and is tin proteasome pathway. The ubiquitin
expressed at high levels only during the proteasome pathway is one of the major
RHY3 2/6/04 3:52 PM Page 63

ENTRAINMENT OF DROSOPHILA CIRCADIAN RHYTHM BY LIGHT 63

(a) (b)

(c) GAL4
In the
eye GAL4 UAS cry eyes of
CRY cry b flies
Oscillations of TIM are restored
only in the eyes.

GAL4 In the
LN GAL4 UAS cry lateral
CRY neurons
Locomotor activity be comes arrhythmic in LL of cry b flies
and is reset by short light pulses.

Figure 3.15. CRY undergoes a light-influenced association with TIM and acts intracellularly.
(a) The yeast two-hybrid assay (described in Fig. 3.7a) shows that CRY associates with TIM in
the light but not the dark. Proteins designated “LA-” are expressed as fusions with the LEXA
DNA-binding domain, while those designated “VP16-” are expressed as fusions with the VP16
RNA polymerase-activating domain. CRY could also form a three-component (ternary
complex) with PER, if TIM was present to complex with both PER and CRY (hence the inter-
action detected with LA-CRY, TIM, and VP16-PER; however, as described in the text, subse-
quent studies also found direct interactions between PER and CRY). CRYb cannot associate
with TIM in either light or dark, and PER associates with TIM in either light or dark. (b) CRY
disrupts the repression of dCLK by PER/TIM, but only in the presence of light. CRYb does not
disrupt PER/TIM function. The cell culture assay is described in Figure 3.7b. [Reprinted with
permission from Ceriani et al. (1999), copyright (1999) American Association for the Advance-
ment of Science.] (c) Expression of CRY only in the eyes or only in the lateral neurons rescues
the cry mutant phenotype only in the cells where CRY is expressed. The GAL4/UAS expression
system is described in Figure 3.11b. The eye-specific (“eye”) promoter is the rhodopsin
promoter, and the lateral neuron-specific (“LN”) promoter is the pigment dispersing factor
promoter.

protein degradation pathways in the cell. In macromolecule thought to unfold ubiquiti-


this pathway, lysines on the protein targeted nated substrates and force the polypeptide
for degradation are covalently modified backbone through a proteolytic core, which
with chains of a small peptide monomer degrades the protein. Inhibitors of the pro-
called ubiquitin. These polyubiquitin chains teasome pathway inhibit the degradation
mediate the binding of the modified protein of TIM in response to light (Fig. 3.16). In
to the 26S proteasome—a large, cylindrical addition to ubiquitination, the degradation
RHY3 2/6/04 3:52 PM Page 64

64 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

Proteasome
Light

t
por
ans
CRY e-tr ?
CRY
CRY CRY

TIM TIM TIM Tyr PO4 TIM Tyr PO4

Lys
TIM

Ubi
Ubi
Figure 3.16. Model for the light-dependent degradation of TIM and CRY by the proteasome.
Phosphorylation of TIM on tyrosine (Tyr-PO4) is thought to be necessary, as is the addition of
ubiquitin peptides (Ubi) to lysines in TIM.

of TIM in response to light requires tyro- CRY Mediation of Photoreception in


sine kinase activity. Elevated levels of phos- Clock Cells
photryrosine can be detected in TIM after
a light pulse, and inhibitors of tyrosine If CRY mediates entrainment by directly
kinases prevent the degradation of TIM in binding to TIM, then CRY should mediate
response to light. photoreception within the clock cells, rather
The degradation of CRY in response than in a separate photoreceptor cell type
to light is also inhibited by proteasome that communicates synaptically with the
inhibitors, by inhibitors of electron trans- clock cell. Recall that the compound eye and
port, and by several mutations in cry. The visual transduction pathway are not
model is that light induces a conformational required for clock photoreception. To test
change in CRY, which then donates elec- the possibility that CRY mediates cell-
trons from reduced flavin to downstream autonomous photoreception, experimental
components that signal degradation of control of CRY’s tissue specific expression
CRY (Fig. 3.16). In a cell line expressing is required. As described for SGG above, it
CRY and exposed to light, inhibition of is possible to express genes in different, spe-
electron transport elevates the levels of cific cell types by using the yeast GAL4 tran-
ubiquitinated TIM (in this cell line, TIM is scription factor as an intermediary (Fig.
ubiquitinated, but not degraded in response 3.15c). A promoter that is activated by
to light). Thus, electron transport is GAL4 because it has been genetically engi-
required to turn off CRY-mediated ubiqui- neered to have binding sites for GAL4
tination of TIM in response to light; (termed “UAS”) was linked to a coding
presumably, light converts CRY into an sequence for cry and introduced as a trans-
active form that promotes ubiquitination gene into flies. In addition, promoters that
and degradation of TIM, while electron were active in diverse cell types were linked
transport terminates this activity by con- to the coding region for GAL4, producing a
verting CRY to an inactive form or trigger- number of transgenes, each of which
ing the degradation of CRY. expresses GAL4 in a distinct cell type.
RHY3 2/6/04 3:52 PM Page 65

REGULATION OF CIRCADIAN-CLOCK-CONTROLLED OUTPUTS BY THE OSCILLATOR MECHANISM 65

For instance, by hooking GAL4 to the underneath the retina and project to the
rhodopsin promoter, the transgene will be lateral neurons (the Hofbauer–Buchner
expressed only in certain photoreceptors of eyelet; Fig. 3.17). Therefore, in the gl;cryb
the eye. By hooking it to the pdf promoter, double mutant, there is an absence of both
which is expressed only in the lateral functional CRY protein and any input from
neurons (and that is described in more detail visual photoreceptors external to the lateral
later), the transgene will express GAL4 only neurons. With both types of input gone, the
in the lateral neurons. If a fly carrying a rhythms of PER and TIM are not entrained
tissue-specific GAL4 transgene is crossed to in the lateral neurons, and behavioral
a fly carrying a UAS-cry transgene, any rhythms cannot be entrained. Evidently,
progeny inheriting both transgenes will now there is functional redundancy in the input
express cry in the same tissue-specific pathways to the lateral neurons, such that
pattern as GAL4, since GAL4 is driving the lateral neurons are still photically
expression of cry (Fig. 3.15c). entrainable as long as either a CRY pathway
Using this methodology, it is possible to or visual pathway is available.
show that expression of wild-type cry in a
cryb mutant produces a wild-type pheno-
type (a “rescue”) only in those tissues 䊏 REGULATION OF CIRCADIAN-
where cry is expressed (Fig. 3.15c). If CLOCK-CONTROLLED OUTPUTS BY
rhodopsin-GAL4 is the driver, oscillations
THE OSCILLATOR MECHANISM
of TIM are rescued only in the eye but not
in the body. If the lateral neuron-specific
Function of the Molecular Oscillator
pdf-GAL4 is the driver, the defective PRC
and the lack of arrhythmicity in response to The molecular oscillator drives the oscilla-
LL are rescued in the cryb mutant (recall tions of numerous clock-controlled genes.
that entrainment of lateral neurons and In theory, any gene which has an E-box-
rhythmic locomotor activity is still obtained containing promoter that is responsive to
in the cryb mutant, so there is nothing to the dCLK/CYC heterodimer can be regu-
rescue with respect to these). The results lated by the molecular oscillator described
are consistent with a role for cry that is cell in this chapter (Fig. 3.18). These clock-
autonomous, and with the role of the lateral controlled genes need not be part of the
neurons in circadian locomotor activity. mechanism that generates the molecular
oscillations, but they may encode factors
that translate the molecular oscillator into
Non-cell-Autonomous Entrainment
actual changes in cellular biochemistry
Since circadian rhythms of locomotor activ- and physiology: the clock outputs. Screens
ity can still be entrained to 12-hour : 12-hour for these genes have involved molecular
light : dark cycles in the cryb mutant, CRY approaches that identify mRNAs whose
cannot be the only circadian photoreceptor levels change over the course of the day
for the lateral neurons controlling locomo- (see Chapter 2). Only a handful of such
tor behavior. In support of this conclusion, genes have been described in Drosophila
locomotor activity in a gl;cryb double mutant by single-gene-based approaches, but as
does not entrain to LD cycles. The gl muta- this chapter goes to press, many more can-
tion eliminates all known “visual” photore- didates are emerging from whole-genome-
ceptors in the adult fly: the two compound based approaches (“genomics”).
eyes, the three ocelli (light-sensitive organs In a screen for genes regulated
at the top of the head), and the two internal, in response to LD cycles, 20 genes termed
rhodopsin-positive cell groups that lie dregs (for Drosophila rhythmically ex-
RHY3 2/6/04 3:52 PM Page 66

66 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

gl6oj

Compound eye gl6oj ? Ocelli

H-B eyelet cry b DN1


DN2 gl6oj

LNd
?
I-LNv
cry b s-LNv

Figure 3.17. Redundant entrainment pathways for the lateral neurons as revealed by the
effects of various mutations on entrainment. The cryb mutation disrupts the Drosophila cryp-
tochrome photoreceptor. The gl mutation prevents development of all the cells that express PER
in the adult head except the lateral neurons. Therefore, a gl;cry double mutant has no entrain-
ment pathways. [Reprinted from Helfrich-Forster et al. (2001): The circadian clock of fruit flies
is blind after elimination of all known photoreceptors. Neuron 30: 249–261, copyright (2001)
with permission from Elsevier Science.] The cells that are potentially circadian photoreceptors
include the dorsal and ventral (large and small) lateral neurons (LNd, l LNv, s LNv), the compound
eye, the ocelli (three light-sensing organs on top of the head), the H-B eyelet (a cluster of pho-
toreceptors between the retina and the optic lobe), and a cluster of dorsal neurons (DN). The
pathways blocked by each of the two mutations, gl 60j; and cry b, are indicated by the bars on
the arrows.

pressed genes) were identified in a collec- (takeout) have been identified in separate
tion of cDNAs expressed in the head but not screens for genes whose mRNAs exhibit cir-
the early embryo.All but three of these have cadian oscillations or regulation.
evening peaks of mRNA expression, like
those of per and tim, while the expression of
vrille
the other three peaked in the morning.
Some of these genes show regulation by LD vrille is a clock-controlled gene that also
cycles and food, in addition to (or instead of) affects clock function. vrille is a member of
regulation by the endogenous clock, while the basic leucine zipper class of transcrip-
others show oscillations in DD and require tion factors, and it has a dCLK/CYC
per function. Only one of the genes (alcohol responsive site in its promoter. As is pre-
dehydrogenase) has a known function. In dicted by this feature of its promoter, vrille
addition, two transcription factors (vrille mRNA exhibits oscillations that are in
and crg-1) and a ligand binding factor phase with those of per and tim in LD and
RHY3 2/6/04 3:53 PM Page 67

REGULATION OF CIRCADIAN-CLOCK-CONTROLLED OUTPUTS BY THE OSCILLATOR MECHANISM 67

Figure 3.18. A molecular model for the circadian clock in Drosophila. The rhythmic accumu-
lation of PER and TIM proteins results in the formation of a PER/TIM complex, which negatively
regulates two feedback loops involving dCLK/CYC. In one of these loops (solid lines) dCLK/CYC
positively regulate per and tim transcription, and in the other loop (dotted lines) dCLK/CYC
negatively regulates dClk mRNA by activating vri expression. Light, CRY, and SGG regulate a
lag in nuclear feedback by PER/TIM through their effects on TIM, and light entrains the oscil-
lator through CRY-mediated degradation of TIM. DBT regulates the initiation and termination
of nuclear feedback through its effects on PER. In addition to driving transcription of per and
tim, dCLK/CYC is thought to activate transcription of numerous clock-controlled genes, which
translate the rhythm of the circadian oscillator into circadian outputs of biochemistry and cell
physiology.

DD, and the oscillations are eliminated by fore derive from effects on dClk. vrille is
the per0 mutation. In the adult head, vrille therefore part of the central oscillator
is expressed in photoreceptor cells and the mechanism, although it was isolated in a
lateral neurons, in a pattern that is identical screen for output genes.
to the expression pattern of tim. While
mutations in vrille are homozygous lethal,
takeout
heterozygotes are viable and have slightly
shortened circadian periods. Overexpres- The takeout (to) gene mediates resistance
sion of vrille in clock cells produces long to starvation. Its mRNA oscillates with a
periods and arrhythmicity and depresses phase that is several hours delayed relative
expression of per, tim, dClk and pigment to per and tim mRNA. While the 5¢
dispersing factor (PDF, a neuropeptide upstream region of to has a potential E box,
released by the lateral neurons; more on this region of the to promoter does not
PDF below). The dClk promoter is nega- confer circadian oscillations to reporter
tively regulated by vrille, and all these genes and does not confer inducibility by
effects on clock gene products may there- dCLK/CYC in the cell culture assay. There-
RHY3 2/6/04 3:53 PM Page 68

68 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

fore, the mechanism for circadian regula- nism described in this chapter. Many genes
tion of to is not well understood, although that do not exhibit circadian oscillations
it is clear that known clock genes are nevertheless exhibit altered levels of con-
involved, because to mRNA is downregu- stitutive expression in dClkJrk or per0
lated in several null mutations of known mutant flies. The basis for these altered
Drosophila clock genes. In addition to expression levels is not known.
circadian oscillations, to mRNA exhibits
induction in response to starvation. For
Posttranscriptional Regulation
reasons that are not clear, this induction
requires the presence of a functional circa- On the basis of our current model for this
dian clock, as it does not occur in clock-null oscillator, promoters with E boxes seem
mutants (per0, tim0, and cyc0). The to gene likely to be the most direct target in output
confers resistance to starvation, because a pathways. However, rhythmically expressed
mutation in to leads to more rapid lethality transcripts will produce rhythmically
in response to starvation. It is possible that expressed proteins. These proteins can
clock regulation of to contributes to meta- produce downstream rhythms of other pro-
bolic and food-related oscillations. This teins, whose transcription is not necessarily
possibility is intriguing, in light of the regu- rhythmically controlled.
lation of mammalian PAS-bHLH factors by
redox state (see Chapter 4).
Cyclic Activity of cAMP Response
Element Binding (CREB) Protein
Genomic Methods
The capacity of CREB to activate transcrip-
Genomic approaches have yielded several tion from a cAMP response element
hundred possible clock-controlled genes. (CRE)-driven luciferase reporter oscillates
The Berkeley Drosophila Genome Project with a circadian rhythm that is affected by
resulted in the nucleotide sequence of the per mutations just as circadian behavior is
majority of the genes in Drosophila. This affected. Since CRE sites are targets of
allowed microarray analysis of the type CREB, the cycling of this reporter is indica-
described in Chapter 2 to identify circadian tive of cyclic CREB activity. The level of
changes in gene expression. Initial attempts CREB does not oscillate, so it must be
to analyze circadian changes in gene regulated posttranslationally—perhaps by
expression by several groups have pro- phosphorylation. Mutations in the Creb
duced somewhat different tabulations of gene shorten the period of circadian
clock-controlled genes. With refinement of behavior and blunt per mRNA and protein
the technology and confirmation of candi- oscillations. In fact, the per promoter has
date genes, a comprehensive list should several putative CRE elements, so CREB
emerge. Already it seems that many of the activity may feed back to regulate the per
output genes differ in phase from per, tim, promoter. Alternatively or additionally,
and dCLk. Many of them are involved in CREB activity may be part of an output
common processes, such as learning and pathway regulating locomotor activity, as
memory/synapse function, vision, olfaction, circadian regulation of the CREB activity is
locomotion, detoxification, proteolysis, blunted in mutants that specifically affect
immunity, and metabolism. None of the output pathways rather than the central
clock-controlled genes exhibit circadian clock mechanisms. For instance, the NF1
oscillations in the ClkJrk mutant, so there is mutation, which upregulates a mitogen acti-
no evidence for any circadian oscillator vated protein kinase pathway involved in
independent of the core oscillator mecha- output to locomotor rhythms, and the dfmr1
RHY3 2/6/04 3:53 PM Page 69

REGULATION OF CIRCADIAN-CLOCK-CONTROLLED OUTPUTS BY THE OSCILLATOR MECHANISM 69

mutation, which disrupts synapse forma- micity of locomotor activity and eclosion.
tion, both blunt the rhythm of CREB activ- This result indicates the importance of this
ity (see text below). Since CREB is involved region to the clock output pathway.
in consolidation of memories, circadian reg- Because neither elimination nor overex-
ulation of CREB may couple the circadian pression of PDF completely eliminates
clock with learning and memory, which is rhythms of locomotor activity, it is likely
thought to be consolidated during sleep in that other locomotor outputs exist.
vertebrates. The importance of proper synaptic con-
nections for the PDF-releasing lateral
neurons is suggested by studies of Dro-
pdf
sophila dfmr1 mutants. These mutants do
The neuromodulator, pigment dispersing not express a homolog of a human gene
factor (pdf), is an important circadian that is mutated in one of the most common
output for circadian control of locomotor forms of mental retardation: the fragile X
activity and eclosion. The work that led syndrome. The Drosophila mutants display
to this knowledge entailed an elegant com- a high frequency of arrhythmic circadian
bination of comparative endocrinology locomotor activity. While oscillations of
and molecular genetic analysis. Pigment PER and TIM are normal in the lateral
dispersing hormones were isolated as neurons, the projections of the lateral
substances that mediate dispersion and neurons are abnormal. Hence, while the
translocation of light-adaptational pig- lateral neuron circadian oscillator is appar-
ments in crustaceans. Antibodies to these ently normal in these flies, it is not able to
crustacean hormones detect immunoreac- communicate its temporal information to
tivity in the Drosophila lateral neurons, its outputs, because of abnormal axonal
which are the site of the clock that controls pathfinding or synapse formation.
rest–activity rhythms. Therefore, a molecu-
lar analysis of Drosophila pigment dispers-
lark
ing factor (pdf ) was undertaken, in order
to determine what role this factor might The lark mutation was isolated in a screen
have in Drosophila circadian rhythms. for mutations affecting the timing of eclo-
Several clock mutants affect the expression sion (Table 3.1). Heterozygous lark mutant
of pdf; mutations in dClk and cyc reduce flies eclose early in an LD cycle, but do not
pdf mRNA, and overexpression of vrille exhibit an altered circadian period of eclo-
reduces the level of pdf peptide (PDF) sion in DD, nor any alterations in circadian
without affecting the levels of pdf mRNA. control of locomotor activity. The muta-
While levels of pdf mRNA and protein do 0tion produces embryonic lethality when
not oscillate, PDF rhythmically accumu- homozygous. lark protein (LARK), which
lates at the terminals of the lateral neurons, is predicted to be an RNA binding protein,
and this rhythm is absent in the per0 and exhibits an oscillation at the protein level,
tim0 mutants. It is likely that this rhythm is while lark mRNA does not oscillate. High
effected by rhythmic transport and/or levels of protein are expressed during the
release from the lateral neuron terminals. day, and the oscillation persists in DD and
A pdf mutant that does not express this requires functional PER. The high levels
peptide has a high frequency of arrhythmic of LARK during the day, when eclosion
locomotor activity. In addition, the dorsal behavior is suppressed, are consistent with
central brain, to which the lateral neuron a function for LARK as a repressor of eclo-
axons project, is the only site of overex- sion. While LARK is localized in the nuclei
pression that significantly alters the rhyth- of most or all pupal neurons, it is also
70
䊏TABLE 3.1. Circadian Mutants in Drosophila
Gene Name Phenotype of Mutants Function of Protein Circadian Targets
period (per)a,c ar, short, or long circadian behavioral and Interacts with TIM, dCLK and CYC to Oscillator
molecular rhythms negatively regulate dCLK/CYC
timeless (tim)a,b,c ar, short, or long circadian behavioral and Interacts with PER, dCLK and CYC to Oscillator
RHY3 2/6/04 3:53 PM Page 70

molecular rhythms negatively regulate dCLK/CYC


dClock (dClk)a ar circadian behavioral and molecular rhythms Interacts with CYC to positively regulate Oscillator
per and tim mRNA and to negatively
regulate dClk mRNA; both types of
regulation repressed by PER/TIM
cycle (cyc) ar circadian behavioral and molecular rhythms Interacts with dCLK to positively regulate Oscillator
per and tim mRNA and to negatively
regulate dClk mRNA; both types of
regulation repressed by PER/TIM
doubletime (dbt)c ar, short, or long circadian behavioral and Casein kinase I ortholog which interacts Oscillator
molecular rhythms with PER to produce phosphorylation
of PER; regulates stability and timing
of nuclear feedback by PER
shaggy (sgg) Short or long circadian behavioral and Glycogen synthase kinase III ortholog Oscillator
molecular rhythms which phosphorylates TIM to regulate
the timing of TIM’s light sensitivity and
nuclear feedback
vrille (vri)a Short, long, and damped circadian behavioral Transcription factor which is positively Oscillator
and molecular rhythms regulated by dCLK/CYC and which
negatively regulates per, tim and dClk
mRNA
cryptochrome Arrhythmic and high levels of PER and TIM Blue-light photoreceptor which dimerizes Input; peripheral
(cry)b,d in most tissues; rhythmic behavior and with TIM to effect degradation of both oscillators
molecular oscillations in lateral neurons. TIM and CRY
larke Early eclosion in heterozygotes; homozyogus RNA-binding protein that may regulate Eclosion (output)
lethal the mRNA of a hormone for eclosion
takeout (to)d Hypersensitive to starvation; no effect on Most likely a ligand-binding protein Starvation
molecular oscillations (output)
Crebf per oscillation is damped, circadian period of cAMP response element binding protein: Possibly PDF
locomotor activity shortened transcription factor regulated by response
RHY3 2/6/04 3:53 PM Page 71

phosphorylation (output)
Neurofibromatosis-1 Arrhythmic locomotor activity; no effect on Inactivates signaling through the RAS Possibly PDF
(NF-1) molecular oscillations protein kinase pathway response
(output)
protein kinase A Loss of some locomotor activity rhythmicity; A protein kinase activated by cAMP Locomotor
(PKA) no effect on molecular oscillations activity
(output)
dunce (dnc) Altered phase response curve and slight cAMP phosphodiesterase ?
shortening of period
pigment dispersing Loss of locomotor activity and eclosion Neuropeptide which accumulates Locomotor,
factor (pdf) rhythmicity rhythmically in lateral neuron eclosion
terminals outputs
fragile X syndrome Loss of some locomotor rhythmicity; abnormal mRNA binding protein which alters Locomotor,
gene (dfmr1) synapse formation; no effect on molecular connections of lateral neurons with eclosion
oscillations postsynaptic neurons outputs
disconnected (disco) Loss of locomotor and eclosion rhythmicity Required for proper pathfinding of the Clock cells which
larval and adult optic nerve and lateral control
neuron determination behavior
a
mRNA and protein oscillate with a circadian rhythm.
b
Levels of protein acutely depressed by light.
c
Nuclear localization of protein oscillates with a circadian rhythm.
d
mRNA oscillates with a circadian rhythm.
e
Protein but not mRNA oscillates with a circadian rhythm.
f
Activity oscillates with a circadian rhythm.

71
RHY3 2/6/04 3:53 PM Page 72

72 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

expressed in the cytoplasm of neurons that the vicinity of the dorsal PDF projections
release a peptide involved in triggering and levels of phospho-MAPK are reduced
eclosion. It is possible that LARK con- in the pdf null mutant.
tributes to a translational control mecha-
nism that regulates the synthesis or release
of this hormone (crustacean cardioactive 䊏 PRESENT QUESTIONS AND
peptide). However, mutations in the RNA FUTURE PERSPECTIVES
binding domains of lark do not affect the
circadian regulation of eclosion, so lark While the genetic analysis in Drosophila
may not affect eclosion by binding to has been remarkably successful in integrat-
mRNA. ing many observations and diverse genes
into a coherent model (Fig. 3.18), there
are still some phenomena that are not
PKA and Nf1
explained by the model put forward in this
Protein kinase A (PKA), which is stimu- chapter. One of these phenomena is tem-
lated by cAMP, is specifically involved in perature compensation, which refers to the
the circadian regulation of locomotor activ- relative constancy of circadian period at
ity, because mutations in the PKA catalytic different temperatures and is a defining
and regulatory subunits produce a high fre- feature of circadian rhythms (see Chapter
quency of locomotor arrhythmia without 1). Attempts have been made to explain
affecting eclosion or the oscillations of per. temperature compensation within the
Modulations of cAMP levels may also be framework of the existing Drosophila
involved in some part of the clock mecha- model, but there has been little progress.
nism, as dunce mutations, which reduce the Another unexplained phenomenon is that
level of cAMP-specific phosphodiesterase, the per mutations affect the periodicity of
alter the Drosophila PRC, shorten the cir- an ultradian rhythm of approximately one
cadian period, and enhance a circadian minute. This is a rhythm in the courtship
oscillation of cAMP levels. song that the male Drosophila sings to the
Null mutations of the neurofibromatosis- female—a song that ends with mating, if it
1 (Nf1) gene also specifically disrupt loco- is sung well! The song consists of pulses of
motor activity rhythms. per and tim wing beats, and the frequency of these
oscillations are not affected by these muta- pulses oscillates with a period of about a
tions, consistent with a function downstream minute in wild-type flies. The period is
of the clock. Since the NF1 protein was shorter in perS flies, longer in perL flies, and
known to signal through PKA in flies, the arrhythmic in per0 flies. The role proposed
obvious explanation was that its effects on for per in the circadian molecular oscillator
rhythms were mediated by PKA. However, is on a timescale that cannot explain the
it turned out that NF1 acts through the effects on these ultradian rhythms.
Ras/MAPK (mitogen-activated protein Within the framework of the model pro-
kinase) pathway to effect clock output. posed in this chapter, the oscillations of per
MAPK activity is elevated in NF1 mutants, and tim gene products exhibit complexities
and Ras/MAPK mutations that reduce that are not explained by the model. For
MAPK signaling suppress the arrhythmia instance, other promoter sequences besides
caused by Nf1. It is likely that release of PDF E boxes contribute to the cycling regulation
from lateral neuron terminals stimulates of transcription, and there is also posttran-
MAPK activity in the postsynaptic neurons, scriptional regulation of per mRNA and of
because a circadian oscillation in phospho- some other clock-controlled mRNAs.Addi-
MAPK (the activated kinase) is observed in tionally, circadian rhythms of behavior
RHY3 2/6/04 3:53 PM Page 73

FURTHER READING 73

occur in some transgenic flies in which per, clock, but the other mammalian orthologs
tim, or dClk are expressed from promoters of Drosophila clock genes have clock roles.
that do not produce normal circadian oscil- CRY appears to be more of a central
lations of these mRNAs. Thus, rhythmic component in the mammalian clock; it
expression of per, tim, or dClk mRNA is not serves to facilitate nuclear accumulation
necessary for circadian rhythms. Neverthe- of PER, and it negatively regulates
less, these transgenes support rescue of cir- CLK/CYC. PER is also involved in this
cadian rhythms of PER, TIM, or dCLK negative regulation, in the positive regula-
proteins. It is possible that rhythmic feed- tion of a CYC ortholog, and is induced in
back on per/tim and dClk transcription is response to light. The differences and
not an essential function for the oscillations similarities between the mammalian and
of PER and TIM in the circadian oscillator Drosophila clocks are discussed more thor-
mechanism. Perhaps oscillating transcrip- oughly in subsequent chapters.
tion of one or more of the other genes that As novel genetic screens are devised
are regulated by clock-controlled E boxes and the genome-scale screens for clock-
(e.g., vrille) is sufficient to drive the post- controlled genes are refined, it is highly
transcriptional oscillations of PER and likely that additional genes will be shown to
TIM. Alternatively and more profoundly be part of the clock mechanism. Some of
model-altering, PER and TIM may be able these may be assigned to input and output
to drive their oscillations entirely through pathways, but it is possible that the distinc-
feedback loops that are posttranscriptional. tion between these divisions of the circa-
If the latter possibility materializes, the dian system may begin to blur, as many of
major function of PER and TIM in the cir- these pathways may relate to the central
cadian oscillator has yet to be elucidated. mechanism as well. It has already become
It has been shown that electrical activity clear that many of the output pathways are
of the lateral neuron pacemaker cells is controlled by peripheral oscillators rather
required for persistent circadian oscilla- than distinct output mechanisms. Under-
tions of PER and TIM, as well as for circa- standing the integration of multiple oscil-
dian rhythms of behavior. Loss of electrical lators to produce rhythmicity at the
activity was produced by expressing K+ organismal level presents a major challenge
channel mutants in the lateral neurons. The (see Chapter 9). Clearly, times that are as
expression of these mutant channels does exciting as the late 1990s through 2003 still
not kill the lateral neurons, because oscilla- lie ahead for molecular chronobiologists.
tions of PER and TIM persist if the trans-
genic flies are maintained in LD cycles
instead of DD. Potentially, electrical activ- FURTHER READING
ity contributes in some way to one of the
known feedback loops, or to a novel mech- Allada, R, Emery, P, Takahashi, JS, Rosbash, M
anism that is not yet understood. (2001): Stopping time: The genetics of fly and
One of the successes of the Drosophila mouse circadian clocks. Annu Rev Neurosci
model is that many of its features have been 24: 1091–1119.
extended quite well to the mammalian Allada, R, White, NE, So, WV, Hall, JC, Rosbash,
M (1998): A mutant Drosophila homolog of
clock, which is covered in Chapter 4. The
mammalian Clock disrupts circadian rhythms
mammalian clock has three known per and transcription of period and timeless. Cell
genes, two cry genes, a family of CLK/CYC 93: 791–804.
transcription factors, and a family of casein Blau, J, Young, MW (1999): Cycling vrille expres-
kinase I genes similar to dbt. Apparently, sion is required for a functional Drosophila
the mammalian TIM is not involved in the clock. Cell 99: 661–671.
RHY3 2/6/04 3:53 PM Page 74

74 DROSOPHILA MELANOGASTER: A MODEL SYSTEM FOR MOLECULAR CHRONOBIOLOGY

Ceriani, MF, Darlington, TK, Staknis, D, Mas, P, heterodimer: A basis for circadian transcrip-
Petti, AA, Weitz, CJ, Kay, SA (1999): Light- tion. Mol Cell Biol 19: 5316–5325.
dependent sequestration of TIMELESS by Martinek, S, Inonog, S, Manoukian, AS, Young,
CRYPTOCHROME. Science 285: 553–556. MW (2001): A role for the segment polarity
Darlington, TK, Wager-Smith, K, Ceriani, MF, gene shaggy/GSK-3 in the Drosophila circa-
Staknis, D, Gekakis, N, Steeves, TDL, Weitz, dian clock. Cell 105: 769–779.
CJ, Takahashi, JS, Kay, SA (1998): Closing the McNeil, GP, Zhang, X, Genova, G, Jackson, FR
circadian loop: CLOCK-induced transcrip- (1998): A molecular rhythm mediating circa-
tion of its own inhibitors per and tim. Science dian clock output in Drosophila. Neuron 20:
280: 1599–1603. 297–303.
Edery, I, Zwiebel, LJ, Dembinska, ME, Rosbash, Price, JL, Dembinska, ME, Young, MW,
M (1994): Temporal phosphorylation of the Rosbash, M (1995): Suppression of PERIOD
Drosophila period protein. Proc Natl Acad protein abundance and circadian cycling by
Sci (USA) 91: 2260–2264. the Drosophila clock mutation timeless.
Gekakis, N, Saez, L, Delahaye-Brown, A-M, EMBO J 14: 4044–4049.
Myers, MP, Sehgal, A, Young, MW, Weitz, CJ Price, JL, Blau, J, Rothenfluh, A, Abodeely, M,
(1995): Isolation of timeless by PER protein Kloss, B, Young, MW (1998): double-time is a
interaction: Defective interaction between novel Drosophila clock gene that regulates
timeless protein and long-period mutant PERIOD protein accumulation. Cell 94:
PERL. Science 270: 811–815. 83–95.
Glossop, NR, Lyons, LC, Hardin, PE (1999): Renn, SCP, Park, JH, Rosbash, M, Hall, JC,
Interlocked feedback loops within the Taghert, PH (1999): A pdf neuropeptide gene
Drosophila circadian oscillator. Science 286: mutation and ablation of PDF neurons each
766–768. cause severe abnormalities of circadian
Handler, AM, Konpoka, RJ (1979): Transplanta- behavioral rhythms in Drosophila. Cell 99:
tion of a circadian pacemaker in Drosophila. 791–802.
Nature 279: 236–238. Saez, L, Young, MW (1996): Regulation of
Hardin, PE, Hall, JC, Rosbash, M (1990): Feed- nuclear entry of the Drosophila clock pro-
back of the Drosophila period gene product teins period and timeless. Neuron 17: 911–920.
on circadian cycling of its messenger RNA Stanewsky, R, Kaneko, M, Emery, P, Beretta, B,
levels. Nature 343: 536–540. Wager-Smith, K, Kay, SA, Rosbash, M, Hall,
Hunter-Ensor, M, Ousley, A, Sehgal, A (1996): JC (1998): The cryb mutation identifies cryp-
Regulation of the Drosophila protein time- tochrome as a circadian photoreceptor in
less suggests a mechanism for resetting the Drosophila. Cell 95: 681–692.
circadian clock by light. Cell 84: 677–685. Vosshall, LB, Price, JL, Sehgal,A, Saez, L,Young,
Konopka, RJ, Benzer, S (1971): Clock mutants of MW (1994): Block in nuclear localization of
Drosophila melanogaster. Proc Natl Acad Sci period protein by a second clock mutation,
(USA) 68: 2112–2116. timeless. Science 263: 1606–1609.
Konopka, RJ, Wells, S, Lee, T (1983): Mosaic Williams, JA, Sehgal, A (2001): Molecular com-
analysis of a Drosophila clock mutant. Mol ponents of the circadian system in
Gen Genet 190: 284–288. Drosophila. Ann Rev Physiol 63: 729–755.
Kyriacou, CP, Hall, JC (1980): Circadian rhythm Williams, JA, Su, HS, Bernards, A, Field, J,
mutations in Drosophila melanogaster affect Sehgal, A (2001): A circadian output in
short-term fluctuations in the male’s Drosophila mediated by Neurofibromatosis-
courtship song. Proc Natl Acad Sci (USA) 77: 1 and Ras/MAPK. Science 293: 2251–2256.
6729–6733. Zerr, DM, Hall, JC, Rosbash, M, Siwicki, KK
Lee, C, Bae, K, Edery, I (1999): PER and TIM (1990): Circadian fluctuations of period
inhibit the DNA binding activity of a protein immunoreactivity in the CNS and the
Drosophila CLOCK-CYC/dBMAL1 het- visual system of Drosophila. J Neurosci 10:
erodimer without disrupting formation of the 2749–2762.
RHY4 2/6/04 3:54 PM Page 75

4
MOLECULAR ANALYSIS OF
CIRCADIAN RHYTHMS:
NONMAMMALIAN VERTEBRATES
Julie A. Williams

Nonmammalian vertebrates express 䊏 INTRODUCTION


homologs of many of the known clock
genes, although temporal expression varies In previous chapters, we’ve seen that the
across species. Some novel components roles of clock genes have diverged between
of the circadian clock have also been insects and mammals. For example, the
described. The organization of the circadian blue-light photoreceptor, cryptochrome,
system has diverged in nonmammalian ver- mediates responses to light in flies and
tebrates such that the anatomical compo- plants, but appears to have lost this func-
nents of the clock are widespread. In some tion in mammals. Instead, it functions as
birds, for instance, multiple oscillators a key transcriptional repressor in mammals.
contribute to behavioral rhythmicity. In Three period genes have been described in
zebrafish, many tissues contain self- mammals. Although they are each regu-
sustaining oscillators, although the contri- lated in a slightly different manner, none of
bution of each of the tissues to behavioral them alone is necessary for maintaining
rhythmicity is unknown. In addition, rhythmic behavior. In contrast, flies express
because of the multioscillatory feature of one period gene that is absolutely required
the circadian system, preparation of the for circadian behavior. Thus, studying the
pineal or retina in culture has provided an role of each of clock component in lower
in vitro model for the circadian clock. vertebrates may clarify not only how they

Molecular Biology of Circadian Rhythms. Edited by Amita Sehgal


ISBN 0-471-41824-2 Copyright © 2004 John Wiley & Sons, Inc.

75
RHY4 2/6/04 3:54 PM Page 76

76 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS: NONMAMMALIAN VERTEBRATES

have evolved but also their function in the The relative contribution of each of these
circadian system. components to circadian output varies
A common feature of the circadian widely across species, and may even vary
system in mammals and fruitflies is that a within an individual depending on environ-
central oscillator is localized to one par- mental factors such as temperature and
ticular brain region: the suprachiasmatic photoperiod relating to the season.
nucleus and the lateral neurons, respec-
tively. This “core” clock in nonmammalian
The Pineal Gland
vertebrates, in contrast, appears to consist
of multiple tissues that are all integral com- The pineal gland is required for circadian
ponents for circadian behavior, including behavior in most birds. The pineal gland,
the retina, a suprachiasmatic area, and the considered the “seat of the soul” by
pineal gland. These have been studied most Descartes, functions as both a biological
extensively in birds, and are discussed in clock and as a photoreceptor in most non-
the following section. In zebrafish, many mammalian vertebrates. It develops from
organs contain self-sustaining oscillators an evagination of the roof of the dien-
(discussed below), although their respec- cephalon and remains attached to the brain
tive contribution to circadian behavior via a pineal stalk. In some poikilotherms,
remains unknown. Thus, the molecular there is a second component that detaches
components of circadian rhythms are out- from the brain and is located more super-
lined in three types of nonmammalian ver- ficially beneath the skull. In amphibians,
tebrates, birds, zebrafish, and the African this is known as the frontal organ, and in
clawed frog, Xenopus laevis. Because so some lizards, the parietal eye.
little is known about the molecular basis of The primary cell type in the pineal gland
circadian clocks in reptiles, they are not is similar to photoreceptive cells in the
covered in this chapter. retina such that it has an outer segment
with stacked disks containing photopig-
ments. This morphology indicates that the
animal is capable of detecting changes in
䊏 ORGANIZATION OF CIRCADIAN
light in the environment; however it does
SYSTEM IN BIRDS
not have the complex visual acuity of the
eye. The mammalian pinealocytes have lost
Multioscillatory System
this photoreceptive characteristic and are
The circadian system in birds is multioscil- secretory in appearance (Fig. 4.1). Further-
latory. Circadian outputs in avian species more, in lower vertebrates, the photosen-
include rhythms in plasma levels of mela- sory pineal cells make synaptic contacts
tonin, body temperature, and locomotor with neurons that project to the brain. Once
activity such as perch hopping in sparrows. again, this is in contrast to mammals, where
As mentioned above, the organization of innervation of the pineal is largely sympa-
the circadian system is quite different from thetic and there are no sensory contacts
that in mammals in that it is a multioscilla- from the pineal at all.
tory system. There are three major compo- Thus the pineal gland is an ideal model
nents in this system: (1) the pineal gland, for studying circadian clocks in birds
which is the primary site of melatonin syn- and other species. It contains all three
thesis; (2) a hypothalamic area that may components of the circadian system: (1)
correspond to the suprachiasmatic nucleus a photoreceptive input, (2) a central
(SCN) in mammals; and (3) the retina, oscillator, and (3) a regulated output
which is also a site of melatonin synthesis. (melatonin).
RHY4 2/6/04 3:54 PM Page 77

ORGANIZATION OF CIRCADIAN SYSTEM IN BIRDS 77

Nonmammalian Mammalian of two nuclei—the medial SCN (mSCN),


pinealocyte pinealocyte which is located near the preoptic recess of
the third ventricle; and the visual SCN
(vSCN), which is slightly more lateral and
Outer segment caudal to the mSCN. It is interesting to note
that different anatomical and functional
features are associated with each of these
nuclei.
Inner segment The vSCN receives the majority of pro-
jections from the retina via the retinohypo-
Axon thalamic tract (RHT) and may thus be
important for integrating information
about the photoperiod into the circadian
system. This is also supported by the obser-
Figure 4.1. Comparison of a mammalian
versus a nonmammalian pinealocyte. Mam- vation that the vSCN expresses a crypto-
malian pinealocytes are secretory, and no chrome (cCRY2; see text below), which is
neural projections have been found from the known to be a blue-light photoreceptor in
pineal. Nonmammalian pinealocytes resem- plants and flies. Although its photorecep-
ble rod photoreceptors; they contain an tive properties are unknown in mammals,
outer segment with stacked disks, an inner
segment, and neural projections. CRY is an integral part of the circadian
clock in the mammalian system. Communi-
cation between the pineal gland and the
vSCN is also apparent, since this region
In nearly all birds, the pineal gland is expresses melatonin receptors, and lesions
required for rhythms in locomotor activity, of the vSCN abolish rhythmic turnover of
feeding, and body temperature. Pinealec- norepinepherine in the pineal. Finally, the
tomy abolishes these rhythms, however, uptake of 2-deoxy-glucose in the vSCN is
with some variability across species. In rhythmic, which suggests that neural activ-
sparrows, rhythms do not disappear imme- ity in this region oscillates in a circadian
diately and in quail and pigeon, they persist manner.
despite the removal of the pineal gland. The mSCN, on the other hand, expresses
These observations indicate that other clock genes, including Clk, Per2, and Per3.
oscillators can compensate for the loss of Per RNA in this region is induced by
the pineal gland. Rhythmicity in pinealec- light and oscillates in a circadian manner.
tomized birds can be restored with rhyth- Destruction of the mSCN disrupts circa-
mic doses of melatonin either through dian rhythms in locomotor activity in quail
injection, through infusion, or in the drink- and pigeon, indicating that it is a necessary
ing water. Transplantation of the pineal component of the circadian system in these
organ can also restore rhythms. In such species.
cases, the host bird’s rhythm is in phase with Taken together, these observations
that of the donor. suggest a compartmentalization of the
avian SCN. One nucleus, the vSCN, is likely
important for mediating light signals from
The Suprachiasmatic Nucleus
both the retina and the pineal organ, and
The circadian system in birds includes an the other (mSCN) contains a core oscilla-
SCN. The hypothalamic area in birds is tor and likely relays these signals to other
thought to contain a correlate to the parts of the animal to coordinate behavior
mammalian SCN. The avian SCN consists or other rhythmic outputs.
RHY4 2/6/04 3:54 PM Page 78

78 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS: NONMAMMALIAN VERTEBRATES

The Retina Per3 mRNA has so far been described


in detail only in Japanese quail. The RNA
The retina contains an autonomous oscilla-
cycling is similar to that of qPer2, except
tor that also contributes to control of circa-
the rise in mRNA expression is more
dian behavior. For example, the retina in
gradual during the night. Per2 and Per3 are
birds produces melatonin in a rhythmic
expressed throughout the body with
manner. However, it is not released into
highest levels in the retina and pineal gland.
the bloodstream from the retina in all
Expression was also observed in heart,
species. In sparrow and starling, for
lung, kidney, and ovary. Low levels of
instance, plasma melatonin rhythms are
expression were also noted in liver, skeletal
abolished after pinealectomy, indicating
muscle, spleen, and testis. qPer2 and qPer3
that retinal melatonin is not released
cycle in most tissues examined with peaks
into the bloodstream, nor does it contribute
at ZT0, except in the ovary. Unlike qPer2,
to overt locomotor activity rhythms. In
qPer3 is not induced by light.
contrast, simultaneous destruction of the
Clock and two isoforms of Bmal (brain
eyes and the pineal gland in pigeon and
and muscle arylhydrocarbon receptor
quail abolish melatonin rhythms as well as
nuclear translocatorlike protein), Bmal1
behavioral rhythms. However, in the quail,
and Bmal2, were identified in chicken by
severing the optic tract leaves the rhythm
RT-PCR using degenerate primers and sub-
in plasma melatonin intact, but abolishes
sequent screening of a chick pineal cDNA
rhythms in behavior.
library. The cBmal genes had 93 and 65%
identity with the human Bmal1 and Bmal2
Molecular Components of the Avian
genes, respectively. Up to 100% identity
Clock
was observed in the conserved bHLH and
Most of the known genes that are essential PAS domain repeats. cBmal1 and cBmal2
components of the circadian clock have mRNA cycle opposite to the phase of
been identified in at least two avian species, cPer2. cBmal2 is 4 hours delayed relative to
including chicken and Japanese quail. The cBMal1. cClock RNA also exhibits circa-
genes isolated include at least two period dian cycling, although at reduced ampli-
homologs; Per2 and Per3 (each correspon- tude, with peaks occurring at ZT10–18 in
ding to the mammalian counterpart); two LD and CT10–18 in DD (Fig. 4.2).
forms of Bmal identified in chicken (Bmal1 To determine whether these transcrip-
and Bmal2); Clock, and two homologs of tion factors interact, they were subjected to
cryptochrome, cCry1 and cCry2. glutathione-S-transferase (GST) pulldown
In both chickens and quail Per2 RNA assays. This involves expressing at least part
oscillates in a circadian manner with a peak of the protein fused to GST in a bacterial
in the early morning (ZT0 in quail) and a vector.The proteins are extracted from bac-
trough in the early night (Fig. 4.2). In teria, purified, and added to a mixture con-
constant conditions, mRNA oscillations are taining glutathione-Sepharose beads and
dampened, but still significant. RNA cycling an [35S]-methionine-labeled target protein
also persists in chick cultured pineal cells, such as cBmal2. After an incubation period,
with sustained peak expression from the beads are isolated and washed, and
ZT2–6, although it rapidly declines from the material bound to them is subjected
CT2–6 in DD. Further studies show that to SDS-polyacrylamide electrophoresis.
Per2 is rapidly induced by light in both The GST fusion protein (such as GST-
chicken and quail. The mechanism of this cCLOCK) binds to the beads, and if it inter-
induction is discussed in further detail acts with the radiolabeled target protein,
below. then the latter will show up as a single band
RHY4 2/6/04 3:54 PM Page 79

ORGANIZATION OF CIRCADIAN SYSTEM IN BIRDS 79

1.2

1
Relative RNA expression
0.8 Per2
Per3
0.6 Cry1
Cry2
0.4

0.2

0
2 6 10 14 18 22 2
ZT

1.2
Relative RNA expression

0.8 Clock
Bmal1
0.6 Bmal2
cE4bp4
0.4

0.2

0
2 6 10 14 18 22 2
ZT

Figure 4.2. Summary of cycling clock genes in the chicken. mRNA levels for each gene are
plotted versus time of day [Zeitgeber time (ZT)] in an L : D cycle of 12 : 12. Open bars at the
top represent daytime, starting at ZT0 (lights on); and dark bars represent nighttime, start-
ing at ZT12 (lights off). Levels of RNA do not reflect relative expression of the clock genes to
each other, but rather how they change during the day.

on the polyacrylamide gel. Indeed, these sensus site (CACGTG) in the per and tim
assays showed interactions between genes (or per and cry in mammals) to pro-
cBMAL1 and cBMAL2, as well as between mote their transcription. To determine whe-
cCLOCK and each of the two BMAL ther cCLOCK and cBMAL function in a
proteins. similar manner, Dr. Yoshitaka Fukada and
As described in an earlier chapter, the colleagues carried out transcrip-
CLOCK:BMAL heterodimer comprises the tional assays using luciferase reporter con-
positive arm of the circadian autoregulatory structs transfected into human embryonic
feedback loop. It binds to an E-box con- kidney (HEK) 293 cells. The luciferase
RHY4 2/6/04 3:54 PM Page 80

80 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS: NONMAMMALIAN VERTEBRATES

reporter genes contained the E-box element Two cry genes have been identified in
and flanking sequences within the promoter chicken: cCry1 and cCry2. The mRNA of
regions of cPer2, mPer2, or mouse vaso- both genes oscillates in LD (Fig. 4.2), but
pressin genes. In all cases, both cCLOCK: has reduced amplitudes in DD. cCry1
cBMAL1 and cCLOCK:cBMAL2 dimers exhibits peak expression at midday, or
increased luciferase activity in a dose- mid–subjective day in DD, and cCry2 peaks
dependent manner. Interestingly, cBMAL2 during the late night. Northern blot
inhibits transcription at higher doses. analyses revealed that cCry2 expression is
To test the other arm of the clock, or the widespread throughout the animal.
negative component, cPer2 was cotrans- High levels of expression were observed in
fected into the cells at varying doses. pineal and retina, and also in heart, liver,
cPer2 effectively inhibits cCLOCK: skeletal muscle, intestine, and brain.
cBMAL-mediated transcription at the Detailed analysis of tissue showed cCry2
E-box. Taken together, these observations expression in brain regions associated
indicate that all these proteins have a role with phototransduction and the visual
in transcriptional regulation that is consis- system such as the vSCN, optic tectum,
tent with both the mammalian and and the lateral septum. cCry mRNA was
Drosophila models of the circadian clock. also observed in the photoreceptive layer
Additional studies by Fukada’s group of the retina, and in both photoreceptive
indicated that the chicken clock genes func- and interstitial pinealocytes. These data
tion as such in a relevant system. Overex- suggest that cCry may retain some function
pression of cBMAL1 or cBMAL2 in as a photoreceptor. However, transcrip-
cultured pineal cells abolished the rhythm tional assays revealed that both cCry1
of melatonin synthesis in constant condi- and cCry2 repress transcription of cPer2
tions. No effect was observed in LD. Light by inhibiting CLOCK:BMAL-mediated
is known to suppress melatonin synthesis in transcription.
vivo; thus it is possible that this pathway
overrides that driven by cBMAL.
Phase Shifting the Circadian Clock by
Although cryptochrome (cry) gene
Photic Pathways
products across all species examined,
including plants, mammals, insects, avians, A key feature of a circadian clock is the
amphibians, and fish, are structurally ability to reset itself in response to a change
similar, their role in the circadian clock in the environment. The most drastic
has diverged. In flies, for instance, cryp- change that our clocks are subject to is
tochromes are blue-light photoreceptors that which comes with travel over long
important for phase resettting. Absence of distances—jet lag. Over several days, we
cry in Drosophila does not affect activity adjust to a new light–dark schedule. What
rhythms in constant darkness, but does are the mechanisms that mediate this
affect the flies’ phase shifting ability in response? In many organisms kept in con-
response to light. In mammals, on the stant darkness, a single light pulse can reset
other hand, CRY proteins are an integral the phase of activity or the time of day that
component of the core clock as they the organism is active. This is known as a
are required for normal behavioral phase shift.
rhythms and on a molecular level, act as Intuitively, one would think the most
transcriptional repressors by regulating direct way to transmit signal to the clock is
CLOCK:BMAL activity. To date, there is through the retina. However, in birds and
no evidence that supports a role of mCRY other nonmammalian vertebrates, this is
as a photoreceptor. not the case. Extraretinal photoreceptors
RHY4 2/6/04 3:54 PM Page 81

ORGANIZATION OF CIRCADIAN SYSTEM IN BIRDS 81

were first described in birds in the 1960s. cultured pineal cells results in reduced
More recently, a photoreceptive molecule levels of melatonin, but the phase of the
was cloned from chicken pineal gland rhythm of melatonin synthesis remains
and named pinopsin. Pinopsin localized to unchanged. The second response to light
regions of pinealocytes that corresponded described in the pineal is the phase shift in
to the outer photoreceptive segments as the rhythm of melatonin synthesis. It is
described above. Furthermore, pinopsin likely that pinopsin mediates each of
colocalizes with two GTP binding protein these responses through one of these two
a-subunits known to be involved in medi- G proteins.
ating responses to light in the pineal, Gt1a Several other molecules have been iden-
and Gq/11a. Interestingly, each G protein tified as important components of an
is involved in mediating a different kind of entrainment pathway. One of these compo-
light response (Fig. 4.3). One response is nents is mitogen-activated protein kinase
pertussis toxin (PTX)–sensitive and (MAPK). The MAPK protein is constitu-
involves the immediate inhibition of mela- tively expressed in the pineal gland, but the
tonin synthesis in the presence of light. PTX active form of it, phosphorylated MAPK,
inhibits some, but not all, G proteins. Gt1a exhibits a circadian oscillation with peak
is known to be inhibited by PTX, but activity at night. In response to a light pulse,
Gq/11a is not. Thus, application of PTX to MAPK rapidly dephosphorylates within

1.2

No Light Pulse
1
Relative amount of melatonin

+ Light pulse

0.8

0.6

0.4

0.2

0
60 72 84 96 108 120 132 144 156 168 180
Time in culture

Figure 4.3. Effects of light on melatonin release in cultured chick pineal cells. Pineal cells
were maintained in a light–dark cycle in culture for 4 days, and then transferred to constant
darkness on day 5 (indicated by the light–dark bars at the top of the chart). On day 5, pineal
cells in one of two plates received a four-hour light pulse at CT20 (open circles), while the
other plate was kept in the dark (filled circles). Vertical arrow shows the acute affect of light,
where melatonin release is suppressed. Horizontal arrow shows a phase-shift in the melatonin
rhythm as compared to control. [Adapted from Okano and Fukada (2001): Microsc Res Tech
53: 72–80.]
RHY4 2/6/04 3:54 PM Page 82

82 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS: NONMAMMALIAN VERTEBRATES

minutes of the onset of the light pulse. a core clock component (Fig. 4.4). From the
In addition, application of PD98059, an observations discussed above, cE4bp4 may
inhibitor of a MAPK kinase (MEK), which serve two roles in the clock:
is responsible for the phosphorylation
or activation of MAPK, phase-shifts the 1. Since cE4bp4 mRNA expression
rhythm of melatonin production in cultured (and likely protein expression) is
chick pineal cells. This effect is both dose- increasing during the day when cPer2
dependent and time-dependent. Together, mRNA is decreasing, one role of
these observations indicate a role of MAPK cE4bp4 may be to down regulate
in an entrainment mechanism. cPer2 in a circadian fashion.
In an attempt to identify other genes 2. Possibly more importantly, both
involved in phase resetting, Dr. Fukada cE4bp4 and cPer2 are induced by
and colleagues used a technique known light; thus cE4bp4 may act to
as differential display PCR (DD-PCR). curtail the response to light by
DD-PCR involves a low-stringency ampli- repressing (and thereby “slowing”)
fication of RNA collected from different cPer2 induction.
conditions. cDNA fragments that are dif-
ferentially expressed are isolated, reampli- Another interesting product that was
fied, and cloned for further analysis. In this isolated from the DD-PCR screen is
case, RNA was collected from pineal glands cHsp90a. This gene increases within 5
that were isolated from chicks exposed to minutes after light onset, and peaks at 90
light pulses at various times throughout the minutes to fourfold of baseline levels.
day. Of approximately 5700 PCR products, cHsp90a mRNA cycles during the day with
99 were affected by light. So far, two of a peak in the early morning. The cycling
these products have been described in persists in DD, but at a lower amplitude.
detail. The cHsp90a protein colocalizes with
One product was a chicken homolog pinopsin in pinealocytes and was found in
of a bZIP transcription factor, E4bp4 both cytoplasm and nucleus, but no cycling
(cE4bp4). Among all Drosophila se- in subcellular localization was detected. To
quences, this gene had highest similarity to date, the role of cHsp90a in entrainment is
vrille, one of the Drosophila clock genes unknown. It is likely that its cycling and
(see Chapter 3). cE4bp4 mRNA exhibited light-induced expression are related to
a circadian oscillation, and was rapidly circadian and light-induced changes in
induced by light at all times of day tested. temperature that are known to occur in the
Analysis of the 5¢ flanking region of the pineal. The human Hsp90a interacts with
cPer2 gene revealed two consensus cE4bp4 BMAL1 and NPAS2, which indicates some
recognition sites. Using this sequence from role in the central clock.
the cPer2 gene, transcriptional assays were
performed to determine whether cPer
expression is affected by cE4bp4. Indeed, 䊏 ORGANIZATION OF CIRCADIAN
cotransfection of a cE4bp4 expression con-
SYSTEM IN ZEBRAFISH
struct repressed cPer2 expression in a dose-
dependent manner. Interestingly, only one
Locomotor Activity and
of the two cE4bp4 recognition sites is
Melatonin Synthesis
required for this transcriptional repression.
These observations have lead to filling Zebrafish display rhythms of locomotor
in another piece of the clock: a light- activity and melatonin synthesis. Activity
responsive gene that interacts directly with rhythms have been recorded from both
RHY4 2/6/04 3:54 PM Page 83

ORGANIZATION OF CIRCADIAN SYSTEM IN ZEBRAFISH 83

E4BP4
Cytoplasm

E4BP4
CRY
PER

CRY

PER
BMAL
CLK
Nucleus
E Box Cry1,2
E4BP4
E4BP4

BMAL
CLK

LRE E4bp4 E Box Per 2

LRE ClkRE cE4bp4

Figure 4.4. Role of the transcription factor cE4bp4 in the circadian clock. Transcription
of chicken clock genes, per and cry, is mediated by CLOCK and BMAL heterodimers; PER
and CRY proteins form heterodimers and translocate back to the nucleus, where they
inhibit CLOCK:BMAL transcription; Per2 transcription is further repressed by cE4BP4 binding
to a target sequence in the per promoter (LRE = light-responsive element; ClkRE = clock-
responsive element). [Adapted from Doi et al. (2001): Proc Natl Acad Sci (USA) 98: 8089–8094.]

adult and larval zebrafish. Adults placed in nents of the circadian clock. The behavioral
small recording chambers with infrared assay allows for the isolation of mutants
motion sensors display behavioral rhythms displaying altered periods or arrhythmic
with a large amount of variability. About behavior. Other circadian outputs that have
70% of the animals show robust rhythms been recorded in zebrafish include visual
with highest activity during the subjective sensitivity and melatonin synthesis.
daytime. Activity rhythms in larval Rhythms in visual sensitivity have been
zebrafish measured by video image analysis detected using a couple of different assays.
are more robust; 95% of larvae show rhyth- One involves measuring the electroretino-
mic activity, with highest levels recorded gram (ERG), which is a simple field
during the day. In both larvae and adults, potential, measured from the cornea, that
the average period in constant conditions is represents neural activity in the retina.
approximately 25 hours. Another way to quantify visual sensitivity
Because of the availability of mutagene- involves the ability of the fish to respond to
sis and transgenic methods in zebrafish, a threatening visual stimulus. The limit of
this makes an ideal system for delineating detection is measured by the extent to
the function of the molecular compo- which a threatening object is illuminated
RHY4 2/6/04 3:54 PM Page 84

84 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS: NONMAMMALIAN VERTEBRATES

(and therefore visible to the fish) to evoke Molecular Components of the


an escape response. A rhythmic component Zebrafish Clock
has been reported in both assays, with
Clock and Bmal
peaks occurring at subjective dawn, and
troughs at subjective dusk. Zebrafish Most of the clock genes that have been
expressing a dominant mutation, night- identified in zebrafish to date were identi-
blindness b (nbb), are deficient in rhythms fied using either PCR or by low-stringency
of visual sensitivity. Several neuronal screening of a cDNA library. Shortly after
deficiencies were found in nbb mutants, the cloning of the mouse Clock (Clk) gene,
including a reduced number of retinal Paolo Sassone-Corsi and colleagues iso-
dopaminergic interplexiform cells. Indeed, lated the homologue in zebrafish. The
chemical destruction of these cells leads to predicted gene product of zfClk is an
a more profound defect in visual sensitivity 894 amino acid protein that is highly similar
rhythms, indicating a role of the dopamine to that in mouse, sharing 80% identity,
system in the circadian control of visual but it shares only 53% identity with
sensitivity. the Drosophila Clk gene. zfClk contains
Rhythmic melatonin synthesis and a bHLH-PAS DNA binding and
release has been recorded from cultured protein–protein interaction domain, a
zebrafish pineal gland as well as from PAS-B region, and a glutamine-rich region
retina. Melatonin synthesis in culture is corresponding to the transcriptional activa-
light-sensitive, and rhythms persist in con- tion domain, consistent with the Clk pro-
stant conditions for at least a week. This cir- teins found in mouse and fruitfly.
cadian output therefore has the potential Analysis of Clk mRNA expression by
for determining how various mutations can RNase protection assay revealed a robust
affect rhythms at the cellular level. It is oscillation in both the pineal and retina
interesting to note, however, that the con- with peak expression at the onset of the
tribution of the pineal gland or of the retina dark period in an L : D 14 : 10 cycle (Fig.
to activity rhythms has not been deter- 4.5). Clk oscillation persists in DD, indicat-
mined in zebrafish. Preliminary experi- ing that it is likely an integral component
ments show that activity rhythms in of the zebrafish clock. Clk mRNA also
zebrafish are maintained after removal of exhibits circadian oscillations in other
both the pineal gland and the eye. This is tissues, including brain, heart, kidney, and
inconsistent with earlier studies in other spleen. No cycling is observed in testes.
teleosts in which pinealectomy did abolish Interestingly, when various organs were iso-
behavioral rhythms. Nonetheless, these lated in culture, Clk cycling persisted for
data indicate that melatonin is likely not several days in constant darkness. Further
required for the circadian control of behav- studies of heart and kidney organ cultures
ior in zebrafish. As discussed below, tissues showed that Clk expression in these tissues
throughout the zebrafish contain independ- entrains to LD cycles. These observations
ent oscillators that are sensitive to light. indicate the existence of photosensitive
Perhaps the SCN or any one of these peripheral pacemakers in zebrafish, similar
organs is sufficient for driving rhythmic to those observed in Drosophila (see
behavior or compensates for the loss of the Chapter 9 for further discussion of multiple
pineal and/or the eyes. Whatever the case, oscillators).
the question of a central oscillator in In order to identify other components of
zebrafish is an issue that requires further the zebrafish clock, Sassone-Corsi’s group
study. proceeded to isolate partners of the zfClk
RHY4 2/6/04 3:54 PM Page 85

ORGANIZATION OF CIRCADIAN SYSTEM IN ZEBRAFISH 85

1.2

Relative RNA expression


1

0.8
Clock
Bmal1
0.6
Bmal2
0.4

0.2

0
0 2 4 6 8 10 12 14 16 18 20 22 24
ZT

1.2

1
Relative RNA expression

0.8
Per1
0.6 Per2
Per3
0.4

0.2

0
0 3 6 9 12 15 18 21 24
ZT

1.2
Relative RNA expression

1
Cry1a
0.8 Cry1b
Cry2a
0.6 Cry2b
Cry3
0.4 Cry4
0.2

0
0 4 8 12 16 20 24
ZT

Figure 4.5. Summary of cycling clock genes in zebrafish. mRNA levels for each gene
are plotted as described in Figure 4.2. Top panel is mRNA collected from zebrafish eye in a
14 : 10 light : dark cycle. Middle panel is mRNA collected from cultured Z3 cells (see text) in
a 12 : 12 L : D cycle. Bottom panel is mRNA collected from zebrafish eye and whole body in a
14 : 10 L : D cycle.
RHY4 2/6/04 3:54 PM Page 86

86 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS: NONMAMMALIAN VERTEBRATES

protein using the yeast-2-hybrid assay. Per3 mRNA oscillates in embryos in the
This was the same approach used to identify CNS and in the retina from 40 to 128 hours
the CLOCK partner, BMAL, in the mam- post-fertilization, with peaks during the
malian system (see Chapter 5). Two inter- early light phase from ZT0 to ZT4. Inter-
acting partners, BMAL1 and BMAL2, were estingly, Per3 mRNA cycling could be
identified using this method. Alignment of detected in unfertilized oocytes and in
the amino acid sequence of each protein embryos as early as the 1–4 cell stage.
revealed that BMAL1 shared highest iden- The cycling is independent of LD cycles
tity with human BMAL, whereas BMAL2 and persists in constant conditions. These
was more divergent. BMAL1 and BMAL2 data indicated that embryos synchronize
share 75% identity with each other, with the their clocks to the maternal gene products
most divergence at the C terminus. Both in the oocytes.
proteins, consistent with the mammalian zfPer1 and zfPer2 have not been investi-
homolog, contain a bHLH domain and PAS gated in vivo, but have been studied in a Z3
domain repeats.Analysis of protein–protein cell line established from embryonic tissue.
interactions show that BMAL1 interacts When Z3 cells were subjected to LD cycles,
with zfCLOCK more efficiently than does mRNA oscillations of each of the per
BMAL2. BMAL1 and BMAL2 do not inter- genes could be detected; zfPer1 peaked at
act with each other. ZT0, zfPer2 peaked at ZT3, and zfPer3
Bmal1 and Bmal2 mRNA cycle in all peaked between ZT3 and ZT6. The onset
tissues tested, with the exception of the of expression for Per1 and Per3 could be
testes. Interestingly, peak expression varies detected before lights on, but peak per2
both between the Bmal genes and across levels were detected only at ZT3. In
tissues. For example, in the brain, Bmal1 constant darkness, Per2 does not cycle
mRNA peaks at ZT14, and Bmal2 peaks at and has substantially reduced expression.
ZT10. Clk RNA peaks at ZT14–16 in the Oscillations of Per1 and Per3, on the other
brain. In the liver, Bmal1 peaks at ZT9, hand, persisted in DD, with peak expression
and Bmal2 peaks at ZT15, while their Clk occurring at CT3. The onset of the expres-
partner peaks at ZT9–15. These observa- sion increase began shortly before the
tions could indicate that posttranscriptional end of the subjective night, similar to
modifications may occur and vary across the case in LD cycles. Together, these
tissues. We have yet to learn whether the observations indicate that Per1 and Per3
Clk and Bmal proteins cycle and whether are circadian-clock-dependent, and Per2 is
they are in phase with their respective RNA light-dependent.
cycling or with each other.
Interestingly, as described in Chapters 3
Cryptochromes
and 5, Clock, but not Bmal1, cycles in
Drosophila, while the reverse is true in Six CRY genes have been identified in
mammals (i.e., Bmal1 cycles), but Clock zebrafish. Two pairs of these genes cluster
does not. In zebrafish, both are found to together and show high similarity to human
cycle. This may represent an intermediate CRY. These are designated zCry1a, zCry1b,
state in evolution or alternatively reflect a zCry2a, and zCry2b. A fifth cry also shows
specific adaptation in zebrafish. high conservation with the human CRY,
and is named zCry3. The last isolated cry
gene shows fairly low conservation with
Period
both human CRY1 and CRY2 and with a
Zebrafish homologs of each of the three Xenopus photolyase. This one was desig-
mammalian per genes have been cloned. nated zCry4.
RHY4 2/6/04 3:54 PM Page 87

ANALYSIS OF CIRCADIAN RHYTHMS IN XENOPUS 87

The function of the zCrys was deter- clock in mammals (see Chapter 5). In this
mined by using an in vitro luciferase system, rev-erba is positively regulated by
reporter assay. The reporter construct con- CLOCK:BMAL-mediated transcription
tained a promoter region of the mouse argi- and feeds back on the clock by binding to
nine vasopressin gene that carries an E-box a target region in the Bmal1 promoter to
consensus sequence, CACGTG, which is repress its transcription. Thus, rev-erba and
the transcriptional activation site recog- Bmal1 mRNA cycle in opposite phases.
nized by the CLOCK:BMAL heterodimer. As mentioned above, Bmal1 and Bmal2
Transfecting the cells with CLOCK and mRNA cycle with different phases across
BMAL effectively increased the luciferase zebrafish tissues. One prediction is that
reporter activity. However, when cells were rev-erba would also cycle in the opposite
also transfected with zCry1a, zCry1b, phase, depending on the tissue. It is likely
zCry2a, or zCry2b, reporter activity was that widespread expression of rev-erba in
reduced. zCry3 and zCry4 had no effect on zebrafish is also an important contributor to
the luciferase reporter activity. These peripheral oscillators in this system.
data indicate that the zCry1 and zCry2 A second example of a circadian con-
pairs share a similar function with the trolled gene is the interphotoreceptor
mammalian CRY proteins and inhibit retinoid binding protein (IRBP). This
CLOCK:BMAL-mediated transcription. It protein is thought to be involved in retinoid
is possible that zCry3 and zCry4 resemble trafficking between photoreceptors and
the function of the Drosophila CRY, but retinal pigment epithelium. Irbp mRNA
this has yet to be determined. expression is high during the day and low
All the zebrafish Cry RNAs are at night. However, IRBP protein is main-
expressed throughout the body, brain, and tained at constant levels, although turnover
eye, with highest levels of expression in the of the protein is known to be higher during
eye. zCry mRNAs all cycle in both eye and the day. Thus, oscillation in mRNA expres-
brain, but with lower amplitudes in the sion may compensate for this in order to
body. All the Crys cycle with different keep protein levels constant.
phases: Cry1a peaks at ZT0–4, Cry1b peaks
at ZT9–13, both Cry2a and 2b peak at
ZT13–15, Cry3 peaks at ZT23–1, and Cry4 䊏 ANALYSIS OF CIRCADIAN
peaks at ZT9 (Fig. 4.5).
RHYTHMS IN XENOPUS

Others Measuring Rhythms in Frogs


Several other clock-controlled genes have Most of what is known about circadian
been identified in zebrafish. One of these is biology in the African clawed frog,
a member of a ligand-activated nuclear Xenopus laevis, is based on studies in iso-
receptor family of transcription factors, rev- lated retinal cultures. The Xenopus eyecup
erba. Because the ligand of this nuclear preparation has been advantageous in
receptor has not been identified, it is several respects: (1) with the appropriate
considered an orphan nuclear receptor. culture conditions, output rhythms can be
Rev-erba mRNA was reported to cycle in measured from a single eyecup (Fig. 4.6),
zebrafish embryos, with peak expression thus eliminating individual differences that
occurring between ZT20 and ZT0. can occur within a population; and (2) the
Although the role of rev-erba is unknown retina is known to contain all the features
in zebrafish, it was discovered to be an that qualify it as a circadian oscillator. Two
important component of the circadian of these features are an ability to entrain to
RHY4 2/6/04 3:54 PM Page 88

88 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS: NONMAMMALIAN VERTEBRATES

CO2 To fraction collector


O2 Medium

Pump

Eyecup

Figure 4.6. Diagram of the Xenopus eyecup preparation. Culture medium is supplemented
with 5-hydroxy-L-tryptophan in order to detect melatonin from a single eyecup. The medium
is gassed with CO2 and O2, and then delivered to a superfusion chamber containing the
eyecup. The perfusate is delivered to a fraction collector for quantifying concentrations of
melatonin. [Adapted from Anderson and Green (2000): Microsc Res Tech 50: 360–372.]

environmental conditions such as light and melatonin at the beginning of the night. By
a freerunning rhythm in constant condi- the end of the night, melatonin and mela-
tions. Because of the presence of known tonin metabolites start inducing rod disk
clock genes in the retina, this tissue offers a shedding, the peak of which is at the
complete circadian system: an input light-to-dark transition. Dopamine, on the
pathway that mediates signals from the other hand, is released during the day and
environment, a central oscillator, and a reg- induces cone contraction. These processes
ulated output such as melatonin. are known to continue in the absence of a
A number of circadian outputs have light–dark cycle, indicating an underlying
been observed in the retina, including clock-controlled mechanism.
rhythms in the ERG, rhythms in rod disk
shedding and cone elongation, and, as men-
Clock Genes in Xenopus
tioned above, rhythms in melatonin synthe-
sis. The rhythm in melatonin release is Most of the known clock components have
known to be due to circadian control of been identified in Xenopus using RT-PCR
enzymes in the synthetic pathway, such as and subsequent screening of a retinal
N-acetyltransferase (NAT). This enzyme cDNA library. These components include
is also strongly inhibited by light and the positive arm of the feedback loop,
dopamine via D2 receptors. The D2 recep- CLOCK and BMAL, as well as two period
tor is a G-protein-coupled receptor that genes (xPer1 and xPer2) and three cry
negatively regulates adenylyl cyclase and genes as described below. Precisely how
cAMP. Since NAT activity is induced by these components function together to
cAMP, D2 receptor activation results in a produce rhythmic output has not been
decrease in NAT activity by inhibiting determined, but the utility of a unique
cAMP. Thus, light and darkness control the transgenic method called restriction
levels of melatonin and dopamine, and enzyme-mediated integration (REMI) is
both these environmental and neurochem- beginning to uncover how each of these
ical factors can control changes in photo- components is involved.
receptor morphology. For example, cone Xper was identified using degen-
elongation is induced by either darkness or erate primers of mouse and Drosophila
RHY4 2/6/04 3:54 PM Page 89

ANALYSIS OF CIRCADIAN RHYTHMS IN XENOPUS 89

per mRNA. The isolated gene resembles levels in DD. These observations indicate
mPer2, with high homology throughout the that as in zebrafish and birds, xPer2 is reg-
entire protein including the PASB and cyto- ulated by light, and the circadian clock
plasmic localization domains. However, regulates xPer1.
despite the high homology in the Further analysis of the effects of light on
N-terminal region, no bHLH domain was per indicate that xper2 mRNA, in particu-
detected. RT-PCR was performed to deter- lar, shows significant increases 3 hours
mine whether other per genes exist, and a after the onset of light, and peak expression
partial sequence was identified that resem- occurs after 6 hours of lights on. Dopamine
bles the mPer1 protein. Using this partial (DA), a neurotransmitter that is thought
sequence as a probe, as well as a piece of to be a component of the entrain-
the xPer2 gene, expression patterns were ment pathway, had a similar effect on
studied both anatomically and temporally. xper2, where significant induction was not
These studies revealed that xPer1 and detected until 3 hours after dopamine
xPer2 have highest expression levels in the treatment. DA induced xper2 mRNA inde-
retina and show lower expression through- pendent of the time of day. The effect of
out the body, including brain, heart, liver, DA is blocked by the D2 receptor antago-
spleen, and testes. Although tissue expres- nist eticlopride. Interestingly, eticlopride
sion is similar for both mRNAs, temporal does not block xper2 induction by light,
expression is quite different. Both xper indicating a separate pathway. Induction of
RNAs exhibit robust cycling in LD, but with xper1 by either light or DA, on the other
a 4–8-hour phase difference (Fig. 4.7). xPer1 hand, is restricted to the early part of the
continues to oscillate in a circadian manner day. These data support the notion that
in DD. xPer2 does not oscillate in constant xper1 is relatively insensitive to light,
conditions, but is expressed at constant whereas xper2 is affected by phase shifting
peak levels in LL, and at constant trough agents, light, and DA.

1.2

1
Relative RNA expression

0.8 Pe r1
Pe r2
0.6 Cry1
Cry2a
0.4 Cry2b

0.2

0
0 4 8 12 16 20 24
ZT

Figure 4.7. Summary of cycling clock genes in Xenopus. mRNA levels for each gene are
plotted as described in Figure 4.2. mRNA was collected from Xenopus retina in L : D 12 : 12.
xClock mRNA does not cycle and is therefore not plotted here (see text).
RHY4 2/6/04 3:54 PM Page 90

90 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS: NONMAMMALIAN VERTEBRATES

Although studies have shown how the tional transgenesis involves injecting
xpers are regulated (one having a circadian plasmid DNA into embryos and looking for
component, and the other by the entrain- surviving adults expressing the transgenic
ment pathway), there are no data available DNA. In frogs, particularly in Xenopus
showing whether these genes are necessary laevis, this technique is problematic in that
for circadian behavior. As discussed above, the gestation period is lengthy, and success-
dopamine, itself released in a circadian ful insertion into the genome is fairly infre-
manner, exerts effects in both melatonin quent. If the transgene is not inserted
synthesis and photoreceptor morphology. into the genome, promoter-driven spatial
The observation that it also affects a known expression does not occur with adequate
clock gene suggests an important interac- fidelity, and the embryo often expresses
tion between the clock and this neuro- the transgene in a mosaic pattern. REMI,
chemical system. on the other hand, involves introducing
Three cry genes have been identified plasmid DNA into the male genome. Low
in Xenopus. xCry1 has 86% identity with concentrations of restriction enzyme are
mCry1 and exhibits the highest amplitude used to integrate the transgene into the
in mRNA oscillation with a peak at ZT16. frog sperm. Afterward, the sperm nuclei are
The two other crys, xCry2a and xCry2b, are transplanted into unfertilized eggs, and
90% similar to mCry2. It is possible that transgenic embryos are produced.
xCry2a and xCry2b are duplicate versions Dr. Green’s group designed a transgene
of the same gene, since full sequences that mimicked the Clkjrk mutation
were not identified, particularly the 5¢ end. described in both Drosophila and mouse.
However, both mRNAs oscillate in both This gene was designed to express all the
LD and DD, but are 4 hours out of phase functional domains of the CLOCK protein,
with each other. Furthermore, all three crys except for that needed for transcriptional
are expressed in the same tissues, including activation. The expressed protein therefore
retina, brain, heart, liver, spleen, and testes. competes with the endogenous protein
xCry2b expression is relatively weak, and by binding with partners (such as BMAL)
xCry1 expression is higher in the brain as required for transcriptional activation.
compared to the other cry genes, indicating Functional assays in vitro using an mper1-
a tissue-specific function. luciferase reporter showed that the
xClock is found in many tissues, includ- mutated xClk could effectively block tran-
ing retina, brain, heart, liver, muscle, spleen, scription induced by a wild-type Clk. When
and testes. Similar to the mammalian coun- the transgene was expressed in frog retina
terpart, xClk contains bHLH, PASA, and using the eye-specific promoter, IRBP,
PASB functional domains, with 94% simi- melatonin cycling was abolished in a dose-
larity in these regions. Consistent with its dependent manner. These data were the
role as a transcription factor, xClk contains first evidence that xClk is required for
a glutamine-rich region or a transcriptional circadian regulation of an output. Overall
activation domain at the C terminus. xClk levels of melatonin were not affected, nor
mRNA does not cycle in the retina in either were levels of other genes in the melatonin
LD or DD. synthetic pathway, although temporal
Studies of xClk have revealed a similar expression of these genes in the presence
function for this protein in clock mecha- of the mutant xClk transgene was not
nisms. Using the method mentioned above determined.
(REMI), Dr. Carla Green and co-workers Other clock molecules have been identi-
produced a transgenic frog expressing a fied in Xenopus, one of which is currently
dominant negative form of xClk. Tradi- being investigated in mammals and insects.
RHY4 2/6/04 3:54 PM Page 91

FURTHER READING 91

Nocturnin (noc) was identified as a cycling ing input mechanisms, oscillator mecha-
transcript in a differential-display PCR nisms, and especially outputs. How does the
screen. Noc mRNA expression peaks at molecular clock signal to the rest of the cell
night, accounting for its name. Sequence and the entire organism to produce rhyth-
analysis revealed that the noc gene encodes mic behavior? With a variety of measurable
a protein containing a leucine repeat outputs available in the in vitro prepara-
domain and a domain homologous to the tions such as the Xenopus eyecup or chick
yeast carbon catabolite repression 4 protein pineal gland, these are ideal systems for
(CCR4), which is a transcription coactiva- addressing these issues.
tor. Noc mRNA expression is restricted
to the Xenopus retina. Interestingly, noc
expression in the mouse is widespread. Not FURTHER READING
only is it expressed in most tissues exam-
ined, but it also cycles in nearly all tissues Anderson, FE, Green, CB (2000): Symphony of
in a circadian manner. Thus, noc is likely an rhythms in the Xenopus laevis retina. Micro
important component that mediates a cir- Res Tech 50: 360–372.
cadian output. Whether it affects behavior Bailey, MJ, Chong, NW, Xiong, J, Cassone, VM
or some other physiological process (2002): Chickens’ Cry2: Molecular analysis of
remains to be determined. an avian cryptochrome in retinal and pineal
photoreceptors. FEBS Lett 513: 169–174.
Cahill, GM (2002): Clock mechanisms in
zebrafish. Cell Tissue Res 309: 27–34.
䊏 CONCLUSIONS Cahill, GM, Hurd, MW, Batchelor, MM (1998):
Circadian rhythmicity in the locomotor
Although the clock machinery in nonmam- activity of larval zebrafish. Neuroreport 9:
malian vertebrates is fairly conserved with 3445–3449.
that in mammals, the organization of their Cermakian, N, Whitmore, D, Foulkes, NS,
circadian system is more diverse.The model Sassone-Corsi, P (2000): Asynchronous
systems outlined above have been useful in oscillations of two zebrafish CLOCK part-
identifying novel components of the circa- ners reveal differential clock control and
dian clock such as cE4bp4 and nocturnin, function. Proc Natl Acad Sci (USA) 97: 4339–
4344.
although a role for the latter in the circa-
Delaunay, F, Thisse, C, Marchand, O, Laudet, V,
dian clock or its output has yet to be deter- Thisse, B (2000): An inherited functional
mined. Several questions have yet to be circadian clock in zebrafish embryos. Science
addressed in these systems. First, as men- 289: 297–300.
tioned above, some of the clock genes cycle Doi, M, Nakajima, Y, Okano, T, Fukada, Y
in these organisms, while in others they do (2001): Light-induced phase-delay of the
not. In the previous chapter, we have seen chicken pineal circadian clock is associated
that mRNA cycling may not be necessary with the induction of cE4bp4, a potential
for driving rhythmic behavior, but protein transcriptional repressor of cPer2 gene. Proc
cycling is required. Thus, the regulation of Natl Acad Sci (USA) 98: 8089–8094.
the various clock proteins needs to be Doi, M, Nakajima, Y, Okano, T, Fukada, Y
(2002): Light-dependent changes in the chick
addressed in these nonmammalian systems.
pineal temperature and the expression of
Furthermore, a key advantage to study- cHsp90a gene: A potential contribution of in
ing nonmammalian vertebrates is the avail- vivo temperature change to the photic-
ability of in vitro preparations that contain entrainment of the chick pineal circadian
a complete circadian system. Many ques- clock. Zool Sci 19: 633–641.
tions in the field remain with respect to Green, CB, Besharse, JC (1996): Identification of
every aspect of the circadian clock, includ- a novel vertebrate circadian clock-regulated
RHY4 2/6/04 3:54 PM Page 92

92 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS: NONMAMMALIAN VERTEBRATES

gene encoding the protein nocturnin. Proc chick pineal clock oscillation. J Neurosci 20:
Natl Acad Sci (USA) 93: 14884–14888. 986–991.
Gwinner, E, Brandstätter, R (2001): Complex Steenhard, BM, Besharse, JC (2000): Phase shift-
bird clocks. Phil Trans Roy Soc Lond B 356: ing the retinal circadian clock: xPer2 mRNA
1801–1810. induction by light and dopamine. J Neurosci
Hayasaka, N, LaRue, SI, Green, CB (2002): In 20: 8572–8577.
vivo disruption of Xenopus CLOCK in the Underwood, H, Steele, CT, Zivkovic, B (2001):
retinal photoreceptor cells abolishes circa- Circadian organization and the role of the
dian melatonin rhythmicity without affect- pineal in birds. Microsc Res Tech 53: 48–
ing its production levels. J Neurosci 22: 62.
1600–1607. Wang, Y, Osterbur, DL, Megaw, PL, Tosini, G,
Hurd, MW, Debruyne, J, Straume, M, Cahill, GM Fukuhara, C, Green, CB, Besharse, JC (2001):
(1998): Circadian rhythms of locomotor Rhythmic expression of Nocturnin mRNA in
activity in zebrafish. Physiol Behav 65: multiple tissues of the mouse. BMC Dev Biol
465–472. 1: 9.
Kobayashi, Y, Ishikawa, T, Hirayam, J, Daiyasu, Whitmore, D, Foulkes, NS, Sassone-Corsi, P
H, Kanai, S, Toh, H, Fukuda, I, Tsujimura, T, (2000): Light acts directly on organs and cells
Terada, N, Kamei, Y, Yuba, S, Iwai, S, Todo, T in culture to set the vertebrate circadian
(2000): Molecular analysis of zebrafish clock. Nature 404: 87–91.
photolyase/cryptochrome family: Two types Whitmore, D, Foulkes, NS, Strähle, U, Sassone-
of cryptochromes present in zebrafish. Genes Corsi, P (1998): Zebrafish clock rhythmic
Cells 5: 725–738. expression reveals independent peripheral
Li, L, Dowling, JE (2000): Disruption of the circadian oscillators. Nature Neurosci 1:
olfactoretinal centrifugal pathway may relate 701–707.
to the visual system defect in night blindness Yamamoto, K, Okano, T, Fukada, Y (2001):
b mutant zebrafish. J Neurosci 20: 1883– Chicken pineal Cry genes: Light dependent
1892. up-regulation of cCry1 and cCry2 transcripts.
Li, L, Dowling, JE (2000): Effects of dopamine Neurosci Lett 313: 13–16.
depletion on visual sensitivity of zebrafish. Yoshimura, T, Suzuki, Y, Makino, E, Suzuki, T,
J Neurosci 20: 1893–1903. Kuroiwa, A, Matsuda, Y, Namikawa, T,
Natesan, A, Geetha, L, Zatz, M (2002): Rhythm Ebihara, S (2000): Molecular analysis of
and soul in the avian pineal. Cell Tissue Res avian circadian clock genes. Mol Brain Res
309: 35–45. 78: 207–215.
Okano, T, Yamamoto, K, Okano, K, Hirota, T, Yoshimura, T, Yasuo, S, Suzuki, Y, Makino, E,
Kasahara, T, Sasaki, M, Takanaka, Y, Fukada, Yokota, Y, Ebihara, S (2001): Identification of
Y (2001): Chicken pineal clock genes: the suprachiasmatic nucleus in birds. Am J
Implication of BMAL2 as a bi-directional Physiol 280: R1185–R1189.
regulator in circadian clock oscillation. Zhu, H, Green, CB (2001): Three cryptochromes
Genes Cells 6: 825–836. are rhythmically expressed in Xenopus
Okano, T, Fukada, Y (2001): Photoreception and laevis retinal photoreceptors. Mol Vision 7:
circadian clock system of the chicken pineal 210–215.
gland. Microsc Res Tech 53: 72–80. Zhu, H, LaRue, S, Whiteley, A, Steeves, TDL,
Pando, MP, Pinchak,AB, Cermakian, N, Sassone- Takahashi, JS, Green, CB (2000): The
Corsi, P (2001): A cell-based system that Xenopus Clock gene is constitutively
recapitulates the dynamic light-dependent expressed in retinal photoreceptors. Mol
regulation of the vertebrate clock. Proc Natl Brain Res 75: 303–308.
Acad Sci (USA) 98: 10178–10183. Zhuang, M, Wang, Y, Steenhard, BM, Besharse,
Sanada, K, Hayashi, Y, Harada, Y, Okano, T, JC (2000): Differential regulation of two
Fukada, Y (2000): Role of circadian activa- Period genes in the Xenopus eye. Mol Brain
tion of mitogen-activated protein kinase in Res 82: 52–64.
RHY5 2/6/04 3:55 PM Page 93

5
GENETIC BASIS FOR CIRCADIAN
RHYTHMS IN MAMMALS
J. D. Alvarez

䊏 INTRODUCTION humans and a mutation in a circadian


gene has been established underlining the
In this chapter, studies of the molecular importance of the field.
basis of mammalian circadian rhythms are Using classical genetics to study mam-
discussed. At the time of this writing, the malian circadian systems is not an easy task
field is advancing with lightning speed; for a number of reasons. The generation
therefore, by the time readers set their eyes time of mammals is long, especially com-
on this book, much more information may pared to model systems such as Drosophila
be known. Still, a discussion of how mam- sp. or C. elegans. Another reason is purely
malian genetics and molecular biology logistical—conducting large-scale mutation
has contributed to a basic understanding studies on mammals requires an enormous
of chronobiology is quite useful. Utilizing amount of space and manpower to handle
insect genetics to dissect circadian systems the numbers of necessary animals. Still, the
has been enormously successful, and we wealth of knowledge that will result from
now know that there is great conservation such studies is so vast that a number of
of the basis for circadian rhythms among all groups have initiated large-scale mutagen-
species, including mammals. Mammals are esis screens in mice. Indeed, it is clear that
quite a bit different from fruitflies, though, when applied, forward genetics can be very
with unique and complex physiologic out- successful as demonstrated by the elegant
puts. A link between a genetic disease in identification of the Clock gene in mice, the

Molecular Biology of Circadian Rhythms. Edited by Amita Sehgal


ISBN 0-471-41824-2 Copyright © 2004 John Wiley & Sons, Inc.

93
RHY5 2/6/04 3:55 PM Page 94

94 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

first cloned mammalian circadian gene. Components of a circadian system


Additionally, sometimes Mother Nature is
kind and presents us with a naturally occur-
ring genetic mutant that affects circadian
rhythms; the best example is the tau muta-
tion in hamsters. Furthermore, although
Input Core oscillator
mammalian genetic studies are not as easy
as those in lesser organisms, the use of
transgenic and gene-targeting technology Output
in mice allows us great control over the
genetics in a mammalian system. Such Figure 5.1. Schematic of the three-part
model of a circadian system. At the heart of
manipulation is already beginning to show the system is the central oscillator. This is the
us the effects of loss of function mutations main timekeeper of the system. The central
of core circadian genes in the murine oscillator is under the influence of input
system. Hopefully, transgenic technology factors. Input is often from environmental
will soon allow us to examine gain of func- factors, such as the light–dark cycle. The
central oscillator imposes a rhythm on
tion mutants in these animals as well. In this various aspects of physiology via output
chapter, we discuss primarily studies that pathways. Output can represent both direct
have been performed in rodents, particu- neuronal connections and secreted factors.
larly mice, because it is in this model system
that most of research on mammalian circa-
dian biology has been performed, particu- systems operate in concert with each other
larly with respect to molecular biology. is a very active area of research.
Currently (as of 2003), it appears that the The central oscillator is the time-
system is conserved in all mammals, but the keeper—it maintains the rhythm. In the
reader should note that such an assumption mammalian core circadian system, the
is not completely proven yet. central oscillator is located in a paired
cluster of neurons in the brain called the
suprachiasmatic nuclei (SCN). Output from
the SCN represents the mechanisms by
䊏 PROPERTIES OF A MAMMALIAN
which this core central oscillator regulates
CIRCADIAN SYSTEM the physiology of the whole organism.
Examples of such outputs include the
Components
sleep–wake cycle and the daily activity of
Historically, before the advent of gene rodents as measured by running on wheels
cloning, most research in mammalian circa- (see text below). The central oscillator is
dian biology focused on phenomenology under the influence of the input, which
and anatomy. These studies found that cir- are factors that can reset or “entrain”
cadian systems in mammals consist of three the central oscillator. The most notable
components, just as in nonmammals. These example of an input factor is light, which is
components are the input, the output and detected by the retina of the eye and trans-
the central oscillator (Fig. 5.1). In mammals, duced to the SCN via direct neural connec-
a core circadian system is responsible for tions. An example of entrainment is “jet
maintaining the rhythms of the entire lag,” a condition initially experienced by
organism. However, circadian systems exist international travelers when changing time
in nearly every organ of the body. In fact, zones, following which the central oscillator
evidence is mounting that circadian systems becomes reset or entrained to the new
exist in individual cells. How all these light–dark cycle after about a week.
RHY5 2/6/04 3:55 PM Page 95

PROPERTIES OF A MAMMALIAN CIRCADIAN SYSTEM 95

Numerous studies have characterized the 4. Although mammals are able to regu-
effects of many different environmental late their body temperature, studies
factors on circadian rhythms, and have led performed with cultured mammalian
to a defined set of properties that charac- cells indicates that mammalian
terize these rhythms. rhythms display the requisite prop-
erty of temperature compensation, in
that they are able to maintain a con-
Circadian Properties Common to
stant period over a wide temperature
All Species
range.
Circadian rhythms display properties that
are conserved across species. Mammalian
Physiologic Aspects
circadian clocks are characterized by the
same properties displayed by clocks in all Circadian clocks control multiple aspects of
organisms: physiology. Although we have established
rules for circadian rhythms in mammals, we
1. First and foremost, they cycle with have yet to discuss what activities are actu-
close to a 24-hour period, although ally under circadian control. These output
there is variability in period length activities allow us to observe the clock in
among different species. action, and to determine the effects of envi-
2. As mentioned above, mammalian ronmental conditions and genetic muta-
circadian clocks can be “entrained” tions on clock function. In truth, it is almost
by environmental cues. Indeed, a harder to find a physiologic output that
single episode of environmental per- does not show a daily variation than one
turbation is enough to shift the phase that does. Of note, the word “daily” is used
of the circadian clock. For instance, a here because in many instances, the
brief light pulse given to an animal observed rhythm has been studied only in
during the normal “night” time will the presence of environmental changes,
shift the phase of the circadian such as light–dark cycles. Therefore, until
rhythm. If the light pulse is given studies are performed under constant con-
early in the night, the phase will be ditions, we should reserve use of the word
delayed, and if given later in the “circadian.” Nevertheless, there are some
night, the phase will be advanced. well-studied examples of circadianly con-
Moreover, varying the intensity of trolled physiologic outputs.
light to which an animal is exposed
affects the freerunning period of the
Rest–Activity Cycle
animal. Other environmental factors
that phase-shift circadian rhythms The rest–activity rhythm is the classic
include social cues and regulated example of a mammalian circadian rhythm.
feeding times. Mammals show sustained bouts of activity
3. Mammalian circadian clocks are and inactivity (rest) that follow a circadian
endogenous. In other words, they will rhythm. Normally, these bouts correspond
persist in the absence of environmen- to times when the animal is either asleep
tal cues, called “freerunning” condi- or awake. However, the rest–activity
tions. This property is demonstrated rhythm is separate from the sleep–wake
in mammals by the fact that the cir- cycle. The best example of a rest–activity
cadian rhythm continues even when rhythm is that of wheel running by rodents
the organism is housed in constant kept in cages, also called a locomotor
darkness. activity rhythm. Rodents maintained on a
RHY5 2/6/04 3:55 PM Page 96

96 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

light–dark cycle will immediately begin yielding insight into the genetic basis of
running on a wheel on lights off, and they sleep regulation and is discussed in more
will stop running on lights on. This activity detail later in this chapter.
cycle shows a very precise rhythm of 24
hours, and continues in constant darkness
Temperature
although with the endogenous periodicity
of the animal rather than the imposed 24- Although mammals are warm-blooded,
hour period (Fig. 5.2). As discussed below, their core body temperature does vary with
this rhythm is useful for the identification the time of day. The peak is normally during
of circadian mutants. the animal’s active phase, and the trough is
during the rest period. Generally, the dif-
ference is only a few degrees centigrade,
Sleep–Wake Cycle
but it is reproducible, is endogenous and
The sleep–wake cycle is probably the can be entrained, making it a true circadian
most obvious of the circadian rhythms in rhythm.
mammals. As noted above, this cycle is
separate from rest–activity rhythms. Under
Endocrine Function
some conditions, mammals will sleep when
they are normally active and may be awake The levels of numerous hormones in
when they normally rest. Exciting work is mammalian serum vary with defined daily

7 pm 7 am 7 pm 7 am

Day
1

3
LD
4

10
DD
11

12

13

14

Figure 5.2. Schematic of activity rhythms of a mouse as measured by running on a wheel.


The bar at the top shows when lights are on (light bar) and off (dark bar). As indicated, the
lights come on at 7 am and turn off at 7 pm. This is a standard 12-hour light–dark (LD) cycle.
Activity of the mouse is indicated by the dark bars for each day. Note that the data are double-
plotted; that is, day 1 is shown on the first line, followed by day 2. The second line starts with
day 2 and ends with day 3. This allows a comparison of days both horizontally and vertically.
At the end of day 6, the animal is shifted from an LD cycle to constant darkness (DD). This is
called freerunning conditions. Note that the periods of activity shift slightly in constant dark-
ness. This is due to the fact that the endogenous periodicity of most mouse strains is slightly
shorter than 24 hours.
RHY5 2/6/04 3:55 PM Page 97

LIGHT: THE PRIMARY INPUT FACTOR CONTROLLING MAMMALIAN CLOCKS 97

variations. Some of the best studied are year. An awareness of the time of year
melatonin and glucocorticoids. These are requires measurement of day length,
discussed further below. Also, aldosterone which again is a function related to circa-
and renin, responsible for salt and water dian regulation.
retention, are circadianly regulated which
may account for observed circadian func-
tions of the kidney. 䊏 LIGHT: THE PRIMARY INPUT
FACTOR CONTROLLING
Reproduction MAMMALIAN CLOCKS
The levels of most reproductive hormones,
Light Detection by the Retina
such as testosterone, are circadianly regu-
lated in mammals. However, most rhythms Input in mammals consists primarily of
governing reproduction are primarily infra- light. The eye is responsible for light detec-
dian (>24 hours). Still, the circadian system tion; enucleation of various mammals
does play a major role in regulating these disrupts circadian responses to light. The
infradian rhythms. One such rhythm is retina is the part of the eye that detects
the estrus cycle of a rat, which is 4 days light. Photic signals from the retina are
long. Each day brings defined changes in conveyed to the SCN directly via a neural
hormone levels and in the response of the pathway called the retinohypothalamic
female reproductive tract. The changes of tract (RHT). Additionally, an indirect
each day occur at the same time of that day. neural pathway courses through the inter-
For example, ovulation occurs at the same geniculate leaflet (IGL) (Fig. 5.3). Still, the
time of day each estrous cycle, so it is clear big questions are what retinal cell, and what
that circadian rhythms influence ovulation. molecule in that cell, act to detect light and
Additionally, many mammals are reproduc- to convey this information to the clock? In
tively capable only at specific times of the other words, what is the circadian photo-

IGL

Light

Retina
GHT
SCN

RHT

Eye

Figure 5.3. Schematic of photic input pathways to the clock. Light from the environment is
detected by cells of the retina. This photic signal is transduced to the SCN through two main
pathways. The first is direct, which connects the retina to the SCN via the retinohypothalamic
tract (RHT). The second pathway is indirect and first connects to the intergeniculate leaflet
(IGL), which in turn connects to the SCN via the geniculohypothalamic tract (GHT).
RHY5 2/6/04 3:55 PM Page 98

98 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

receptor? With respect to vision, there are Cones


two types of photoreceptor cells in the
There are no naturally occurring mutant
retina: rods and cones. Rods are the
mice that completely lack cones. However,
primary photoreceptor and contain
mice that lose their cones were produced
the photopigment rhodopsin. Rods are
using transgenic methodology. These mice
responsible for vision in dim light. Cones
express the cl transgene, which contains the
contain the photopigment opsin. Cones
diphtheria toxin gene under the control of
are responsible for color vision and for
the human opsin promoter. As is the case
vision in bright light. Frustratingly, neither
with the rodless mutant mice, cl mice do not
rods nor cones are necessary for circadian
show deficits in circadian photoreception.
photoreception.
Finally, by breeding cl mice with either rd
or rdta mice, mouse lines that completely
lack both rods and cones were produced.
Rods and Cones
However, these double mutant lines are
Genetic mutations are useful in studies normal in circadian photoreception, further
directed at identifying the circadian underlining the conclusion that neither
photoreceptor. These mutations have rods nor cones are necessary for circadian
allowed us to exclude certain candidates photoreception.
but unfortunately have not fingered a
culprit yet.
Novel Photopigments
Because neither rods nor cones are the
Rods answer to the circadian photoreceptor
The naturally occurring mouse mutant, quest, it is likely that other retinal cells
retinal degeneration (rd), carries an auto- contain novel photopigments that are the
somal recessive mutation that results in the true circadian photoreceptor. A number of
complete loss of rods by 10 weeks of age. interesting candidates have been described,
These mice also lose a majority of their but, to date, none have been specifically
cones, and are blind. Despite these losses, rd linked to circadian photoreception in
mice are normally responsive to light with mammals.
respect to circadian responses. Another
natural mutation, retinal degeneration slow
Cryptochrome
(rds), also leads to massive rod and cone
loss, but, like rd, does not appear to affect One interesting development was the dis-
circadian responses to light. Additional covery of mammalian cryptochrome (cry)
information comes from studies using mice genes. These genes code for putative flavin
that express the rdta transgene. This trans- binding proteins, which, in other systems,
gene contains the diphtheria toxin gene are responsive to blue light and to UVA
under the control of the human rhodopsin light. This type of photopigment was origi-
promoter. These rdta transgenic mice have nally identified in plants, but homologs in
a massive early loss of rods coupled with a animals exist. In insects, there is a single
slow degeneration of cones. Despite these cryptochrome gene that appears to repre-
losses, rdta mice are still responsive to light. sent the true circadian photoreceptor. In
Taken together, these studies indicate that mammals, however, the story is not as clear
rods are not solely responsible for circadian because the cry genes are apparently part
photoreception. of the central clock. A more in-depth dis-
RHY5 2/6/04 3:55 PM Page 99

LIGHT: THE PRIMARY INPUT FACTOR CONTROLLING MAMMALIAN CLOCKS 99

cussion of the cry genes is found later in this pathways responsible for light responsive-
chapter. ness is actively being pursued. Like many
aspects of circadian biology, however, much
clarification is still necessary.
Melanopsin
Recall that photic signals from the retina
Melanopsin is a novel opsin-related pho- are conveyed along the RHT to the SCN.
topigment that is found in a subset of The primary neurotransmitter of this
retinal ganglion cells (RGCs). RGCs act pathway is glutamate. On reaching the
primarily as relay cells that convey infor- SCN, the glutamatergic signal activates a
mation from rods and cones to various glutamate specific receptor called the
parts of the brain involved in vision. Some N-methyl-d-aspartate (NMDA) receptor.
of these RGCs connect with the SCN. It has Activation of this G-protein-coupled recep-
been found that the subset of RGCs that tor leads to an increase in intracellular
connect with the SCN is the same subset of calcium, which in turn activates mitogen-
RGCs that contain melanopsin. Further- activated protein kinase (MAPK). Activa-
more, these melanopsin-containing RGCs tion of this kinase leads to phosphorylation
are responsive to light. In fact, these cells of a transcription factor called cyclic AMP
are not involved in image formation, but response element binding (CREB) protein.
rather are sensitive to the level of light, The phosphorylated form of CREB is the
called luminance. Tying these lines of evi- active form. Strikingly, CREB activity is
dence together leaves us with a small group upregulated by a light pulse given during
of melanopsin containing RGCs that are the subjective night, but not during the sub-
responsive to luminance and also synapse jective day, indicating that CREB is part
with neurons in the SCN. However, mice of the signal transduction pathway that
lacking melanopsin are still responsive to mediates phase resetting. Binding sites for
light. CREB are found in the promoters of some
Of course, there may not be a single cir- circadian genes that are core components
cadian photoreceptor in mammals. It is pos- of the circadian clock. These genes, called
sible that this system is so important that Period-1 (Per1) and Period-2 (Per2),
redundancy is in place. In other words, cryp- are normally induced by light pulses
tochromes and melanopsin, and possibly given in the middle of the night and are
others, may all be involved. Indeed, mice discussed in further detail below (see also
that lack cryptochromes and rods and cones Fig. 5.5).
are partially impaired in their responses to Adding complexity to the circadian
light. Moreover, mice that lack rods, cones, response to light is the fact that both CREB
and melanopsin completely lack photic activity and MAPK activity are normally
responses. Therefore, redundancy does circadianly regulated in the SCN. The use of
appear to function in mammalian circadian antibodies specific for the phosphorylated
photoreception. form of CREB demonstrated that the level
of active (phosphorylated) CREB is rhyth-
mic in the SCN. The active form of CREB
Circadian Response to Light
is at its highest level in the middle of
Light entrains the clock, and a light pulse the night. Correspondingly, the activity of
given to an animal during the night resets CREB in the SCN is also circadianly regu-
the clock by either delaying or advancing lated, as demonstrated using a transgenic
the phase of the circadian rhythm (Fig. 5.4). reporter construct that is responsive to acti-
Characterization of the signal transduction vated CREB. Expression of the reporter
RHY5 2/6/04 3:55 PM Page 100

100 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

7 pm 7 am 7 pm 7 am

Day
1

2 LD
3

6
Phase 7
delay
8

10
DD
11
Phase 12
advance
13

14
15

16

Figure 5.4. Schematic of activity rhythms of a mouse showing effects of a light pulse to gen-
erate either a phase delay or a phase advance. The mouse is first entrained to a LD cycle and
then allowed to freerun in constant darkness. When a light pulse (indicated by the upside-
down triangle) is administered at the beginning of the night, a phase delay occurs as indi-
cated by the delay in the active period during the next night. The overall period stays the
same, however. Similarly, a light pulse administered late in the subjective night results in a
phase advance as indicated by the earlier onset of activity. Again, note that the overall period
stays the same. Often at the onset of a phase change, there is a transient period of near
arrhythmicity, which is not illustrated here. Presumably, this occurs while the clock is being
“reset.”

construct was highest in the middle of the additional signaling cascades may be at
day and lowest in the middle of the night. work.
The lag time between the peak of active Light responses of the clock are even
CREB levels (middle of the night) and the more complex than indicated above
peak of reporter gene expression (middle because there may be more than one
of the day) is due to the fact that it takes pathway at work. When a light pulse is
about 12 hours to see the full effect of tran- given to an animal at the beginning of the
scriptional activity. MAPK activation is also night, a phase delay occurs. The pathway
endogenously rhythmic in the SCN. Like outlined above is concerned primarily with
CREB, MAPK can be phosphorylated, such delays. In contrast, a light pulse deliv-
and this phosphorylation event activates ered toward the end of the night results in
the enzyme. Using antibodies specific for a phase advance. There is some evidence
phosphorylated MAPK, it was found that that the pathways for phase delays and
the level of active MAPK peaks at the phase advances are different. In phase
end of the subjective day. Note that this advances, activation of the NMDA receptor
peak is actually out of phase with the peak leads to an increase in nitric oxide. Nitric
of phosphorylated CREB, suggesting that oxide is a relatively recently described
RHY5 2/6/04 3:55 PM Page 101

LIGHT: THE PRIMARY INPUT FACTOR CONTROLLING MAMMALIAN CLOCKS 101

Phase delays Phase advances


Light Light

RHT RHT
(glutamate) (glutamate)

NMDA R NMDA R* NMDA R NMDA R*

NO
Ca2+

GTP cGMP
MAPK MAPK*

MAPK MAPK*

CREB CREB*
CREB CREB*

Transcription ON Transcription ON
mPer1, mPer2 mPer1

Figure 5.5. Molecular pathway for either phase advances or phase delays in response to a
light pulse during the subjective night. During a phase delay, the RHT releases glutamate,
which is bound by the NMDA receptor. The activated receptor (indicated by the asterisk) leads
to an increase in calcium. Through a series of proteins not shown here, the calcium leads to
phosphorylation and activation of MAP kinase (asterisk). Active MAPK leads to activation of
the transcription factor CREB by phosphorylation (asterisk). Active CREB then induces tran-
scription of the circadian clock genes, Period-1 and Period-2. A light pulse given at the latter
part of the subjective night also results in activation of the NMDA receptor. In contrast,
though, at this time of night, the active NMDA receptor leads to an increase in nitric oxide
(NO). NO activates an enzyme, guanylyl cyclase, which converts GTP into cyclic GMP. The rise
in cGMP levels also results in activated MAPK and eventually activated CREB. However, tran-
scription of only Period-1 is induced. The reason underlying the dichotomy of responses is not
clear, but presumably there are additional factors necessary for transcriptional activation that
are available in the early night, but not in the late night.

second messenger that activates guanylyl Involvement of Neuropeptide PACAP in


cyclase, an enzyme responsible for the pro- Photic Input
duction of cyclic GMP (cGMP). High levels
of cGMP activate cGMP-dependent kinase, Although glutamate is the primary neuro-
which in turn activates MAPK. As pointed transmitter of the RHT, the neuropeptide
out above, activation of MAPK leads to pituitary adenylate cyclase activating
activation of CREB. Note that both the polypeptide (PACAP) is also important for
phase advance pathway and the phase circadian clock function, especially with
delay pathway, while different, utilize acti- regard to input pathways. PACAP is formed
vated MAPK (Fig. 5.5). How this similar in the same retinal ganglion cells that
outcome leads to two different effects produce melanopsin, as discussed earlier.
depending on the time of night is still an The nerve terminals of these cells synapse
area of extensive study. with SCN neurons and express PACAP in
RHY5 2/6/04 3:55 PM Page 102

102 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

a circadian manner. Levels are highest at Functions of VIP and VPAC Receptors
night and lowest during the day. There is
VIP (see above), which is related to
both in vitro and in vivo evidence that
PACAP, is also implicated in photic input.
PACAP functions in the clock. PACAP
Unlike PACAP, which is produced in retinal
induces phase shifts in the SCN in much the
ganglion cells, VIP is produced by neurons
same way as in response to light. This
in the SCN that receive input from the
includes induction of the Per1 and Per2
retina. These neurons synapse with other
genes. PACAP may actually act as a modu-
regions of the hypothalamus. VIP is impli-
latory molecule because it modulates
cated in the phase shifting response of the
glutamate effects on the clock. PACAP
SCN to light both in vitro and in vivo. As
appears to dampen the glutamate derived
mentioned above, VIP shares receptors
phase advance seen late at night and
with PACAP, namely, VPAC1 and VPAC2.
enhance the glutamate-driven phase delay
Of these, only VPAC2 is expressed in the
seen in the early night. Still, precisely how
SCN. Indeed, in the nervous system, the
PACAP figures into the input pathway is
SCN is the area of highest VPAC2 expres-
not yet clear.
sion. Moreover, expression in the SCN is
There are three receptors for PACAP.
circadian, as it is highest during the day and
All are G-protein-coupled receptors. Two
lowest at night. The importance of VPAC2
of these, VPAC1 and VPAC2, bind both
in photic input was established by the cre-
PACAP and another neuropeptide, vaso-
ation of transgenic mice that overexpress
active intestinal peptide (VIP), equally
the human homolog of VPAC2. These
well. The third receptor is PAC1, which
transgenic mice reset their activity rhythms
essentially is specific for PACAP. Mice
more quickly in response to an advance in
lacking PAC1 were created using gene
the light–dark cycle. The creation of a
targeting technology. Experimentation with
knockout mouse lacking VPAC2 validated
these knockout mice revealed that PAC1 is
the function of this receptor in photic input.
not absolutely essential for entrainment
Mice lacking VPAC2 fail to induce
of circadian rhythms by light. When con-
mPer1 and mPer2 in response to light;
fronted with a light pulse in the early
therefore, it is clearly necessary for the
night, PAC1-deficient mice responded with
response to photic signals. VPAC2 is
phase delays greater than that observed
involved in more than just photic input,
with wild-type mice. The induction of
though. Evidence indicates that it is neces-
Per1 and Per2 by light was not affected in
sary for maintaining core circadian activity
PAC1-deficient mice, which may account
(see text below).
for the fact that these mice still respond to
light. The only notable deficiency displayed
by the knockout mice was an inability to
Other Retina–SCN Pathways
generate phase advances in response to a
light pulse given late at night. In contrast, Light is not the only input onto the central
such a light pulse caused PAC1-deficient oscillator in the mammalian SCN. As men-
mice to respond with phase delays. The rel- tioned above, other pathways indirectly
atively mild phenotype of PAC1-deficient connect the retina to the SCN. Because
mice suggested that one or both of the these pathways have relay stations, they
other PACAP receptors, VPAC1 or VPAC2, do not solely transmit information from
is the key receptor for light entrainment the retina. These pathways integrate in-
or that redundancy among these receptors formation from other parts of the brain
exists. and thus also transmit nonphotic input to
RHY5 2/6/04 3:55 PM Page 103

THE INTRACELLULAR MAMMALIAN CENTRAL OSCILLATOR 103

the SCN. One pathway from the retina to ablation of the SCN in rats resulted in a
the SCN first synapses in the IGL. The loss of circadian rhythms as measured by
IGL innervates the SCN via the geniculo- adrenal corticosterone secretion, drinking
hypothalamic tract (GHT); the main neu- behavior, and locomotor activity. More-
rotransmitter is neuropeptide Y. The IGL over, transplanting SCN cells from one
also functions in mediating nonphotic input animal into an animal with an ablated SCN
to the SCN. Induced physical activity shifts reversed the loss of circadian rhythms. In
the phase of an animal’s circadian rhythm, fact, the period of the rhythms of the trans-
and the IGL is necessary for this activity- planted animal always followed those of the
induced shift. Another indirect pathway donor.
from the retina to the SCN courses through
the raphe nuclei. Serotonin is the primary
Intracellular Rhythms of the Core
neurotransmitter of this pathway. The sero-
Circadian Oscillator
tonergic input may have a modulatory
effect on input from the RHT and may also A circadian rhythm of neuronal firing has
function in nonphotic phase shifting. been demonstrated in the SCN using brain
Finally, hormones have input onto the SCN; slices. However, many elegant studies
the most widely studied is melatonin. Mela- have established that the basis for rhythm
tonin is produced cyclically by the pineal generation is a cumulation of intracellular
gland under the control of the SCN, and is rhythms from individual SCN neurons.
regarded primarily as an output molecule Reppert’s group showed that dissociated
(see next section). However, melatonin can rat SCN neurons plated on microelectrode
feed back and influence the SCN, so it is arrays display sustained rhythms in their
mentioned here. Still, very little is known electrical activity. These findings were
about the mechanisms underlying any non- extended using a mutant hamster line
photic input; therefore, they are not dis- called tau, which has a shortened circadian
cussed further. period of wheel-running and a mutant
mouse line, Clock, which has a long period
in the heterozygous state and becomes
䊏 THE INTRACELLULAR arrhythmic when homozygous. Dissociated
MAMMALIAN CENTRAL SCN neurons from these mutants display
electrical activity that correlates well with
OSCILLATOR
the altered behavioral rhythms. The tau
mutant cells had shortened periods of elec-
Housing of the Mammalian Central
trical activity. Heterozygous Clock mutant
Oscillator in the SCN
SCN cells had long periods, and homozy-
As mentioned above, the SCN is home to gous cells were arrhythmic. Results with
the mammalian central clock. This structure these mutant animals indicated that the cir-
is located just above the optic chiasm in the cadian clock is cell-autonomous.
anterior hypothalamus. Several lines of evi- Notably, the circadian rhythms of in-
dence established the SCN as the central dividual cells are more variable than the
pacemaker of mammals. Neuroanatomical behavioral rhythms. It is hypothesized that
studies mapped the abovementioned con- the periodicity of the overt rhythm is the
nections from the retina to the SCN, and average of the periods displayed by indi-
other research demonstrated strong circa- vidual neurons. This is supported by elegant
dian rhythms of glucose utilization and neu- studies utilizing chimeric mice in which
ronal firing rate in the SCN. Additionally, the SCN are composed of a mixture of cells
RHY5 2/6/04 3:55 PM Page 104

104 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

from two different mouse strains of dif- NCAM is postulated to allow changes in
ferent period lengths. The period of the tissue architecture. The importance of PSA-
rest–activity rhythm depends on the rela- NCAM in the SCN was demonstrated
tive ratio of the two cell types. How the using mice deficient for this molecule.
individual cells communicate with each Under freerunning conditions, these mice
other to generate a unified rhythm is un- show a significantly shorter period of activ-
known at this time, and is one of many ity rhythm that degenerates into arrhyth-
fascinating, unanswered questions in the micity. Additionally, enzymatic removal of
field. PSA from the SCN of adult animals simi-
larly shortens the freerunning period.
GABA and PSA-NCAM are just two
Multifaceted Synchronization of
examples of molecules necessary for syn-
SCN Neurons
chronization of clock neurons. These indi-
How the individual neurons of the SCN cate that both secreted factors and direct
are synchronized is unclear. The primary intercellular contact are involved in estab-
neurotransmitter of these neurons is lishment of the unified core clock. A
g-aminobutyric acid (GABA). GABA nor- number of other factors, both secreted and
mally acts to inhibit neuronal activity. nonsecreted, are suggested as being impor-
Within the SCN, the exact function of tant for this clock. The evidence for the
GABA is unclear. One report suggested majority of these is incomplete; therefore,
that GABA excites SCN neuron firing they are not discussed further.
during the day, and inhibits such firing at
night. Unfortunately, this finding has been
A SCN Cell Line
difficult to replicate, and it is currently
believed that GABA only acts in an in- Utilizing rat fetal precursors of the SCN, a
hibitory manner in the SCN. Still, work with SCN cell line was established in 1999 by
dissociated SCN neurons in culture sug- immortalization with the adenovirus E1A
gests that GABA is important for intercel- gene. This cell line demonstrates many
lular communication. GABA synchronizes properties found in the intact SCN,
the phase of firing rate rhythms in SCN including circadian rhythmicity of glucose
cells in culture. Also, GABA treatment can utilization and neurotrophin expression.
phase-shift the electrical activity of these Moreover, transplantation of this cell line
cells. The direction of the phase shifting, into the brains of rats with SCN lesions led
either advancing or delaying, is dependent to the reestablishment of circadian
on the circadian time. locomotor activity. In contrast, a fibroblast
Because the SCN is composed of so cell line, which can be induced to display
many closely apposed cells, it is possible circadian expression of clock genes
that direct cell–cell contacts may contribute (see section below), is unable to reestablish
to intercellular communication. The SCN such rhythms. Also, the SCN cell line is
expresses a specialized neural cell adhesion able to drive circadian rhythms of glucose
molecule (NCAM) that contains a high uptake and circadian gene expression
level of polysialic acid (PSA-NCAM). in a cocultured fibroblast cell line. There-
NCAMs are normally important for cellu- fore, much like the intact SCN, this cell line
lar contact and intracellular signaling in can regulate rhythms of other cells via
neurons. The presence of polysialic acid secreted factors. Hopefully, this SCN cell
moieties on NCAM is found primarily in line will be beneficial in understanding the
the developing nervous system and in intra/intercellular nature of the circadian
plastic areas of the brain. Therefore, PSA- clock.
RHY5 2/6/04 3:55 PM Page 105

THE CENTRAL OSCILLATOR IS COMPOSED OF A FEEDBACK LOOP 105

䊏 THE CENTRAL OSCILLATOR IS cyclic transcription/translation feedback


COMPOSED OF A FEEDBACK LOOP loop that is based on the one originally
described in drosophila (Fig. 5.6). Two
It is clear that phenomenological research genes, period (per) and cryptochrome (cry),
in chronobiology has been quite successful are cyclically transcribed over the course of
in leading to an understanding of both the 24 hours; the peak transcript levels occur in
properties of, and the anatomical basis for, the middle of the day and the trough occurs
mammalian circadian rhythms. However, in the middle of the night. Translation is
since the early 1990s molecular biology and coupled to transcription; therefore, as the
genetics have provided insight into the level of transcript rises during the day, more
molecular mechanisms underlying the and more protein is produced. As the levels
central clock in mammals. The molecular of these proteins rise in the cell, they
basis of the mammalian circadian clock is a heterodimerize and translocate to the

Protein Degradation CYTOPLASM


P P

CRY P
P
PER
NUCLEUS

Kinases CLOCK::BMAL1

PER CRY
period
PER
PER
PER2
PER1
PER3
cryptochrome
CRY1 CRY2

Figure 5.6. Schematic of the feedback loop. The feedback loop consists of four main com-
ponents: PERIOD (PER), CRYPTOCHROME (CRY), CLOCK, and BMAL1. CLOCK and BMAL1 are
positively acting transcription factors. They heterodimerize and activate transcription of the
Per and Cry genes. There are actually three isoforms of PER and two isoforms of CRY as indi-
cated in the figure. The PERs form both homodimers and heterodimers and also hetero-
dimerize with the CRYs. On dimerization, PER and CRY translocate to the nucleus and inhibit
the activity of the CLOCK:BMAL1 heterodimer. Evidence suggests that CRY is the dominant
repressor. Suppression of CLOCK:BMAL1 activity causes transcription of Per and Cry to be shut
off. The feedback loop is not permanently disrupted, however, because the PER and CRY pro-
teins are targeted for degradation. The targeting signal is probably phosphorylation. The PERs
are phosphorylated by casein kinase Ie. The CRYs are also phosphorylated, although currently
the kinase responsible for this is unknown. On degradation of PER and CRY, transcriptional
repression is relieved and the cycle begins anew. The end result of the feedback loop is daily
oscillation of both mRNA and protein levels of the core components. The exception is CLOCK,
which is expressed at the same level throughout the day.
RHY5 2/6/04 3:55 PM Page 106

106 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

nucleus. In the nucleus, the PER:CRY het- been determined if GSK has a similar role
erodimer blocks the activity of two transcri- in mammals.
ption factors, CLOCK and BMAL1. In the
absence of PER and CRY, CLOCK and
BMAL1 form a heterodimer that activates
䊏 THE Clock GENE
transcription of the per and the cry genes.
In other words, when levels of PER and
Clock was the first cloned mammalian cir-
CRY are high, transcription of per and
cadian gene.
cry is repressed. Transcription is not per-
manently halted; however, because the
PER and CRY proteins are eventually tar-
A Forward Genetic Screen
geted for degradation, which decreases the
protein levels in the cell. When the levels The murine Clock (for circadian locomotor
fall enough, the transcriptional repression output cycles kaput) gene was the first mam-
is relieved and the cycle begins anew. malian circadian gene to be cloned. Its iso-
Notably, in mammals, there are actually lation is a beautiful example of the power of
three homologs of per: Per1, Per2, and Per3; forward genetics in a mammalian system.
and two homologs of cry: Cry1 and Cry2. The strategy to identify this gene was based
The reasons for having multiple homologs on that of Konopka and Benzer, who iden-
are not completely clear at this time, and tified the first circadian gene, period, in
are discussed further below. An important Drosophila. The basic idea is to induce
point is that the central clock machinery is genetic mutations in the germ cells of an
apparently at work in many tissues, not just organism by exposing it to a chemical
the brain; that is, cyclic expression of Per, mutagen. The exposed organisms are then
Cry, and Bmal1 is found in organs such bred to nonexposed organisms.If a mutation
as liver and kidney. Clock expression is occurs in a gene important for circadian
constant in both the SCN and peripheral rhythms, the offspring that inherit that
organs. The function of such peripheral mutation will show alterations in circadian
clocks is discussed in more detail later in behaviors. Because the offspring will only be
this chapter. heterozygous for any induced genetic muta-
As noted above, both PER and CRY tions, this type of screen identifies dominant
proteins are targeted for degradation, mutations. Takahashi and his colleagues
which allows the molecular circadian cycle undertook such an ambitious screen, and
to be continually reset. The signal to the published their results in 1994. In their
cell’s degradation machinery is hypothe- original screen, C57BL/6J male mice
sized to involve protein phosphoryla- were exposed to the mutagen, N-ethyl-N-
tion. Research has shown that casein kinase nitrosurea (ENU). These mice were bred to
Ie (CKIe) is responsible for phosphoryla- nonmutated female mice, and the progeny
tion of PER. CKIe and its role in the cir- were screened for alterations in circadian
cadian clock are discussed in further detail behavior, specifically wheel running. Labo-
below. Regarding the degradation of CRY, ratory mice normally are most active at
however, the story is not as clear as with night and show sustained wheel-running
PER. Interestingly, in Drosophila, TIME- activity with an average period of slightly
LESS (TIM), not CRY, partners with PER less than 24 hours. Approximately 300
to affect transcriptional repression, and progeny animals were screened, and the
glycogen synthase kinase (GSK) phospho- mean period of wheel-running activity was
rylates TIM (Chapter 3). As yet, it has not 23.7 hours. However, the period of one
RHY5 2/6/04 3:55 PM Page 107

THE Clock GENE 107

animal slowly lengthened over a month and positional cloning proved that such a tech-
stabilized at 24.8 hours. Breeding of this nique could be used to find behavioral genes
animal showed that the long period pheno- in mammals.
type was inherited as an autosomal, semi-
dominant mutation. Homozygous Clock
animals have a mean period of 27.3 hours
A PAS-Containing Protein Revealed in
when first moved to constant darkness, but
Clock Cloning
become arrhythmic over time (Fig. 5.7).
These data indicated that Clock is a core The discovery that Clock represented a
component of the mammalian circadian single gene allowed its location to be
system. The successful isolation of Clock by mapped by linkage analysis. Genetic crosses

ENU X
Clock Wild type

X
Male Female
(exposed) (unexposed)
X
WT/WT Clock/WT

G1 offspring (304)
(304)

(25%) (50%) (25%)


Wild type (303) Clock founder (1) WT/WT Clock /WT Clock / Clock

Figure 5.7. Mutation strategy used to generate Clock mutant mice. Male mice were exposed
to the mutagen, ENU. These mice were then bred to unexposed female mice, and the off-
spring (G1) were screened for disorders of circadian locomotor activity rhythms. The vast
majority of mice had a normal period length of 23.7 hours, but one mouse had a period of
approximately 25 hours. Schematics of the locomotor activity records are shown. The arrow
indicates the point where the mice are moved from a light–dark cycle to constant darkness.
The long-period mouse was bred to wild-type females and the offspring were screened for
locomotor periods. Approximately half of these offspring had wild-type periods, and half had
the long-period phenotype, indicating that the mutation was a single-gene autosomal dom-
inant. The long-period phenotype animals were interbred to generate homozygous animals.
The offspring fell into three groups phenotypically with respect to locomotor activity period:
wild type (25%), parental (50%), and an even longer period of approximately 28 hours (25%).
These represented homozygous wild type, heterozygotes, and homozygous Clock mutation,
respectively. Such data indicates that the Clock gene is a single, semidominant gene. Notably,
the homozygous Clock mutant animals degenerate into arrhythmicity after ~2 weeks in con-
stant darkness.
RHY5 2/6/04 3:55 PM Page 108

108 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

mapped the Clock locus in relation to sponded to the bHLH-PAS protein that
simple sequence length polymorphisms was identified by positional cloning. There-
(SSLPs) and assigned the locus to chromo- fore, it was clear that this gene was able to
some 5. The actual cloning of Clock was rescue the Clock mutant phenotype, and
a bit more difficult and was reported in a proved that this candidate gene indeed was
pair of landmark papers in 1997. Two com- the Clock gene.
plementary approaches were used: standard Sequencing of the entire candidate Clock
positional cloning and transgenic rescue. gene showed it to be quite large. It spans 100
With regard to positional cloning, first, high- kb of genomic DNA and contains 24 exons.
resolution genetic mapping using SSLPs The open reading frame encodes a protein
narrowed the region of interest on chromo- of 855 amino acids with a glutamine-rich
some 5 to approximately 400 kb (thousand carboxyl terminus and a bHLH domain
base pairs). This area was further subdi- common to many transcription factors.
vided by constructing yeast artificial chro- Sequencing of this gene in Clock mutant
mosomes (YACs) and bacterial artificial mice showed an A-to-T transversion at the
chromosomes (BACs). Shotgun sequencing splice donor site of intron 19. This mutation
of these constructs was used to identify can- results in a splicing change that removes
didate genes. Strikingly, one of these candi- exon 19 and therefore leads to a loss of 51
dates had sequence similarity to the PAS amino acids in the glutamine-rich region.
domain. This domain is found in many pro- These groundbreaking experiments proved
teins, including certain basic helix– that this PAS-containing protein was the
loop–helix (bHLH) transcription factors. product of the Clock gene, and fully estab-
More importantly, the PAS domain had lished the use of positional cloning to isolate
already been found in the Drosophila circa- mammalian clock genes. Subsequent work
dian protein PERIOD. The link to a known placed CLOCK into the transcriptional
circadian protein was quite compelling, and translational feedback loop that makes up
certainly suggested that this gene repre- the central oscillator.
sented the Clock gene. Additionally, north-
ern blot analysis of hypothalamus RNA
showed that Clock/Clock homozygous mice 䊏 BMAL1
expressed a lower level of this candidate
gene transcript than did wild-type mice, BMAL1 binds to Clock and regulates cir-
further strengthening the hypothesis that cadian transcription.
this candidate was the actual Clock gene.
As a complementary approach to posi-
A Dominant Negative Clock
tional cloning, BAC clones covering the
400-kb area were isolated and used to make Given the fact that the CLOCK protein had
transgenic mice. Lines of mice that were domains common to many transcription
homozygous for the Clock mutation, but factors, such as the bHLH domain and the
also contained individual BAC transgenes, glutamine-rich region, it was hypothesized
were generated. One of these mouse lines that CLOCK functioned in the circadian
had a wild-type phenotype with respect to clock by controlling transcription. Indeed,
locomotor activity rhythms. In other words, cyclic expression of mPer1, mPer2, and
this BAC transgene was able to rescue mPer3 is blunted in Clock mutant mice,
the Clock phenotype. Therefore, the actual thus providing further evidence that
Clock gene had to be contained within this CLOCK controls transcription of circadian
BAC clone. This BAC clone contained only genes. Furthermore, since CLOCK contains
three candidate genes. One of these corre- a PAS domain for protein dimerization, the
RHY5 2/6/04 3:55 PM Page 109

BMAL1 109

obvious experiment was to search for the hypothesis was confirmed by the demonstra-
CLOCK partner. The first partner isolated tion that a CLOCK:BMAL1 heterodimer
was BMAL1 (brain–muscle ARNT-like bound to an E box found in the promoter
protein 1), also known as MOP3 (member of the Drosophila period gene. Moreover, the
of PAS superfamily 3). Strikingly, this CLOCK:BMAL1 heterodimer activates
protein is also a bHLH transcription factor transcription of a reporter gene fused to the
that contains a PAS domain. promoter sequence of the mPer1 gene, which
Members of the family of bHLH tran- also contains E boxes. The establishment of
scription factors bind as dimers to the DNA transcriptional activity for CLOCK nicely
sequence,CACGTG,also known as an E box. explained the observed phenotype of Clock
Interestingly, at the time of CLOCK and mutant mice. Recall that the Clock mutation
BMAL1 isolation, E boxes were beginning results in a deletion of 51 amino acids from
to be found in the promoter regions the glutamine-rich carboxyl terminus. Such
of many circadian genes.Moreover,extensive regions often act as transcription activation
analysis of the Drosophila period gene,which domains. Therefore, it appears that the
was the best characterized clock gene, mutant CLOCK protein is able to het-
revealed that these E boxes were necessary erodimerize with BMAL1 and bind to the E
for proper transcription. The hypothesis was box, but this heterodimer cannot activate
that PAS-bHLH proteins bound to the E transcription. In other words, the protein
boxes of circadian gene promoters and regu- product of the Clock mutation acts as a dom-
lated transcription of these genes. This inant negative (Fig. 5.8).

bHLH PAS Glutamine-rich


Wild type

Clock

CLOCK
Clock

Q-Rich Q-Rich

BMAL1 BMAL1

PAS PAS PAS PAS

bHLH bHLH bHLH bHLH

Wild-type Clock
transcription ON transcription OFF

Figure 5.8. The Clock mutation results in a dominant negative transcription factor. The Clock
mutation leads to alternative splicing, resulting in a protein that lacks exon 19. This exon is
part of the glutamine-rich region that is important for transcriptional activation. CLOCK
normally partners with BMAL1 by direct interaction of the PAS domains. The CLOCK:BMAL1
heterodimer is transcriptionally active. The CLOCK mutant protein can also heterodimerize
with BMAL1. However, this complex is not transcriptionally active because of the truncated
glutamine-rich region.
RHY5 2/6/04 3:55 PM Page 110

110 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

Function of BMAL1 in Circadian genes is also abolished. As we will discuss


Rhythms in Mice later in this chapter, peripheral organs also
show rhythmic expression of many genes,
The finding that BMAL1 binds to CLOCK the function of which is not at all clear. In
and activates transcription of the the peripheral organs of BMAL1 knockout
drosophila period gene strongly suggested animals there is low-level, nonrhythmic
that this protein was a core component expression of many of these genes.
of the mammalian circadian clock. Fur- The BMAL1 knockout animals had
thermore, Bmal1 expression is cyclically additional, although more subtle, pheno-
expressed in the SCN, as would be expected types. For example, while the animals show
of a core circadian component. Addition- rhythmic behavior in light–dark conditions,
ally, the highest expression of Bmal1 occurs the phase of activity onset is variable. Also,
during the latter portion of the night. This a good portion of the activity occurs during
is the opposite phase to mPer1 and mPer2, the lights-on time. These phase and activity
consistent with the idea that BMAL1, along disturbances indicate that BMAL1 knock-
with its partner, CLOCK, activates the out mice have difficulty in responding to
transcription of these two genes. Additional light, which suggests that BMAL1 does
data indicate that BMAL1 itself functions play a role in entrainment, as had been sug-
in a loop where its protein product regu- gested earlier. Moreover, these mice do not
lates the levels of its mRNA. PER2 posi- respond to a light pulse in the middle of
tively regulates Bmal1, resulting in the the night, either, further supporting that
interlocking of the BMAL1 loop with the hypothesis.
PER/CRY loop. Finally, BMAL1 also plays Examination of the BMAL1 knockout
a role in the light input control over cir- animals reveals an additional phenotype.
cadian rhythms. For example, the actual These animals show an overall lower
protein level of BMAL1 is reduced in amount of activity than do their wild-type
response to a light pulse delivered in the littermates. Such a result suggests that loss
middle of the night. In contrast, the Bmal1 of BMAL1 disrupts not only the core clock
transcript in the SCN shows no change in but also the clock’s communication with the
level on exposure to such a light pulse. Such organism’s physiology as a whole. In other
a result indicates that regulation of BMAL1 words, BMAL1 may have a role in circa-
by light is posttranslational. dian output pathways, such as behavioral or
To fully characterize the role of BMAL1 metabolic outputs. Clearly, BMAL1 is one
in mammalian circadian rhythms, a genetic of the most important circadian proteins,
knockout in mice was created. In light–dark but its function is still far from being com-
conditions, these mice run on wheels with pletely understood.
regular periodicity. However, when moved
to constant darkness, these knockout mice
display an immediate and complete loss of 䊏 THREE MAMMALIAN
circadian rhythms (Fig. 5.9). Such a result HOMOLOGS OF PERIOD
proves that BMAL1 is a central clock com-
ponent. Moreover, the knockout animals So far, our discussions have focused on
have lost rhythmic expression of mPer1 and CLOCK and BMAL1, which together
mPer2 in the SCN. In fact, overall expres- activate transcription of circadian genes.
sion levels of these genes are very low. However, recall that, in accordance with
These findings proved that BMAL1 is nec- models derived from insects, there are
essary for mPer1 and mPer2 transcription. negative regulators of the circadian
Interestingly, rhythmic expression of other clock; the primary ones are PERIOD and
RHY5 2/6/04 3:55 PM Page 111

THREE MAMMALIAN HOMOLOGS OF PERIOD 111

7 pm 7 am 7 pm 7 am

Days

Figure 5.9. Schematic activity record of Mop3 knockout mice. The knockout mice entrain to
a light–dark cycle without difficulty and show the standard 24-hour period. However, on
placement in constant darkness, the mice immediately become arrhythmic. Note that this
result contrasts to the Clock mutant animals, which display long periods in constant darkness
for approximately 14 days before becoming arrhythmic. The immediate arrhythmicity
observed in the Mop3 knockout animals indicates that this gene is indispensable for main-
taining circadian rhythms in mice.

TIMELESS. Now we turn our attention to no particular reason, except for the order
the negative regulators found in the mam- of isolation. The presence of multiple
malian circadian clock. homologs does indicate, not surprisingly,
that mammalian chronobiology is a bit
more complex than that found in insects.
Three PER Homologs
Two groups originally reported the
The Drosophila period gene was originally isolation of the first mammalian period
identified in a mutation screen by Konopka gene, Per1. One group fortuitously found
and Benzer in 1971 and was eventually Per1 when analyzing cDNAs derived
cloned in 1984. However, despite numerous from human chromosome 17. One of these
attempts to clone a mammalian homolog, cDNAs had a PAS domain. Complete
none was found. This lack of success led cloning of this gene, which was originally
many to believe that the entire basis for the called RIGUI, demonstrated that it was
mammalian circadian clock was unrelated highly homologous to the Drosophila
to that found in insects. After a decade period gene. The murine homolog, called
of waiting, though, a mammalian period m-rigui, was also isolated. The expression of
homolog was finally isolated. In fact, the this gene was shown to oscillate in both the
field was rewarded with three homologs. retina and the SCN of mice. These oscil-
These are named Per1, Per2, and Per3 for lations not only continued in constant
RHY5 2/6/04 3:55 PM Page 112

112 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

darkness, but they were also phase-shifted involves the response to light. The expres-
by a pulse of light. These lines of evidence sion of both mPer1 and mPer2 is induced on
strongly suggested that RIGUI represented exposure to light. However, mPer1 induc-
the functional homolog of Drosophila tion is much stronger and more rapid than
period. that for mPer2. These differences may have
The other group that first reported the functional consequences. Mice lacking the
sequence of Per1 utilized a PCR-based mPer1 gene do not phase-advance their
screen to amplify products from human locomotor rhythms in response to a light
genomic DNA that had regions homolo- pulse given early in the night. In contrast,
gous to the PAS domain of Drosophila mice lacking the mPer2 gene do not phase-
period. One of the amplified genomic prod- delay their locomotor rhythms in response
ucts was used to isolate a corresponding to a light pulse given in the latter part of
cDNA. This cDNA had high homology the night. In contrast to mPer1 and mPer2,
to Drosophila period, and was called expression of mPer3 is immune to light
hPER. Using hPER as a probe, the mouse induction, suggesting that it has no role in
homolog mPer (identical to m-rigui) the input to the circadian clock.
was also cloned. Like the first group, this Another key difference between the
second group also showed that mPer was proteins is the expression pattern in vari-
expressed in the SCN, and that the levels of ous tissues. Strikingly, there is differential
the transcript varied throughout the day. It expression of the Per genes within the SCN
was quite clear that the search for a mam- itself. Silver and colleagues have shown
malian counterpart for drosophila period that, in hamsters, one region of the SCN
was over. expresses Per1, Per2, and Per3 in a rhythmic
manner. Another region displays constant
Per3 expression and light-induced expres-
Possible Functional Differences
sion of Per1 and Per2. These results suggest
The other two PER homologs were found in functional differences between the PER
a much simpler manner than that used for homologs. Additionally, there are differen-
PER1. Reppert’s group knew that more and tial expression patterns among nonneuro-
more sequence data were being deposited nal tissues as well. As mentioned earlier,
into a public database daily. By continuing peripheral tissues rhythmically express the
to run computer searches of the database for core circadian genes. The mPer genes are
homologs to Drosophila PER and hPER1, not exceptions. However, each of these
two additional genes were identified. These genes is expressed in specific subsets of
were called Per2 and Per3, and their identi- peripheral tissues and at different levels in
fication suggested that in mammals, fami- these tissues. The reasons for this differen-
lies—rather than individual members— tial expression are unknown, but suggest
represented the circadian proteins. that the Per homologs may have tissue-
Reasons for having multiple homologs specific functions. Clearly, deciphering
were not at all clear at the time of discov- the individual functions of the different
ery, and while by no means complete, some homologs will give great insight to the func-
key differences are emerging. The tran- tion of the clock in physiology.
script levels of all three homologs cycle in
the SCN, albeit with staggered phases. Still,
Knockout Mice
levels of all three are highest during the
day and fall to a nadir during the night. As we indicated, one advantage that
One primary difference among the genes mammals have over insects is the ability to
RHY5 2/6/04 3:55 PM Page 113

THREE MAMMALIAN HOMOLOGS OF PERIOD 113

manipulate the mammalian genome to gen- PER1


erate animals that lack certain genes. To
date, the PER genes have all been targeted The most recent of the Period genes to be
for deletion, and such knockout animals genetically deleted is mPer1. Deletion of
prove that the PER proteins are core com- mPer1 was carried out by three different
ponents of the central clock. Moreover, evi- groups, and surprisingly, two interesting but
dence from knockout mice now suggests slightly different phenotypes are found. All
that mPER1 acts posttranscriptionally, and groups found that, much like the mPer2
that mPER2 is a transcription regulator. knockout animals, the circadian rhythm of
Also, mPER3 is hypothesized to be an mPer1 knockout mice was shorter (approx-
output molecule. imately one hour) than wild type when
measured by wheel running. Additionally,
all groups found that, unlike the mPer2
PER2
mutant animals, the mPer1 mutant mice
The first gene that was deleted was mPer2, maintained their rhythms in constant dark-
and it was actually the first circadian gene ness, at least for some time. Extremely inter-
deleted in such a manner. In this original esting, though, is the fact that the mPer1
knockout, the PAS domain of mPer2 was mutant animals are quite variable in the
targeted for deletion, creating an allele length of their periods. Specifically, it
called mPer2brdm1. These mice display a appears that mPER1 is important for main-
shorter period than do wild-type mice when taining the precision of the circadian period,
measured for wheel-running activity. This and not necessarily the rhythm itself.
shortened period gradually degrades into Reppert’s group found that their mPer1
arrhythmicity when the mice are housed knockout mice (mPer1Idc) completely lose
in constant darkness, much like the Clock rhythmicity after about 2 weeks in constant
mutant mice. These findings definitely darkness; however, the other two groups did
established PER2 as a component of not observe any loss of rhythm. Currently, it
the central clock. Interestingly, this mPer2 is unclear as to why there is a difference in
knockout also showed diminished RNA the phenotypes of the three lines of knock-
levels of the mPer1 and mPer2 genes in out mice; how-ever, the targeting strategy
the SCN, indicating that it may control for the mPer1Idc mouse included disrupting
transcription of these genes in vivo either the start codon. This mouse presumably
directly or indirectly. Surprisingly, such a makes no transcript. In contrast, the other
result does not fit with the hypothesis two groups disrupted primarily the PAS
that the PER proteins act as transcriptional domain of the mPer1 gene. Indeed, in both
repressors as discussed above. In fact, the of these mouse lines a small partial tran-
opposite hypothesis is suggested; that is, script is detected. It is possible that these
PER2 has a positive effect on transcription transcripts code for partial proteins that
of circadian genes. This result was repeated have some residual mPER1 function.
by another group that produced a mPer2 Despite their differences, all three
knockout animal. The phenotype and the groups found that there is no apparent
effect on transcription is the same. There- effect on circadian gene transcription in the
fore, while it is clear that mPER2 is a core SCN of mPer1 knockout animals; namely,
clock component, its role with respect to the mPer2, Bmal1 and mCry1 (see text
transcription is not clear. below) genes are still rhythmically ex-
pressed at high levels. However, despite the
lack of effect on transcription, the levels
RHY5 2/6/04 3:55 PM Page 114

114 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

of these proteins are strikingly reduced in “double knockout” animals, that is, ani-
the SCN. Thus, the primary function of mals that lack two genes (or more). These
mPER1 may be to mediate protein stabil- types of studies have been accomplished.
ity rather than transcriptional control. This Double-knockout animals of mPer1 and
phenotype is in striking contrast to that mPer2 show a much stronger phenotype
found with the mPer2 knockout mice, which than do either single-knockout types alone.
have strong effects on circadian RNA These mice display an immediate loss of
levels. Drawing comparisons between the wheel-running rhythmicity when placed in
mPer1 and mPer2 knockout animals nicely constant darkness. This type of profound
demonstrates the power of gene targeting phenotype is similar to the BMAL1 mutant
technology to allow a better understanding animals described above. In contrast,
of specific functions of two proteins. mPer1/mPer3 and mPer2/mPer3 double-
knockout animals do not show any pheno-
typic differences from the single knockout
PER3
of either mPer1 or mPer2. This result sug-
Finally, a mPer3 knockout mouse has also gests that mPer3 is not really a core com-
been produced. Surprisingly, mutation of ponent of the circadian clock, although with
this gene has very little effect on circadian the caveat that the mPer3 knockout animal
rhythms. The only observed effect is a mod- makes a partial transcript.
erate shortening (0.5 hour) of the circadian
wheel-running period when the mice are
Dimerization
held in constant darkness. It is possible that
mPER3 has a more specialized role in the Just like CLOCK and BMAL1, the PER
mammalian circadian clock, or even that proteins have PAS domains for protein
it is completely dispensable for clock dimerization. Recall that in Drosophila,
function. The short-period phenotype of PER utilizes its PAS domain to het-
PER3 knockout mice is similar to that of erodimerize with TIM, enter the nucleus
mice lacking the output gene, albumin d- and repress transcription by the insect
element binding protein (Dbp) (see discus- homologs of CLOCK and BMAL1. If
sion below). Therefore, some hypothesize mammals were as easy as insects, the PER
that PER3 acts as an output molecule pos- proteins would interact with mammalian
sibly by regulating transcription. Compli- TIM homologs and repress transcription by
cating interpretation of this study, though, CLOCK and BMAL1. However, the
is the fact that a partial transcript is made knockout of the only known mammalian
in these mice. Therefore, similar to two of TIM homolog is embryonically lethal,
the mPer1 knockout mouse lines, the mPer3 making it difficult to address its function
knockout mouse may make a partial in the clock. Also, although the hypothesis
protein. If such a protein is at least partially exists that PER proteins function in
active, it may hide the true null phenotype. transcriptional repression, current data
suggest that only mPER2 regulates
transcription.
Multiple Knockouts
Since all the PER homologs have PAS
Another complication of these knockout domains, it is logical to postulate that they
studies is the possibility that the PER pro- can interact with themselves and with each
teins may be functionally redundant. That other. Indeed, this is the case; multiple
is, if one is missing, another can take its groups have shown that the PER homologs
place, and substitute for its function. The can homodimerize and heterodimerize in
only way to address this issue is to generate all combinations in both tissue culture and
RHY5 2/6/04 3:55 PM Page 115

CRYPTOCHROMES IN THE MAMMALIAN CORE CLOCK 115

the SCN. Still, such studies leave us a long trainment of gene expression. In flies, there
way from determining the function of these is one CRY homolog that functions as the
interactions. Indeed, the PER proteins actual circadian photoreceptor molecule.
actually accomplish their functions by het- Continuing with the theme, though, the sit-
erodimerizing with the CRYs, which were uation is much less clear in mammals. There
mentioned above. Cryptochromes are dis- are two mammalian homologs of CRY:
cussed in greater detail in the next section, CRY1 and CRY2. In the mouse, the two
but for now, recall that there are two cryp- Cry genes are clearly important for circa-
tochrome homologs, CRY1 and CRY2. It dian rhythms. Gene expression of both
appears that heterodimerization with the mCry1 and mCry2 cycles in the SCN
CRYs enhances protein stability of the with message levels highest at the end of
PER proteins. Additionally, such hetero- the day. This is delayed from the peak of
dimerization results in nuclear entry of the Per genes’ expression. Protein expres-
the PER proteins, although there is also evi- sion of CRY1 and CRY2 also cycles in the
dence of CRY-independent nuclear entry. SCN, with the peak following approxi-
In short, the homo- and heterodimerization mately 3 hours after the mRNA peak.
of the various PER protein isoforms is The cyclic expression of the Cry genes is
important for their subcellular localization compelling; however, proof of the
and for their function. However, precisely CRYs’ function in the clock came from
defining the specific functions of specific knockout mice.
dimer combinations in the circadian clock
is a work in progress.
Mammalian CRYs Not Necessary for
Circadian Photoreception
Obviously, the discovery that there were
䊏 CRYPTOCHROMES IN THE
mammalian homologs of the plant and
MAMMALIAN CORE CLOCK
insect circadian photoreceptor set off an
intense search to prove that these homologs
Two Mammalian Homologs of
had similar functions in mammals. Data
Cryptochrome
from knockout animals indicate that this
As mentioned above in the section regard- may not be the case. The first of the two
ing light input to the circadian clock, a genes that were deleted was mCry2. In
novel class of molecules, called cryp- light-dark conditions, the mCry2 knockout
tochromes (CRYs), has been recently mouse displayed a normal 24-hour period
described in mammals. CRYs are homologs of wheel running. However, this period
of blue-light photoreceptors originally lengthened by about an hour when the mice
found in plants and are related to the family were kept in constant darkness. The mCry1
of proteins called UV-dependent pho- knockout animals also showed a 24-hour
tolyases, which function to repair DNA. period of wheel running in light–dark con-
Photolyases bind the cofactors methenylte- ditions. However, in contrast to the mCry2
trahydrofolate (MTHF), which “harvests” knockout animals, the mCry1 knockout
light, and flavin-adenine dinucleotide animals had wheel-running periods that
(FAD), which is necessary for the catalytic were shortened by about 2 hours in con-
function of photolyases (Fig. 5.14). CRYs stant darkness.Taken together, these results
also bind MTHF and FAD; however, they indicate that the two mCRY proteins have
do not have photolyase activity. In plants, opposite effects on the timing of the clock.
CRYs act as blue-light photoreceptors that Also, the fact that these animals could be
regulate flowering and circadian photoen- driven by light–dark conditions indicates
RHY5 2/6/04 3:55 PM Page 116

116 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

that neither of these is absolutely essential mouse and human cryptochromes. More-
for light-driven rhythmicity. over, data suggest that this repression
Of course, it was possible that in a single- occurs through direct interaction of the
knockout animal, the nondeleted homolog mCRYs with BMAL1. Also, both mCRY1
takes over for the disrupted gene. To and mCRY2 can physically interact with all
address this possibility, double-knockout three mPER protein isoforms, particularly
animals were generated. Strikingly, animals with mPER2. Taken together, in mammals
lacking both mCRY1 and mCRY2 have an CRYs have apparently replaced TIME-
immediate loss of rhythms in constant con- LESS as the PER partner. In other words,
ditions. This finding proves that the CRY CRYs and PERs heterodimerize and block
proteins are necessary to generate core cir- the activity of CLOCK and BMAL1. In this
cadian rhythms. In addition, the mCRYs model, the CRYs are the dominant repres-
cannot be the sole circadian photoreceptors sors, and the PERs are necessary for
because these double-knockout mice do nuclear localization.
entrain to light. Furthermore, induction of
mPer1 and mPer2 expression in the SCN in
response to a nocturnal light pulse still
occurs in double-knockout animals. As
䊏 THE Tau GENE
mentioned above, however, rd mice that
Tau is an example of a naturally occurring
lack both Cry genes show deficits in
circadian mutation.
entrainment not seen in either the rd mice
themselves or in the mCry1/mCry2 double
knockouts, leaving open the possibility that
Short Circadian Periods
the mCRYs contribute at least partially to
circadian photoreception. The isolation of Clock is a great example
of identifying circadian components by
forward genetics. The story of tau is an
Mammalian CRYs in the Core
example of how nature occasionally con-
Circadian Clock
ducts mutagenesis experiments for us. The
The fact that mCry1/mCry2 double-knock- tau mutation was originally isolated as a
out animals are arrhythmic indicates that spontaneously arising mutation in a male
they are part of the core circadian Syrian hamster. This male had a very short
clock mechanism. Interestingly, in period of 22 hours in constant darkness as
the mCRY1/mCRY2 double-knockout measured by wheel running. To further
animals, mPer1 and mPer2 are no longer characterize the circadian phenotype, this
cyclically transcribed; the level of expres- male was bred with normal female ham-
sion is constitutively high. This finding sug- sters, and the wheel-running periods of the
gested a transcriptional repressor role for offspring were determined (Fig. 5.10).
CRY1 and CRY2. Furthermore, transcrip- These offspring fell into two distinct groups
tion of mCry1 and mCry2 is attenuated without regard to the animals’ sex. One
in Clock mutant mice, suggesting that group had a normal period of 24 hours, and
CLOCK acts as an activator of mCry1 and the other had a period of 22 hours. Such a
mCry2 transcription. A role for CRY in the pattern was consistent with the tau muta-
circadian transcriptional feedback loop was tion representing a single, semidominant,
further strengthened by the finding that autosomal mutation. To produce homozy-
both mCRY1 and mCRY2 block transcrip- gous animals, the offspring with the shorter
tion of mPer1 by CLOCK : BMAL1 het- period were interbred. Again consistent
erodimers. This finding applies to both with tau representing a single autosomal
RHY5 2/6/04 3:55 PM Page 117

THE Tau GENE 117

Male Female Tau /WT Tau /WT


(mutant) (wild type)
X
X

F2 offspring

F1 offspring
Tau / Tau (25%) WT/WT (25%)
period = 20 hours period = 24 hours

Tau /WT (50%) WT/WT (50%) Tau /WT (50%)


period = 22 hours period = 24 hours period = 22 hours

Figure 5.10. Isolation of the tau mutant hamster line. One male with a locomotor period of
22 hours was isolated as a spontaneously generated mutant in a group of Syrian golden ham-
sters. Note that the normal period for hamsters is 24 hours, so that the period stays the same
even in constant darkness. Mating of this mutant hamster male, called tau, to wild-type
females yielded a 50 : 50 ratio of short and wild-type period phenotypes, irregardless of sex.
Such a result indicates that the tau mutation represents a single, dominant, autosomal gene.
Mating of two heterozygote animals generated three phenotypes with respect to locomotor
periods: 20, 22, and 24 hours. These were in a ratio of 1 : 2 : 1 indicative of classic Mendelian
inheritance of a single semidominant gene.

gene, the phenotype of the offspring fell Despite its importance, the tau gene was
into three groups with respect to period not cloned until 2000, because of the lack of
length: 24, 22, and 20 hours. Presumably, the genetic markers in hamsters. The develop-
group with the shortest period represented ment of an elegant cloning strategy solved
homozygous animals. The reporting of the that problem.
tau mutation in 1988 was extremely useful
to the study of mammalian circadian
Cloning Difficulties
rhythms. The use of hamsters homozygous
for the tau mutation further established the Cloning of the tau gene was difficult
SCN as the seat of circadian rhythms in because of the lack of a genetic map in
mammals, as SCN transplants between hamsters. In other words, classical methods
wild-type and tau mutant hamsters showed of positional cloning such as those used to
that periodicity of wheel-running activity isolate Clock, would not be useful because
was conferred exclusively by the SCN. not enough genetic markers were known in
RHY5 2/6/04 3:55 PM Page 118

118 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

the hamster genome. This difficulty was cir- wild-type and tau mutants by the RT-PCR
cumvented by using a technique called using RNA extracted from the SCN of
genetically directed representational differ- these animals. Comparison of the
ence analysis (GDRDA), which allows the sequences revealed that tau hamsters have
identification of molecular markers linked a C-to-T transition that causes an arginine
to a trait without a genetic map. First, the to cysteine substitution at residue 178. This
tau mutant hamster strain was crossed to a residue is highly conserved among casein
wild-type hamster strain of a significantly kinase isoforms from many species, sug-
different genetic background. The F1 gen- gesting that this transition represented the
eration was interbred to produce F2 actual tau mutation. Although there was no
animals. These crosses produced animals apparent difference in amount of CKIe
that were genetically heterogeneous. Phe- mRNA or protein in the SCN of tau and
notype analysis showed that, as expected, wild-type hamsters, the tau CKIe isoform
the F2 progeny fell into three distinct had diminished kinase activity. Taken
groups in a mendelian ratio with respect to together, these data strongly indicated that
the tau phenotype. DNA was extracted CKIe was the product of the tau locus and
from homozygous wild type and from that the point mutation was responsible for
homozygous tau F2 animals, and was com- the observed phenotype.
pared by using differential hybridization
and the PCR to detect polymorphisms. Two
Regulation of Cyclic Activity
polymorphisms that segregated with the tau
mutation were isolated. Sequencing of The tau mutant phenotype was explained
hamster-derived lambda phage clones con- on the basis of work done in drosophila. In
taining these polymorphisms revealed a Drosophila, it appears that DBT phospho-
microsatellite repeat that was also poly- rylates PER, and this phosphorylation leads
morphic. This microsatellite was conserved to PER degradation. Notably, wild-type
in mice, and mapped to chromosome CKIe phosphorylates both mPER1 and
15. Analysis of the mouse genetic map mPER2 in vitro. Tau mutant CKIe binds to
around this microsatellite revealed a 15- both mPER1 and mPER2, despite the pres-
centimorgan region of chromosomal ence of the C-to-T transition. This is similar
synteny between mouse and hamster. to what was shown for the Drosophila DBT
According to previously established maps, mutant in its interaction with PER.
this region was also syntenic with human However, as mentioned above, the mutant
chromosomes 8, 12, and 22. Surprisingly, the CKIe was much less efficient at phosphory-
human casein kinase Ie (CKIe) gene is lation. Finally, in tau mutant hamsters the
found on chromosome 22. This was an level of Per1 RNA in the SCN is reduced,
extremely exciting finding because muta- and the period of RNA cycling is short-
tions in the doubletime (dbt) gene, the ened. This finding is consistent with the
Drosophila homolog of CKIe, alter circa- shortened circadian period demonstrated
dian rhythms. Therefore, CKIe was a strong by tau mutant animals.
candidate for the tau gene. The means by which CKIe affects the
To confirm that CKIe was the product of clock is still actively being studied. Some
the tau locus, the hamster CKIe was cloned. groups have used tissue culture models to
The mouse homolog of CKIe had not been show that CKIe phosphorylates both
mapped yet; therefore, primers designed mPER1 and mPER2, and suggest that this
from the human CKIe sequence were used phosphorylation event leads to protein
to isolate the hamster CKIe gene in both degradation. However, there is also evi-
RHY5 2/6/04 3:55 PM Page 119

OTHER POSSIBLE COMPONENTS OF THE CLOCK 119

dence that the phosphorylation state of the there are additional levels of control: (1)
PERs affects their localization within a cell: BMAL1 inhibits its own transcription and
nuclear or cytoplasmic. Of course, targeted (2) PER2 actually activates Bmal1 tran-
degradation and subcellular localization scription. Work by Ueli Schibler’s labora-
are not mutually exclusive methods of reg- tory identified REV-ERBa as a link
ulating protein activity. between the positive and negative limbs of
CKIe also phosphorylates mCRY1, the feedback loop by establishing its role in
mCRY2, and BMAL1. It appears that cyclic Bmal1 expression and by demon-
CKIe, the PERs, and the CRYs all form a strating its regulation by PER proteins.
large complex and that phosphorylation REV-ERBa is a member of the ligand
by CKIe regulates the nucleocytoplasmic activated nuclear receptor superfamily of
localization of the dimer. Presumably, this transcription factors. Because its activating
will affect the repressor function of the ligand is unknown, REV-ERBa is called
PER : CRY complex. In contrast, the effect an “orphan” nuclear receptor. REV-ERBa
of phosphorylating BMAL1 is believed to acts as a transcriptional repressor and is
increase BMAL1-mediated transcription. implicated in the regulation of adipogene-
Indeed, the CLOCK:BMAL1 heterodimer sis and of metabolism. However, Schibler’s
is apparently always bound to DNA, but group found that REV-ERBa is a tran-
the phosphorylation state of BMAL1 is cir- scriptional repressor of Bmal1. Addition-
cadianly regulated. Therefore, cyclic phos- ally, Rev-erba expression is cyclic in the
phorylation of the transcription factors may SCN, and Rev-erba knockout mice
regulate their cyclic activity. lack cycling of Bmal1 in the SCN. Surpris-
ingly, though, the circadian phenotype
of these animals is subtle. Knockout mice
䊏 OTHER POSSIBLE COMPONENTS have shortened activity rhythm period
OF THE CLOCK lengths, and a greater diversity in these
lengths, suggesting that REV-ERBa is nec-
So far, we have discussed the main compo- essary for maintaining precision of the
nents of the core circadian clock. These are clock. Such a phenotype is in contrast to
the transcriptional activators CLOCK and Bmal1 knockout mice, which lack circadian
BMAL1, and the transcriptional repressors, activity rhythms altogether. Furthermore,
PER and CRY. As we have pointed out, data suggest that the PER proteins are neg-
in mammals, the complexity of the core ative regulators of Rev-erba, and that Rev-
clock is reasonably high because multiple erba is activated by the CLOCK : BMAL1
homologs of PER and CRY exist. In this heterodimer. Thus, the negative effect of
section, we will expand on that complexity BMAL1 on its own transcription would be
by discussing numerous additional mole- through upregulation of its own repressor
cules for which there is evidence regarding (REV-ERBa).
a function in the mammalian clock.
DEC1 and DEC2
REV-ERBa Two bHLH transcription factors that do not
The four proteins, PER, CRY, CLOCK, and contain PAS domains were implicated in
BMAL1 constitute a feedback loop with circadian processes. These proteins, called
the two former proteins acting as negative DEC1 and DEC2 (so named because they
components and the two latter proteins were identified in differentiated human
acting as positive components. However, embryo chondrocytes) are related to the
RHY5 2/6/04 3:55 PM Page 120

120 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

HAIRY and ENHANCER OF SPLIT pro- the core oscillator is compromised in these
teins of Drosophila that function in neural transgenic mice. Additionally, VPAC2
development by repressing transcription. knockout mice show a unique circadian
Expression of both DEC genes cycles in the phenotype as assessed by locomotor behav-
SCN and in peripheral tissues. The phase of ior. These mice did have a circadian cycle of
the DEC1 and DEC2 RNA oscillation in locomotor activity in constant darkness,
the SCN lags slightly behind mPer1. Con- although the onset and offset of wheel
sistent with this fact, DEC1 and DEC2 can running were not well defined and the
strongly repress CLOCK : BMAL1 activa- overall amplitude of the rhythm was lower
tion of mPer1 in tissue culture. Interest- than in the wild type. Strikingly, however,
ingly, DEC1 expression is activated by a the phase of the rhythm in the knockout
light pulse much like expression of mPer1, animals was completely opposite that of
suggesting that the DECs may connect wild-type animals in freerunning condi-
input pathways to the core clock. Still, there tions. The mechanism underlying the
is no direct evidence yet implicating either reverse phase of activity is not clear, but
DEC protein in the core circadian clock or the authors hypothesize that pacemakers in
in peripheral oscillators. the brain other than the SCN may be
driving these rhythms. Although there is
little evidence for such a postulation, it is
CKId
clear that the oscillator in the SCN is com-
Evidence is accumulating that CKIe is not promised. Molecularly, cyclic expression of
the only kinase important for circadian the core genes, mPer1, mPer2, and mCry1 is
functions. All the core clock proteins previ- abolished in the knockout mice. Moreover,
ously described are phosphorylated in a cir- expression of these genes is at baseline
cadian-dependent manner. Casein kinase Id (trough) levels indicating that CLOCK :
(CKId) is highly homologous to CKIe and BMAL1 activity is compromised. Exactly
is also highly expressed in the SCN, how loss of VPAC2 results in a com-
although not rhythmically. It phosphory- promised core oscillator is not clear, but
lates all three mPER isoforms in vitro, and Hastings and colleagues propose that
is coimmunoprecipitated with CLOCK, intercellular communication within the
BMAL1, mPER1, mPER2, mCRY1, and SCN is important for maintaining the core
mCRY2 in vivo. In terms of circadian func- rhythm. Recall that VPAC2 binds VIP in
tions, CKId is probably not completely addition to PACAP. VIP is released from
redundant with CKIe, or presumably there SCN neurons. Therefore, VIP binding to
would not be as much of an effect on VPAC2 may regulate SCN neurons in a
rhythms in tau mutant animals. Therefore, paracrine manner to maintain the core
the functional significance of phosphoryla- oscillator.
tion by CKId is still being investigated.
Microarray Analysis
VPAC2
Microarray experiments show that many
As indicated above in the section on input genes cycle in the SCN. The development of
molecules, the PACAP receptor, VPAC2, microarray technology allows the simulta-
may have a role in maintaining the core neous assessment of transcript expression
clock. In mice expressing the human of literally tens of thousands of genes. It is
VPAC2 receptor transgene, the activity little wonder, then, that this technique is
rhythm was shortened to 8 hours in con- being applied to discover new genes that
stant darkness. This finding indicates that may be important for clock function. At
RHY5 2/6/04 3:55 PM Page 121

OTHER POSSIBLE COMPONENTS OF THE CLOCK 121

the time of this writing, two laboratories QTL Analysis


have reported microarray analysis of SCN
tissue collected from mice at various times Within the mouse species, different strains
of day. Both of these groups found that lit- are variable in the quantitative aspects of
erally hundreds of genes cycled in their circadian biology. For example, C57BL/6
expression in the SCN. The types of genes mice have a period of 24 hours, but Balb/c
could be grouped by functionality and mice have a period of slightly less than 23
included groups important for transcrip- hours, and CS mice have a period longer
tion, metabolism, protein synthesis, protein than 24 hours. A number of laboratories
processing, and protein secretion. Addi- have reasoned that the allelic differences at
tionally, a number of genes of unknown certain genes underlie this strain variability.
function were found to cycle in the SCN. Determining the genes would therefore
Both of these laboratories also reported provide additional insight into how genes
microarray analysis of gene expression in affect a complex behavior, such as circadian
mouse livers collected at different times locomotor activity. Such a task is easier said
of day. Again, literally hundreds of genes than done, though. The genetic variability
showed cyclic expression in this tissue, but among strains is so high that multiple vari-
surprisingly, the number of genes that ants might affect the behavior being
cycled in both the SCN and the liver was studies. Quantitative trait locus (QTL)
less than 30 in both cases. Such a finding analysis is a genetic technique that allows
indicated that cyclic expression is a tissue- study of such polygenic effects.
specific property. QTL is really a specialized form of
Although the wealth of data obtained genetic mapping. Two disparate strains of
from microarray analysis is extensive, to mice that differ in a circadian measurement
date it has not proved to be helpful for (e.g., period length) are mated. It is
identifying new core clock genes. One of important that a large number of genetic
the major problems has been a lack of markers exist that differentiate these
reproducibility of the data; that is, while two strains. With mice, this is not a problem
both laboratories identified hundreds of given the large number of SSLPs that have
genes that cycle in the SCN, there is sur- been identified. The F1 offspring are
prisingly little overlap between the two intercrossed to generate F2 offspring.
reported results. Additionally, although one These F2 progeny are assessed for the trait
would expect that the limited overlapping being tested. Given the heterogeneous
subset of cycling genes in the SCN and the genetic makeup of these F2 progeny, there
liver might reveal new core clock genes, is much more variability in this trait. The
even here there is little similarity between genomes of these mice are scanned for the
the two laboratories’ results. In truth, the genetic markers. Utilizing computer pro-
only genes that are reported by both labo- grams, one can determine the likelihood
ratories to cycle in both the SCN and the that a trait segregates with a specific
liver are the core clock genes already marker. Such markers are called QTLs, and
mentioned. Therefore, while microarray represent areas of the genome that have a
technology surely will give us a greater high probability of containing genes affect-
understanding of circadian biology, cur- ing the trait (Fig. 5.11). Candidate genes
rently we await additional experiments to from these areas can then be tested to
fully realize these benefits. determine if they regulate the trait under
study.
QTL analysis has been used to isolate
QTLs that affect wheel-running behavior in
RHY5 2/6/04 3:55 PM Page 122

122 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

Strain 1 (23.6 hours) Strain 2 (22.8 hours)

4 4 4 4

Interstrain crosses (F2 generation)

Period measurements
Map with molecular markers
Line 1: 23.3 hours
Line 2: 23.6 hours
Line 3: 23.6 hours

Line 1 2 3

Computer analysis –
links phenotype and genotype data

Circadian period QTL on chromosome 4,


allele from strain 2 contributes to short period

Figure 5.11. Schematic representation of QTL analysis for short-period phenotype in mice.
Two mouse strains that differ in circadian period are interbred to produce an F2 generation.
The genotypes of these animals are determined by mapping with molecular markers. The
circadian period of each of these is determined and correlated with the genotypic data. Only
chromosome 4 is shown here for both strains, as it is the one with the relevant QTL in this
case. In this simplified example, an F2 animal that is homozygous for a strain 2 allele at the
top of the chromosome has a shorter period than do F2 animals that are either heterozygous
or lack the allele. Analysis of numerous F2 animals determines whether this correlation is sig-
nificant. If so, then a QTL is established at the tip of chromosome 4. [Schematic is based on
Salathia, N, Edwards, K, Miller AJ (2002): QTL for timing: A natural diversity of clock genes.
Trends in Genetics 18: 115–118].

mice. Few of these map to the known cir- the genes associated with some of these
cadian genes. One hypothesis is that the QTLs will be cloned soon.
core circadian genes are so essential to cir-
cadian rhythms that little allelic variability
Other Agents
can be tolerated. In any case, identification
of these QTLs gives researchers new direc- Even though we now have a good
tions to identify additional genes involved understanding of the molecular basis of the
in regulating circadian rhythms. Hopefully, circadian clock, it is clear from both
RHY5 2/6/04 3:55 PM Page 123

OTHER POSSIBLE COMPONENTS OF THE CLOCK 123

microarray and QTL studies that additional tional GRP receptor. GRP may function
components may have subtle functions directly in the light-response pathway
affecting the clock. Additionally, the advent because GRP administration increases
of gene targeting technology has revealed mPer1 and mPer2 expression in the SCN
new functions of many genes. Many knock- much as a light pulse does.
out mice have disturbances in circadian
rhythms, although a number of these have
Fyn Kinase
underlying neural defects. Some com-
pounds for which there is experimental evi- Fyn kinase is a member of the Src family of
dence supporting a circadian clock function nonreceptor tyrosine kinases that is highly
are briefly mentioned below. expressed in the brain. Mice lacking fyn
kinase show significantly longer periods of
wheel-running behavior under freerunning
Serotonin
conditions, although they can entrain to
Serotonin is a neurotransmitter implicated light. However, these animals have addi-
in many behavioral processes in mammals. tional behavioral deficits, and the anatomic
As mentioned above, serotonergic neurons morphology of the SCN in these mice is
innervate the SCN in rodents via the indi- abnormal. Therefore, effects of the fyn
rect photic input system. There are multiple kinase deletion may be secondary to neu-
receptors for serotonin, making study of ronal defects.
the function of this molecule in circadian
biology somewhat complicated. Neverthe-
CaMKII
less, many studies using various serotonin
receptor agonists have shown suppression CREB is a known component necessary for
of light-induced responses at both the the induction of mPer1 and mPer2 expres-
behavioral and the molecular level in ham- sion in response to light. As outlined above,
sters. Moreover, depletion of serotonin CREB is phosphorylated in response to
enhances light-induced phase shifts in ham- an increase in intracellular calcium. The
sters. These results suggest the hypothesis calcium/calmodulin-dependent kinase II
that serotonin is inhibitory for photic (CaMKII) is one kinase that activates
entrainment. This story is complicated by CREB. CaMKII itself is phosphorylated in
the fact that similar results are not found in the SCN in response to a light pulse. Intra-
mice. Therefore, whether serotonin has a cerebral injection of a CaMKII inhibitor
general function in mammalian circadian into hamsters inhibited light-induced phase
rhythms, particularly with respect to input, delays of wheel-running rhythms. Also, this
is not clear. inhibitor attenuated the induction of
hamster Per1 and Per2 by a light pulse.
Therefore, CaMKII may have some func-
GRP
tion in the circadian responses to light.
Gastrin releasing peptide (GRP) is a neu-
rotransmitter found in some neurons in the
TIM Homologs
SCN, which respond to a light pulse by
increasing expression of c-Fos, a protein In mammals the role of Drosophila TIME-
marker of cellular activity. Also, GRP LESS is apparently played by mCRY1 and
administration in vivo elicits a phase shift mCRY2. In mammals, there does not
similar to a light pulse given at night. Such appear to be a direct homolog of TIM.
an effect is not seen in mice lacking a func- However, mammalian homologs of a TIM-
RHY5 2/6/04 3:55 PM Page 124

124 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

related Drosophila gene, called timeout, do but they do seem to have functions in envi-
exist. In mice, expression of this mam- ronmental sensing.
malian TIM (mTIM) is found in the SCN,
albeit at moderate levels. Whether mTim
NPAS2
expression cycles in the SCN is unclear.
Reports exist supporting both cyclic and NPAS2, a new PAS-domain-containing
noncyclic expression. It is not completely protein, functions in the forebrain. Steve
clear why disparate results exist, but mTim McKnight’s group searched the GenBank
may produce alternative transcripts which database for previously unidentified pro-
result in two forms of mTIM. Only one of teins that contained PAS domains. They
these may cycle. identified two which were selectively
Although there are some data suggesting expressed in the mammalian brain (fore-
a function for mTIM in the circadian clock, brain) and spinal cord. These were called
such a hypothesis has been difficult to study neuronal PAS domain protein (NPAS) 1
because the knockout of mTIM is embry- and NPAS2. These have also been termed
onically lethal. Embryos homozygous for member of PAS superfamily (MOP) 5 and
a disrupted mTim gene display cellular MOP4, respectively (Table 5.1). Expression
disorganization and necrosis as early as of both of these genes appeared to corre-
embryonic day 5.5, which is around the late with development of the brain. NPAS1
time of implantation into the uterus. Obvi- expression was first detected at the com-
ously, the function of mTIM is essential for mencement of brain organogenesis, and
early development of an embryo. Indeed, NPAS2 expression began shortly after
expression of mTim is found in many birth. In the adult, NPAS2 is expressed pri-
tissues in the developing embryo and the marily in the mammalian forebrain, an area
adult. More recent work suggests that involved in memory formation. A high
mTIM may function in regulating develop- degree of similarity between NPAS2 and
ment of tissues that undergo branching CLOCK was noted and NPAS2 was found
morphogenesis of tubules and ducts, such as to activate transcription by heterodimeriz-
the kidney. ing with BMAL1, much like CLOCK. This
In sum, the circadian function of mTIM observation raised the possibility that
is still open to question. The answer NPAS2 is involved in circadian rhythms.
may lie in developing tissue-specific knock- Furthermore, in tissue culture cells, expres-
outs of this gene. Interestingly, in flies, sion of NPAS2 and BMAL1 leads to
glycogen synthase kinase (GSK) phos- induction of mPer1, mPer2, and mCry1,
phorylates TIM, and regulates its levels but repression of Bmal1 expression.
and its subcellular localization. However, However, NPAS2 is not expressed in the
no such function for GSK has been found SCN, which would suggest that it does
in mice. not function in the central oscillator.
McKnight’s group went on to show that
normally, mPer1, mPer2, mCry1, and Bmal1
䊏 A FAMILY OF bHLH PAS all cycle in the mammalian forebrain.
DOMAIN PROTEINS Notably, Bmal1 cycles out of phase to the
former three. A knockout mouse of NPAS2
As we pointed out above, BMAL1 and was created and originally reported to have
CLOCK are both PAS-domain-containing deficits in memory formation. However,
bHLH transcription factors. It turns out further examination of the knockout mouse
that a family of these proteins exists. Not all revealed that cyclic expression of mPer2 in
of these are involved in circadian rhythms, the forebrain is disrupted. In areas of
䊏TABLE 5.1. Known PAS Proteins
Name Class of Protein Function PAS Protein Binding Partners Other Names
AHR Transcription factor Response to planar aromatic ARNT
RHY5 2/6/04 3:55 PM Page 125

hydrocarbons (PAHs)
HIF1a Transcription factor Hypoxic response ARNT, ARNT2 MOP1
HIF2a Transcription factor Hypoxic response ARNT, ARNT2 EPAS1, HLF, HRF,
MOP2
HIF3a Transcription factor Hypoxic response ARNT, ARNT2 MOP7
ARNT Transcription factor Hypoxic response, PAH response AHR, HIF1a, HIF2a, HIF3a HIF1b
ARNT2 Transcription factor Hypoxic response, neuronal development? HIF1a, HIF2a, HIF3a, SIM1
AHRR Transcription factor Inhibit AHR signal transduction ARNT
BMAL1 Transcription factor Circadian clock CLOCK, NPAS2 MOP3, ARNT3
CLOCK Transcription factor Circadian clock BMAL1, MOP9
PER1 Transcription factor Circadian clock PER1, PER2, PER3
PER2 Transcription factor Circadian clock PER1, PER2, PER3
PER3 Transcription factor Circadian clock PER1, PER2, PER3
NPAS1 Transcription factor Unknown MOP5
NPAS2 Transcription factor Peripheral clocks, memory formation BMAL1 MOP4
NPAS3 Transcription factor Unknown MOP6
SIM1 Transcription factor Neuronal development ARNT2
SIM2 Transcription factor Neuronal development
PASKIN Serine/threonine kinase Regulation of glycolysis? Unknown
MOP9 Transcription factor Regulation of blood clotting? CLOCK CLIF, BMAL2
SRC1 Nuclear receptor Potentiate transcriptional response to
coactivator steroid hormones
TIF2 Nuclear receptor Potentiate transcriptional response to GRIP1
coactivator steroid hormones
RAC3 Nuclear receptor Potentiate transcriptional response to ACTR, pCIP, AIB1,
coactivator steroid hormones TRAM1

125
RHY5 2/6/04 3:55 PM Page 126

126 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

the brain where NPAS2 is not expressed, protein 1 (EPAS1), which is found pre-
mPer2 levels continued to cycle. These ferentially in vascular endothelial cells.
data indicate that NPAS2 is involved in Another group of bHLH-PAS proteins
timekeeping, but presumably in non- act as coactivators for nuclear hormone
SCN rather than central tissues. More receptors. While none of the afore-
evidence to support this hypothesis is mentioned proteins are yet proven to be
reported below. Of note, little additional directly involved in circadian biology, it
information has been reported regarding may turn out that they interact with the
the function of NPAS1 since its identifica- central clock to provide a link to peripheral
tion in 1997. organ physiology.

Possible Function of MOP9 in


Peripheral Tissues 䊏 LARGELY UNKNOWN
OUTPUT PATHWAYS
Another bHLH-PAS protein was cloned
by a number of different groups in 2000.
Output Control by SCN
This has been called BMAL2, MOP9,
and cycle-like factor (CLIF). MOP9 is The preceding discussion demonstrates that
highly homologous to BMAL1. It is much progress has been made towards
expressed in multiple tissues including the determining the molecular bases underly-
SCN. Mop9 expression does not cycle either ing both the input to the clock and the core
in the SCN or peripheral tissues, much oscillator. However, the final part of the cir-
like Clock. It is clear, however, that MOP9 cadian system, the output pathway, is still
functions as a transcription factor. Multiple somewhat of a mystery. How the SCN con-
groups showed that MOP9 interacts trols the physiology of an organism is
with CLOCK to activate expression of largely unknown. Complicating discussion
reporter constructs in tissue culture cells. of this subject is the fact that outputs are
Moreover, a MOP9/CLOCK heterodimer measured by numerous, very disparate
activates expression of plasminogen activa- functions. For example, these can be hor-
tor inhibitor 1 (PAI1). PAI1 is a circadianly monal (Chapter 10), such as daily variations
expressed gene that regulates clotting activ- of testosterone; or behavioral, such as the
ity in blood vessels. Therefore, it is possible daily locomotor activities of rodents. More-
that MOP9 is more involved with periph- over, such outputs may be directly con-
eral oscillators (see text below) than with trolled by the SCN or may be regulated
the central clock, possibly with respect to indirectly through SCN-controlled inter-
hemostasis. mediaries. Such a dichotomy is borne out
through both anatomical and molecular
studies. Anatomically, distinct neural con-
Other PAS-Domain-Containing Proteins
nections from the SCN to other areas of the
As of today (2003), there is a whole family brain exist. These SCN-connected areas
of PAS-domain-containing bHLH tran- then connect to different parts of the body
scription factors. Not all of these are and regulate physiology. While such neu-
involved in circadian rhythms; however, roanatomical studies are quite useful, they
they all appear to have functions in envi- do not contain complete answers, because
ronmental sensing. These include the surprisingly, the SCN can also control
hypoxia inducible factors (HIFs), which rhythms via secreted factors. Molecularly,
activate genes in response to low oxygen output is defined primarily as substances
tension, and endothelial PAS domain that are rhythmically expressed by the
RHY5 2/6/04 3:55 PM Page 127

LARGELY UNKNOWN OUTPUT PATHWAYS 127

SCN. Genes regulated in this fashion are Hormone Release Regulated by SCN via
referred to as “clock-controlled genes” Distinct Neuroanatomic Pathways
(CCGs), and studies indicate that some
Many hormones show distinct circadian or
are transcriptionally regulated by the core
diurnal rhythms. Output pathways demon-
pacemaker. In this section, we discuss spe-
strating SCN control have been described
cific candidates for regulating distinct
for some of these. A more detailed dis-
output pathways, and attempt to place them
cussion of hormonal rhythms is found in
in a greater context with respect to the
Chapter 10; however, we briefly mention
SCN.
some of these here to illustrate examples of
SCN output pathways (Fig. 5.12).
SCN Connection to Distinct
Brain Areas
Melatonin
Just as there are a number of different input
pathways to the SCN, there are also a As mentioned above, melatonin can act as
number of different neuronal output path- an input factor on the SCN. But it also acts
ways. Most of these pathways connect to as an output factor, and primarily translates
other areas of the brain, particularly within the length of the photoperiod to the rest of
the hypothalamus. It is the connection to the organism. Melatonin is cyclically pro-
these areas that causes rhythmic release of duced by the pineal gland with levels
hormones that act as output molecules. highest at night. The pathway that controls
Retrograde tracing and immunohistochem- melatonin production is multisynaptic. The
istry have allowed the construction of an SCN synapses with the PVN, which in turn
anatomic map of the SCN output pathways. connects to the intermediolateral column
The main output pathway connects the of the spinal cord. This structure connects
SCN to the subparaventricular zone (SPZ) with the superior cervical ganglion, which
below the hypothalamic paraventricular then sends connections to the pineal gland.
nucleus (PVN). A few fibers also connect to Melatonin apparently has different effects
the dorsal medial nucleus of the hypothal- depending on the species of mammal
amus (DMH). Additional efferent connec- studied. In many species, seasonal varia-
tions run to the lateral geniculate nucleus tions in photoperiod, and thereby mela-
(LGN) and the raphe nuclei. These con- tonin production, regulate reproductive
nections may act as a modulatory influence activity. Humans, however, are not pho-
on the input signals that originate in the toperiodic, and the function of melatonin is
retina. We discuss the importance of some largely unknown, although it does have
of these connections in the next section. Of some sleep promoting effects. More infor-
course, there are additional efferents from mation on melatonin can be found in
the SCN, the function of which is still Chapter 10.
unknown. For example, connections to the
paraventricular nucleus of the thalamus
Corticosterone
(PVT) exist. The PVT regulates activity;
therefore, the SCN may regulate locomotor Corticosterone levels are circadianly regu-
activity through this connection. However, lated in mammals. Corticosterone is the
such a link has not been conclusively immediate precursor of aldosterone in
determined. mammals and therefore regulates sodium
retention and blood pressure. It is released
by the adrenal glands under the control of
adrenocorticotropic hormone (ACTH),
RHY5 2/6/04 3:55 PM Page 128

128 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

SCN
Vasopressin

DMH MPN

PVN

CRF GnRH
MELATONIN

IMLCSC
Anterior Pituitary

SCG
ACTH
LEUTEINIZING
HORMONE

Pineal Gland Adrenal Gland

MELATONIN CORTICOSTERONE
Figure 5.12. Output pathways of the SCN that regulate hormonal rhythms. The three most
widely described hormonal outputs of the circadian system are melatonin, corticosterone, and
leuteinizing hormone production. The melatonin pathway is shown connected by solid
arrows. The SCN connects to the paraventricular nucleus (PVN), which in turn connects to the
interomediolateral column of the spinal cord (IMLCSC). The IMLCSC connects to the pineal
gland via the suparcervical ganglion (SCG). The corticosterone pathway is shown by dotted
arrows. There are multiple pathways controlling the corticosterone rhythm: (1) similar to the
melatonin pathway, the SCN connects to the PVN, which in turn connects to the IMLCSC—
however, the IMLCSC connects directly to the adrenal glands; (2) the PVN also regulates cor-
ticosterone release by producing corticotroph releasing factor (CRF), which stimulates the
anterior pituitary to release adrenocorticotrophic hormone (ACTH, which stimulates the
adrenals); and (3) the SCN also connects to the CRF, producing neurons of the PVN indirectly
via the dorsal medial nucleus of the hypothalamus. The pathway regulating leuteinizing
hormone release is shown by dashed arrows. LH is released by the anterior pituitary in
response to gonadotrophin releasing hormone (GnRH) from the medial preoptic nucleus
(MPN) of the hypothalamus. The SCN regulates the MPN both directly and indirectly via rhyth-
mic vasopressin production.

which itself is released from the anterior directly to the adrenals via the interomedi-
pituitary gland. In turn, ACTH release is olateral column of the spinal cord. Together
under the control of neurons of the PVN these three pathways work to establish the
that release corticotroph releasing factor corticosterone rhythm.
(CRF). There are several routes of SCN
control over the corticosterone rhythm: (1)
Gonadotropins
there are direct synapses from the SCN to
the CRF producing neurons of the PVN; Luteinizing hormone (LH) is released by
(2) the SCN synapses with the DMH, which the anterior pituitary in a circadian fashion.
in turn synapses with the PVN; and (3) This hormone has different effects in males
there is a pathway that connects the PVN and females, but primarily regulates repro-
RHY5 2/6/04 3:55 PM Page 129

LARGELY UNKNOWN OUTPUT PATHWAYS 129

ductive functions. The medial preoptic regulated wheel-running behavior. To


nucleus (MPN) of the hypothalamus accomplish this goal, they first isolated
releases gonadotropin releasing hormones numerous peptides that are normally
(GnRH) in response to vasopressin from secreted by the SCN in hamsters. These
the SCN. There are also direct connections peptides were tested for effects on locomo-
from the SCN to estrogen-containing tor activity rhythms by infusing them into
neurons in the MPN. Therefore, the SCN the third ventricle of hamsters and observ-
has both direct and indirect ways of regu- ing their wheel-running rhythms. One
lating the gonadotropin rhythm. peptide, transforming growth factor a
(TGFa), completely and reversibly blocked
wheel running by the hamsters, suggesting
Output Control by SCN Using
that it acts as an inhibitory factor for loco-
Secreted Factors
motor activity. TGFa is a peptide signaling
As mentioned above, SCN transplants from molecule best known for its function in cel-
one animal can restore rhythms in an lular growth control and differentiation.
animal that has an ablation of the SCN. In The receptor for TGFa is the epidermal
1996, Silver and colleagues extended these growth factor receptor (EGFR). Strikingly,
findings to show that transplanted SCN do epidermal growth factor (EGF) also
not require synaptic connections to affect blocked locomotor activity in hamsters,
rhythm control. SCN transplants from tau further suggesting that TGFa acts through
mutant hamsters were encapsulated in a the EGFR. Additional work showed that
semipermeable membrane prior to trans- tgfa is rhythmically expressed in the SCN,
plant into wild-type animals with ablated and that EGFR is expressed in the SPZ. As
SCN. These encapsulated transplants did mentioned above, this area of the brain is
not form synaptic connections. However, innervated by the SCN. Lesions of the SPZ
recovery of circadian rhythms, as judged by demonstrate its importance for locomotor
locomotor activity, was still accomplished. rhythms. It was hypothesized that rhythmic
The recovered rhythms matched that of the release of TGFa from the SCN acts on
donor. These experiments proved that a EGFRs in the SPZ to regulate locomotor
secreted factor from the SCN could at least activity. However, mice with a mutation in
regulate circadian locomotor activity. Of the EGFR called waved-2, which reduces
course, these results did not rule out the receptor activity by 90%, were completely
necessity of synaptic connections for the rhythmic in constant darkness. In LD con-
control of other circadian rhythms. For ditions, waved-2 mice were rhythmic, but
example, certain rhythms are never were abnormally active during the light
restored by any transplants. These include time. The phenotype of these EGFR
reproductive responses to daylength and mutant mice indicates that this receptor
certain endocrine rhythms. Therefore, may actually mediate masking effects of
synaptic connections are necessary to main- light rather than the persistence of circa-
tain some rhythms. However, this classic dian rhythms under freerunning conditions.
study proved that a secreted factor from the
SCN was capable of controlling locomotor
Regulation of Locomotor Behavior
output.
by PK2
Another molecule that was implicated in
Regulation of Locomotor Activity
locomotor output in rodents is the secreted
by TGFa
protein, prokineticin 2 (PK2). PK2 was
Building on the abovementioned study, originally identified as a protein that causes
Weitz’ group sought to identify factors that contraction of gastrointestinal smooth
RHY5 2/6/04 3:55 PM Page 130

130 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

muscle. General examination of the expres- SCN. Presumably, not all of these necessar-
sion pattern of PK2 revealed that it is ily have output functions. However, the
expressed in the hypothalamus. Further receptor of PK2 (PK2R) is expressed in
study showed circadian expression in the numerous areas where the SCN has its con-
SCN, peaking during the first half of the nections, although this does not include the
subjective day. Genomic DNA analysis SPZ. The most compelling evidence that
revealed E boxes upstream of the PK2 PK2 is truly an output protein came from
gene. These E boxes are necessary for tran- studies involving the intracerebroventricu-
scription. Moreover, CLOCK and BMAL1 lar injection of PK2 in rats. Administration
can activate transcription of PK2, and this of PK2 in this way suppressed the wheel-
activation is blocked by the PERs and the running behavior of rats during the subjec-
CRYs. The oscillations of PK2 expression tive night. This effect is much like that seen
are disrupted in Clock mutant mice and for TGFa (Fig. 5.13).
mCry1/mCry2-deficient mice, indicating
that these core circadian proteins regulate
Arginine Vasopressin
PK2 expression in vivo. Of course, as men-
tioned above, microarray experiments show The classic example of a CCG is arginine
that expression of many genes cycles in the vasopressin (AVP), also known as antidi-

(a)
Days

TGF-a

(b)
Days
1

2
PK2
3

5
6

Figure 5.13. TGFa and prokineticin 2 both suppress locomotor activity. (a) Hamsters were
entrained to a 14–10 light : dark cycle for two weeks and then moved to constant darkness.
At the indicated time, TGFa was constantly infused into the third ventricle through an intra-
ventricular cannula. Locomotor activity almost immediately stopped, and very little activity
occurred while infusion was being carried out. When the infusion was stopped, the animals
were active again. (b) Similarly, rats subjected to recombinant human PK2 injection into the
ventricular system almost immediately stop running on wheels. Interestingly, there is abnor-
mal activity observed the day after the PK2 injection.
RHY5 2/6/04 3:55 PM Page 131

CLOCKS EVERYWHERE: PERIPHERAL OSCILLATORS 131

uretic hormone (ADH). AVP is a neu- mice have defects in circadian locomotor
ropeptide that is synthesized in the PVN activity (a 30-minute shorter period than
and the supraoptic nuclei (SON) of the wild type and overall less activity), they are
hypothalamus and then released from the still rhythmic in freerunning conditions.
posterior pituitary. AVP causes an organism Such findings indicate that dbp is more
to retain water and sodium; therefore, it reg- likely an output gene. Indeed, the dbp pro-
ulates osmotic pressure and blood pressure. moter contains E boxes, and transcription
AVP is also found in SCN neurons where of dbp is dependent on CLOCK. Further
both its synthesis and release are circadi- supporting an output function for DBP is
anly regulated. Synthesis of AVP is con- the fact that expression of this gene cycles
trolled at the transcriptional level in the not only in the SCN but also in numerous
SCN. Indeed, the avp gene contains E peripheral tissues. Moreover, disruption of
boxes, which are transcriptionally regulated DBP causes a lack of circadian expression
by the CLOCK : BMAL1 heterodimer. of other genes in the liver, many of which
Moreover, in Clock mutant mice, the level function in metabolism of steroid hor-
of avp is markedly diminished and non- mones. Therefore, DBP may serve to
rhythmic. In contrast, the generation of avp amplify circadian signals in nonneuronal
in the SON is unchanged in these mutant tissues by regulating circadian expression
mice, indicating that different regulatory of other genes in these tissues. This type of
elements are at work in different areas of function introduces the next topic: periph-
the brain. It could be argued that rhythmic eral oscillators.
expression of avp in the SCN implicates it
in the generation of the core oscillator.
However, in a rat model that lacks avp,
䊏 CLOCKS EVERYWHERE:
locomotor and drinking rhythms still exist,
PERIPHERAL OSCILLATORS
although there are slight differences in
the amplitude and entrainability of the
Peripheral Oscillators: Organs with
rhythms.As such,AVP is considered a CCG.
Their Own Clocks
As briefly mentioned numerous times
D-Element Binding Protein
above, one of the most intriguing aspects of
d-Element binding protein (DBP) is circadian outputs is the presence of tissue-
another classic example of a CCG. Albumin specific clocks in many organs other than
d-element binding protein (DBP) is a the brain. Evidence that peripheral oscilla-
member of the PAR-domain-containing tors exist comes from the fact that all the
basic leucine zipper family of transcription known circadian genes that cycle in the
factors. Members of this family contain an SCN—Bmal1, Per1, Per2, Per3, Cry1, and
amino-terminal activation domain that is Cry2—are also rhythmically expressed in
rich in proline and acidic residues, called nearly every organ examined. Additionally,
the PAR domain. Transcription of dbp as observed in the SCN, microarray exper-
oscillates strongly in the SCN, which origi- iments identified hundreds of genes that
nally suggested that it may be a component cycle in peripheral tissues. The purpose of
of the core oscillator. Also, DBP can bind these peripheral oscillators presumably is
to the mPer1 promoter and cooperate with to impart local control of circadian func-
CLOCK and BMAL1 to activate transcrip- tions in a tissue-specific manner.
tion. However, knockout mice lacking the Peripheral oscillators do not normally
dbp gene still show robust circadian gene function in a vacuum, however. The core
expression. In addition, although these circadian pacemaker in the SCN is hypoth-
RHY5 2/6/04 3:55 PM Page 132

132 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

esized to maintain control over these types of oscillators interact represent


peripheral clocks. Several lines of evidence another great unknown area of circadian
support this hypothesis: (1) peripheral biology. Notably, however, there are
oscillation of clock gene expression is dis- instances in which the slave oscillators can
rupted in SCN-lesioned animals; (2) the operate independently of the SCN. Such a
phase of rhythmic expression of the clock phenomenon illustrates that the peripheral
genes is delayed with respect to the phase oscillators can override the SCN control
of expression in the SCN, suggesting that when physiologically advantageous (Fig.
the SCN controls their expression; and (3) 5.14).
peripheral oscillation of clock gene expres-
sion in explanted tissues dampens after
Peripheral Oscillators: Genes that Cycle
about a week in culture. In contrast, sec-
in the Periphery
tions of SCN will continue to cycle indefi-
nitely. These lines of evidence have yielded Just as is seen in the SCN, literally hundreds
the theory that the SCN houses the “master of genes cycle in peripheral organs of the
oscillator” which is in control of numerous rodent. This is borne out by microarray
“slave oscillators” found in peripheral studies. Strikingly, again, the subset of genes
tissues. The mechanisms by which these two that cycles in one tissue is quite different

Master oscillator

Restricted feeding or
other Input

Liver

Kidney
SLAVE OSCILLATORS

Figure 5.14. Master–slave oscillator relationship. Under normal conditions, the core clock in
the SCN elicits control over all other peripheral oscillators, represented by the liver and the
kidney. Control may be by indirect neuronal connections, secreted factors, or a combination
of both. These peripheral oscillators have a phase that lags the core clock by approximately
4 hours. Under conditions of stress, such as restricted feeding, the slave oscillators can be lib-
erated from the master oscillator. The phase of oscillations in peripheral organs will change,
but the phase of oscillations in the SCN remains unchanged. The mechanisms underlying both
the uncoupling event and the imposition of a new phase are unknown.
RHY5 2/6/04 3:55 PM Page 133

CLOCKS EVERYWHERE: PERIPHERAL OSCILLATORS 133

from that in another tissue, further under- sion in the periphery. There are some data
lining the possibility of tissue specific func- from studies of chickens that E4BP4 may
tions for the peripheral oscillators. Some of also function in entrainment by light;
the best studied genes that show oscillatory however, such experiments have not yet
expression in peripheral tissues are dis- been performed in mammals.
cussed below. Notably, although many of
these also cycle in the SCN, they are con-
REV-ERBa
sidered output genes.
The orphan nuclear receptor REV-ERBa
was discussed above as a possible compo-
PAR Proteins
nent of the core oscillator in the SCN.
The classic example of a PAR protein is However, like many core clock genes, rev-
DBP. As discussed above, expression of this erba is circadianly expressed in peripheral
gene cycles in the SCN, but also cycles tissues. Such peripheral circadian expres-
robustly in most organs of the body. sion has been best studied in the liver.
Expression of two other PAR domain Indeed, REV-ERBa was originally impli-
genes, hepatic leukemia factor (hlf), and cated in lipoprotein metabolism and adipo-
thyrotroph embryonic factor (tef), also genesis. Possibly REV-ERBa regulates
cycles robustly in the liver (and in the circadian aspects of metabolism in the liver.
SCN), suggesting that PAR proteins func- It is clear from knockout mice that core
tion generally in circadian output. clock gene expression is disrupted in the
However, the exact function of PAR pro- liver of these knockout mice. However, no
teins in peripheral oscillators is not com- information is yet available regarding any
pletely clear. Although expression of dbp is disruption to liver function in these mice.
regulated by CLOCK, such regulation has
not been shown for other PAR genes.
RORb
The retinoid-related orphan receptor b
E4BP4
(RORb) is another orphan nuclear recep-
E4BP4 is a basic leucine zipper trans- tor. RORb is expressed primarily in the
cription factor originally isolated as an E4 central nervous system, although it is also
adenovirus E4 promoter binding protein. found in the retina and in the pineal gland.
E4BP4 lacks a PAR domain, but its DNA Interestingly, although expressed in the
binding domain is homologous to those in SCN, rorb expression does not cycle there.
DBP, TEF, and HLF. E4BP4 is the mam- However, there is robust oscillation of rorb
malian homolog of the Drosophila gene in both the retina and the pineal gland, sug-
vrille, which is necessary for circadian gesting that it has a function in peripheral
rhythms. Examination of e4bp4 expression oscillators in these tissues. Mice lacking rorb
revealed that it is circadianly expressed in were generated.These mice have a neuronal
both the SCN and that of peripheral tissues, phenotype that may result from develop-
much like the PAR genes. However, E4BP4 mental defects. Additionally, RORb-
cycles in a phase opposite DBP. Moreover, deficient mice show retinal degeneration;
E4BP4 acts as a transcriptional repressor however, they are still able to entrain to
and competes for the same DNA binding light dark cycles, indicating that a functional
site as the PAR proteins. According to input pathway exists. In freerunning condi-
those data, it is hypothesized that E4BP4 tions, the locmotor activity period of these
and the PAR proteins act in opposite direc- knockout mice increased by approximately
tions to “fine-tune” circadian gene expres- 25 minutes. Such a phenotype may result
RHY5 2/6/04 3:55 PM Page 134

134 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

from a defect intrinsic to the core oscillator this is the case for individual cells in tissue
or to defective feedback from the pineal culture. Schibler’s group in 1998 demon-
gland. However, measurements of pineal strated that a serum shock delivered to
function, namely, melatonin production, both a Rat-1 fibroblast cell line and a rat
have not been performed. Such measure- hepatoma (liver) cell line induced cyclic
ments are necessary to fully elucidate the oscillations of the rat Per1 and Per2 genes.
function of RORb in peripheral oscillators. Moreover, cyclic expression of the tran-
Notably, the RORb-related proteins, RORa scription factors rev-erba, tef, and dbp was
and RORg, may also regulate circadian also induced by serum shock. The periods
gene expression in the liver because they of these rhythms were 22.5 hours and oscil-
form complexes with promoter elements latory expression could be maintained for
in a cyclic manner. As such, the ROR family at least three cycles in vitro. Strikingly,
of proteins may be general regulators of the temporal sequence of the individual
chronobiology in the periphery. rhythms mimicked that found in the liver.
That is, normally in the liver the peak of
rev-erba expression precedes that of Per1
Microarray Experiments
and Per2. The same result was found in the
Microarray experiments have been per- cell lines following the serum shock. Taken
formed on peripheral tissues collected over together, these results indicated that the
a 24-hour period just as has been per- clockworks were intrinsic to many cells in
formed on the SCN. Many tissues have the body. Additional studies have shown
been profiled, and it is a safe assumption that numerous mammalian cell lines,
that more profiling is occurring as this is including murine fibroblasts and vascular
written. While most of these experiments endothelial cells, display rhythmic clock
need to be repeated and individual gene gene expression on serum shock. The estab-
cycling confirmed, a few conclusions can be lishment of circadian gene expression in
drawn: (1) in each tissue, literally hundreds tissue culture cells was an extremely impor-
of genes show circadian expression in tant finding for it opened up a new, easily
individual tissues; (2) there is little overlap manipulated model system to study periph-
between the subsets of genes that cycle in eral oscillators.
the different tissues; (3) many genes that
are involved in specific cellular processes,
Retinoid Acid Receptor (RAR)
such as glycolysis, all show cyclic expression
in the same tissue; and (4) the number and Sometimes in science, a laboratory search-
diversity of genes that show cyclic expres- ing for an answer to one question fortu-
sion suggest that the core clock transcrip- itously discovers an answer to another.
tion factors are not sufficient for directly FitzGerald and colleagues made just such a
driving rhythmic expression in peripheral discovery that lent great insight into periph-
tissues. As more and more microarray eral oscillator mechanisms. To examine
experiments are performed, a greater mechanisms that control vascular smooth
understanding of how cyclic expression is muscle contraction, a search for protein
established will emerge. binding partners of the retinoid X receptor
(RXR) was initiated. This screen yielded
NPAS2. Additional experiments showed
Oscillators Revealed in Every Cell
that RXR also interacted with CLOCK.
Through Tissue Culture
This interaction was specific. Only CLOCK
Recall that the SCN is composed of multi- and NPAS2 interacted with RXR. BMAL1
ple oscillators in individual cells. Strikingly, did not. Also, interaction was found only
RHY5 2/6/04 3:55 PM Page 135

CLOCKS EVERYWHERE: PERIPHERAL OSCILLATORS 135

with RXR and the highly related retinoic expression in fibroblasts grown in culture.
acid receptor (RAR). Binding to other Moreover, dexamethasone injection into
steroid receptors such as peroxisome pro- mice shifts the phase of circadian RNA
liferator-activated receptor (PPAR) and expression in liver, kidney and heart. No
thyroid hormone receptor (TR) did not shift was seen in the SCN, which does not
occur. The interaction with the clock pro- express the glucocorticoid receptor. To
teins was greatly enhanced by retinoic acid prove their hypothesis, Schibler’s group
(RA) binding to either the RXR or the used a mouse line that has an inactive
RAR, suggesting that RA might affect a glucocorticoid receptor only in the liver.
function of the interacting protein complex. Dexamethasone injection into these mice
A functional importance for this interac- phase shifted circadian gene expression in
tion was provided by the finding that RAR heart and kidney, but not in liver. These
and RXR both blocked transcriptional studies were quite beneficial because they
activation by the NPAS2:BMAL1 and link a specific cellular pathway to changes
CLOCK:BMAL1 heterodimers in tissue in peripheral circadian gene expression.
culture cells. Expression of all the core The importance of this pathway under
clock genes cycles in the vascular smooth- physiological conditions has yet to be deter-
muscle cells. Most importantly, this cyclic mined however.
gene expression is phase-shifted on injec-
tion of retinoic acid into mice. These exper-
Restricted Feeding
iments established a direct mechanism by
which a steroid hormone could affect circa- Restricted feeding can uncouple peripheral
dian gene transcription by the core clock oscillators from SCN control. Restricted
components. feeding had long been known to entrain
Given the abovementioned circadian locomotor activity in rodents. Normally
functions of the REV-ERB and ROR fam- rodents feed at night, when they are active.
ilies of nuclear receptors, the possibility is However, if food availability is restricted
emerging that the nuclear receptor protein to the daytime, these animals will become
family has a general function in peripheral active a few hours prior to food availability,
oscillators, much like the PAR family of such that they are now active during the light
proteins. Additional data supporting this phase. Two groups examined the effects of
hypothesis come from studies regarding restricted feeding on the molecular oscilla-
the glucocorticoid receptor, discussed in the tions seen in peripheral tissues and in the
next section. SCN. Both groups found similar results.
Altered feeding regimens shifted the phase
of cyclic expression of both core clock genes
Glucocorticoids
and other genes in peripheral tissues. Of
Glucocorticoids can phase-shift peripheral note, the phase shift occurred faster in the
circadian gene expression. The finding that liver than in other organs such as the lung
a serum shock induces circadian gene and the kidney. It is unclear why this differ-
expression in fibroblasts suggested that a ential response occurs. It is possibly due to
bloodborne factor could signal circadian differences in sensitivity to an entraining
time to organs in the body. Schibler’s group factor. Notably, the liver is intimately related
reasoned that glucocorticoids were good to feeding; therefore it makes intuitive sense
candidates for such signaling factors that such an organ would be more sensitive
because they are expressed in circadian to feeding regimens.
cycles. Consistent with this hypothesis, Possibly the most striking finding from
dexamethasone induces circadian gene these experiments, however, was that
RHY5 2/6/04 3:55 PM Page 136

136 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

restricted feeding did not induce phase in peripheral tissues. Using a neuroblas-
shifts of gene oscillations in the SCN. This toma cell line, McKnight’s group identified
finding established that peripheral oscilla- a number of target genes activated by the
tors could be uncoupled from the SCN. NPAS2:BMAL1 heterodimer. One such
Therefore, while the SCN normally does act gene is lactate dehydrogenase A (LdhA),
as a master pacemaker over the peripheral, which catalyzes the reduction of pyruvate
slave oscillators, these oscillators can be to lactate. While not obviously interesting,
“liberated” under the right conditions. Such McKnight’s group realized that LDHA
a dichotomy may allow an organism local utilizes reduced nicotinamide adenine din-
control over specific organs and thus to ucleotide (NADH) as a cofactor, generat-
better respond to abrupt shifts in environ- ing the oxidized form, NAD+. They
mental conditions. reasoned that these cofactors might have
Because glucocorticoid release is inti- influence over the activity of the
mately tied to feeding, it was possible NPAS2:BMAL1 heterodimer. They found
that restricted feeding-induced phase shift- that DNA binding activity was enhanced by
ing was acting through glucocorticoids. NADH (reduced form) and inhibited by
However, several lines of evidence indicate NAD (oxidized form). Because the
that this is not the case: (1) a line of mice absolute cellular concentration of cofactor
that have a mutated glucocorticoid receptor is constant, it was hypothesized that binding
specifically expressed in the liver can be is a function of the ratio of the reduced to
entrained by restricted feeding as measured the oxidized form. Consistent with this
by cyclic gene expression in the liver and hypothesis, maximal binding was found
(2) repeated injections of corticosterone when the ratio of NADH to NAD was at
into mice fed ad libitum at times when this least 75%. Interestingly, the binding of
hormone peaks in feeding-restricted CLOCK:BMAL1 heterodimers was also
animals do not shift oscillations in the liver. enhanced by NADH. However, NADH had
The function of glucocorticoids may actu- no effect on DNA-binding of BMAL1:
ally be to inhibit food-induced phase shift- BMAL1 homodimers. Therefore, it appears
ing of peripheral oscillators when animals that only NPAS2 and CLOCK are respon-
are fed in the daytime. Consistent with this sive to NADH. Indeed, using deletion
hypothesis, adrenalectomized mice respond mutations, the responsive site in NPAS2,
with more rapid phase shifting of circadian and presumably CLOCK, was localized to
gene expression in the liver in response the bHLH domain; the PAS domain was
to food restriction than did nonmanipulated dispensable for responsiveness.
animals. Of course, numerous hormonal The oxidation state of NADH may affect
and signaling pathway changes occur in transcriptional activity in two ways that
response to feeding, so there may not be a are not mutually exclusive: (1) it
single signal that is responsible for food- enhances binding of heterodimers to
induced shifting of peripheral oscillators. DNA and therefore presumably enhances
Alternatively, changes in cellular metabolic transcriptional activity, and (2) NADH
state in response to feeding may be apparently promotes the formation of
involved, as is discussed in the next section. NPAS2:BMAL1 heterodimers at the
expense of BMAL1:BMAL1 homodimers.
This sets up an elegant system such that
The Cellular Redox State
when the reduced form is high, transcrip-
Recall from above that NPAS2 is a tional activation occurs. However, when the
homolog of CLOCK that can heterodimer- oxidized form is high, BMAL1:BMAL1
ize with BMAL1 and activate transcription homodimers, which are transcriptionally
RHY5 2/6/04 3:55 PM Page 137

CLOCK COMPONENT MUTATION LEADING TO SPECIFIC DISORDERS 137

inactive, bind, and transcriptional repres- sarily reproduce what would happen in the
sion is the result. wild. To fully understand the importance of
The finding that redox state can affect an intact circadian clock, field studies must
DNA binding by the core circadian tran- be performed. Such heroic studies are
scription factors raises intriguing questions actually being undertaken using SCN-
regarding food-induced entrainment. The lesioned ground squirrels released into the
presence of food causes cells to activate wild. These studies suggest that the SCN-
metabolic pathways, which results in higher lesioned animals are more likely to fall
concentrations of reduced cofactors. For prey to predators than are their non-
example, feeding activates glycolysis and lesioned counterparts possibly because of
thus generates NADH. In contrast, food excessive nocturnal activity. However,
restriction results in utilization of reduced while we await results from more field
cofactors. Although food-induced entrain- studies, familial genetics and careful labo-
ment may be influenced by a number of ratory analyses are at least proving to us
different mechanisms, redox state is a novel that disruption of specific clock compo-
possibility. nents can have profound effects on an
organism.

䊏 CLOCK COMPONENT MUTATION


Sleep Disorders
LEADING TO SPECIFIC DISORDERS
One of the most exciting findings is the
Most of the preceding sections focused on demonstration that humans with a mutated
describing the current state of knowledge Per2 gene display a specific sleep disorder.
regarding the molecular basis for circadian Patients with familial advanced sleep phase
rhythms. Obviously, much has been learned syndrome (FASPS) are characterized by a
since, but still more remains to be under- normal sleep pattern, but it is advanced by
stood. One aspect of circadian biology that approximately 4 hours. That is, these
has not been adequately addressed is how patients fall asleep at around 7:30 pm every
all the molecular data fit into the larger evening and spontaneously awake at
picture of organismal physiology and around 4:30 am every morning. Patients
behavior. Everyone seems to agree that with this disorder also have advanced mela-
circadian rhythms are important. Indeed, tonin and temperature rhythms. FASPS is
it is hard to believe that Mother Nature inherited as a simple autosomal dominant
would put in place a conserved timekeep- disorder, indicating that it is a single-gene
ing system in organisms from cyanobacte- mutation. Ptácek and colleagues originally
ria to mammals, unless such a system was mapped the FASPS gene to the telomeric
vital. It would stand to reason, therefore, portion of the long arm of chromosome 2
that disruption of this system, à la murine in a family afflicted with this disorder. Inter-
gene knockouts, would have profound estingly, the human Per2 gene resides in the
effects on the animal. Surprisingly, this does same area as the putative FASPS gene.
not seem to be the case. Aside from running Because it is a circadian gene, Per2 was an
on wheels in the middle of the day, so far excellent candidate for the FASPS gene.
the animals with artificially disrupted circa- Sequencing of the Per2 gene from affected
dian rhythms appear to be just fine. There- individuals showed a transition from A to
fore, we are left with the question, is a G that changed a serine to a glycine. This
circadian clock necessary? However, substitution occurs in the binding site for
remember that utilizing mammals under the human casein kinase Ie protein, which,
strict laboratory conditions does not neces- you may recall, is the homolog of the
RHY5 2/6/04 3:55 PM Page 138

138 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

hamster TAU protein. Moreover, this muta- 䊏 CONCLUSION


tion leads to hypophosphorylation of
PER2. Taken together, these findings This chapter has attempted to introduce the
strongly suggest that the PER2 mutation is reader to the rapidly expanding world of
the cause of FASPS in this afflicted family, molecular circadian biology in mammals.
and provide the first link between circadian Notably, mammalian clockworks are very
gene function and human disease. similar to those of insects. However, in
many instances it appears that the issue is
PER2 as a Possible Tumor Suppressor more complicated. Also, this chapter is very
rodent-centric. As pointed out in the intro-
Another extremely exciting finding is that duction, it is a fair assumption that there are
PER2 may have a role in oncogenesis, differences in aspects of circadian biology
specifically as a tumor suppressor. Lee and between mice and humans. These differ-
colleagues observed that PER2 knockout ences are illustrated by the mere fact that
mice were more prone to develop tumors one is nocturnal and the other diurnal.
with age than were wild-type mice. These However, the rodent model has been
mice specifically developed salivary gland extremely useful in our understanding of
hyperplasia, genital teratomas, and lym- mammalian circadian biology and no doubt
phomas. Moreover, these knockout mice will continue to be so.
are more sensitive to g irradiation than are
wild-type mice as assessed by tumor for-
mation, primarily lymphoma. Also, thymo- FURTHER READING
cytes from the knockout mice are resistant
to apoptosis induced by g irradiation. Nor- Entries are listed in order of relevance or prior-
mally, thymocytes will die by apoptosis in ity, not alphabetically.
response to irradiation. If they do not, they
may accumulate genetic defects that lead to Photoreception and Input
cancer formation. Mechanistically, Lee and
Ginty, DD, Kornhauser, JM, Thompson, MA,
colleagues found that the tumor suppres- Bading, H, Mayo, KE, Takahashi, JS,
sor, p53 is not induced by irradiation in Greenberg, ME (1993): Regulation of CREB
PER2-deficient thymocytes. Such a finding phosphorylation in the suprachiasmatic
suggested that PER2 functions in the tran- nucleus by light and a circadian clock.
scriptional response to irradiation. Con- Science 260: 238–241.
sistent with this hypothesis, all the core Obrietan, K, Impey, S, Storm, DR (1998): Light
circadian clock genes are induced by irra- and circadian rhythmicity regulate MAP
diation, but such induction is abrogated in kinase activation in the suprachiasmatic
PER2 knockout animals. Additionally, the nuclei. Nat Neurosci 8: 693–700.
oncogene, c-myc, is normally expressed in a Shigeyoshi,Y,Taguchi, K,Yamamoto, S,Takekida,
S,Yan, L,Tei, H, Moriya,T, Shibata, S, Loros, JJ,
cyclic fashion in peripheral tissues. In PER2
Dunlap,JC,Okamura,H (1997):Light-induced
knockout animals, though, c-myc expres- resetting of a mammalian circadian clock is
sion is still cyclic, but is greatly upregulated. associated with rapid induction of the mPer1
This finding led to the proposal that PER2- transcript. Cell 91: 1043–1053.
deficient mice are more susceptible to Albrecht, U, Sun, ZS, Eichele, G, Lee, CC (1997):
cancer formation because of an inability A differential response of two putative
to properly regulate genes involved in cell mammalian circadian regulators, mper1 and
growth control. mper2, to light. Cell 91: 1055–1064.
Shearman, LP, Zylka, MJ, Weaver, DR,
Kolakowski, LF, Jr., Reppert, SM (1997): Two
RHY5 2/6/04 3:55 PM Page 139

FURTHER READING 139

period homologs: Circadian expression and Lowrey, PL, Shimomura, K, Antoch, MP,
photic regulation in the suprachiasmatic Yamazaki, S, Zemenides, PD, Ralph, MR,
nuclei. Neuron 19: 1261–1269. Menaker, M, Takahashi, JS (2000): Positional
syntenic cloning and functional characteriza-
Central Oscillator tion of the mammalian circadian mutation
Moore, RY, Eichler, VB (1972): Loss of a circa- tau. Science 288: 483–492.
dian adrenal corticosterone rhythm following
Additional Components
suprachiasmatic lesions in the rat. Brain Res
42: 201–206. Panda, S, Antoch, MP, Miller, BH, Su, AI,
Welsh, DK, Logothetis, DE, Meister, M, Schook, AB, Straume, M, Schultz, PG, Kay,
Reppert, SM (1995): Individual neurons SA, Takahashi, JS, Hogenesch, JB (2002):
dissociated from rat suprachiasmatic nucleus Coordinated transcription of key pathways in
express independently phased circadian the mouse by the circadian clock. Cell 109:
firing rhythms. Neuron 14: 697–706. 307-320.
Low-Zeddies, SS, Takahashi, JS (2001): Chimera Ueda, HR, Chen, W, Adachi, A, Wakamatsu, H,
analysis of the Clock mutation in mice shows Hayashi, S, Takasugi, T, Nagano, M,
that complex cellular integration determines Nakahama, K, Suzuki, Y, Sugano, S. Iino,
circadian behavior. Cell 105: 25–42. M, Shigeyoshi, Y, Hashimoto, S (2002): A
transcription factor response element for
Core Components gene expression during circadian night.
Vitaterna, MH, King, DP, Chang, AM, Nature 418: 534–539.
Kornhauser, JM, Lowrey, PL, McDonald, JD,
Output Pathways
Dove, WF, Pinto, LH, Turek, FW, Takahashi,
JS (1994): Mutagenesis and mapping of a Silver, R, LeSauter, J, Tresco, PA, Lehman, MN
mouse gene, Clock, essential for circadian (1996): A diffusible coupling signal from
behavior. Science 264: 719–725. the transplanted suprachiasmatic nucleus
King, DP, Zhao, Y, Sangoram, AM, Wilsbacher, controlling circadian locomotor rhythms.
LD, Tanaka, M, Antoch, MP, Steeves, TD, Nature 382: 810–813.
Vitaterna, MH, Kornhauser, JM, Lowrey, PL,
Turek, FW, Takahashi, JS (1997): Positional Peripheral Oscillators
cloning of the mouse circadian Clock gene. Akhtar, RA, Reddy, AB, Maywood, ES, Clayton,
Cell 89: 641–653. JD, King, VM, Smith, AG, Gant, TW,
Antoch, MP, Song, EJ, Chang, AM, Vitaterna, Hastings, MH, Kyriacou, CP (2002): Circa-
MH, Zhao, Y, Wilsbacher, LD, Sangoram, dian cycling of the mouse liver transcriptome,
AM, King, DP, Pinto, LH, Takahashi, JS as revealed by cDNA microarray, is driven by
(1997): Functional identification of the the suprachiasmatic nucleus. Curr Biol 12:
mouse circadian Clock gene by transgenic 540–550.
BAC rescue. Cell 89: 655–667. Storch, KF, Lipan, O, Leykin, I, Viswanathan, N,
Gekakis, N, Staknis, D, Nguyen, HB, Davis, FC, Davis, FC, Wong, WH, Weitz, CJ (2002):
Wilsbacher, LD, King, DP, Takahashi, JS, Extensive and divergent circadian gene
Weitz, CJ (1998): Role of the CLOCK expression in liver and heart. Nature 417:
protein in the mammalian circadian mecha- 78–83.
nism. Science 280: 1564–1569. Balsalobre, A, Damiola, F, Schibler, U (1998): A
Thresher, RJ, Vitaterna, MH, Miyamoto, Y, serum shock induces circadian gene expres-
Kazantsev, A, Hsu, DS, Petit, C, Selby, CP, sion in mammalian tissue culture cells. Cell
Dawut, L, Smithies, O, Takahashi, JS, Sancar, 93: 929–937.
A (1998): Role of mouse cryptochrome blue- McNamara, P, Seo, SP, Rudic, RD, Sehgal, A,
light photoreceptor in circadian photore- Chakravarti, D, FitzGerald, GA (2001): Reg-
sponses. Science 282: 1490–1494. ulation of CLOCK and MOP4 by nuclear
Ralph, MR, Menaker, M (1988): A mutation of hormone receptors in the vasculature: A
the circadian system in golden hamsters. humoral mechanism to reset a peripheral
Science 241: 1225–1227. clock. Cell 105: 877–889.
RHY5 2/6/04 3:55 PM Page 140

140 GENETIC BASIS FOR CIRCADIAN RHYTHMS IN MAMMALS

Balsalobre, A, Brown, SA, Marcacci, L, Tronche, DNA binding by the redox state of NAD
F, Kellendonk, C, Reichardt, HM, Schutz, G, cofactors. Science 293: 510–514.
Schibler, U (2000): Resetting of circadian
time in peripheral tissues by glucocorticoid Disorders
signaling. Science 289: 2344–2347. DeCoursey, PJ, Walker, JK, Smith, SA (2000):
Damiola, F, Le Minh, N, Preitner, N, Kornmann, A circadian pacemaker in free-living
B, Fleury-Olela, F, Schibler, U (2000): chipmunks: essential for survival? J Comp
Restricted feeding uncouples circadian oscil- Physiol A—Sens Neural Behav Physiol 186:
lators in peripheral tissues from the central 169–180.
pacemaker in the suprachiasmatic nucleus. Toh, KL, Jones, CR, He, Y, Eide, EJ, Hinz, WA,
Genes Dev 14: 2950–2961. Virshup, DM, Ptacek, LJ, Fu, Y-H (2001): An
Yamazaki, S, Numano, R, Abe, M, Hida, A, hPer2 phosphorylation site mutation in
Takahashi, R, Ueda, M, Block, GD, Sakaki, Y, Familial Advanced Sleep Phase Syndrome.
Menaker, M, Tei, H (2000): Resetting central Science 291: 1040–1043.
and peripheral circadian oscillators in trans- Fu, L, Pelicano, H, Liu, J, Huang, P, Lee, C (2002):
genic rats. Science 288: 682–685. The circadian gene Period2 plays an impor-
Rutter, J, Reick, M, Wu, LC, and McKnight, SL tant role in tumor suppression and DNA
(2001): Regulation of Clock and NPAS2 damage response in vivo. Cell 111: 41–50.
RHY6 2/6/04 3:55 PM Page 141

Part III
MOLECULAR CONTROL OF
CIRCADIAN RHYTHMS: FROM
CYANOBACTERIA TO PLANTS
RHY6 2/6/04 3:55 PM Page 143

6
CIRCADIAN RHYTHMS IN
CYANOBACTERIA
Nirinjini Naidoo

䊏 INTRODUCTION Cyanobacteria as Photoautotrophs


and Diazotrophs
Cyanobacteria: The Simplest Organisms Cyanobacteria are aquatic, prokaryotic
Known to Possess Circadian Clocks organisms capable of photosynthesis.
The circadian clock was initially assumed These organisms possess chlorophyll a and
to exist only in eukaryotes. Prokaryotic they split water and reduce CO2 to produce
cells, either multicellular or unicellular, oxygen and carbohydrate, respectively:
were considered too simple to express cir-
visible light
cadian behavior. Despite the fact that no 6CO2 + 12H2O (C6H12O6)
evidence existed to support such an idea, it chlorophyll
persisted and became accepted dogma. + 6H2O + 6O2
However, since the early 1990s, circadian
rhythms have been observed and reported They also possess two other pigments,
in several strains of cyanobacteria, making blue phycocyanin and red phycoerythrin.
them the simplest organisms known to Cyanobacteria, often referred to as blue-
possess a circadian clock. Most of the green algae because of the presence of
studies on cyanobacterial rhythms have phycocyanin, are in fact bacteria. They
been carried out on a unicellular strain comprise one of the largest and most
called Synechococcus elongatus. important groups of bacteria on earth.

Molecular Biology of Circadian Rhythms. Edited by Amita Sehgal


ISBN 0-471-41824-2 Copyright © 2004 John Wiley & Sons, Inc.

143
RHY6 2/6/04 3:55 PM Page 144

144 CIRCADIAN RHYTHMS IN CYANOBACTERIA

They are among the oldest known fossils at are unicellular; whereas others are filamen-
around 3.5 billion years and have played tous. Many of both types grow in colonies
an important role in the evolutionary and (Fig. 6.2).
ecological history of the Earth (Fig. 6.1). In addition to producing oxygen, many
The oxygen atmosphere was generated by species of cyanobacteria also fix nitrogen.
cyanobacteria during the Archaean and These cyanobacteria are photoautotrophs
Proterozoic eras. In addition, cyanobacteria and diazotrophs. Diazotrophs are organ-
residing in plant cells gave rise to the isms that can convert atmospheric nitrogen
modern chloroplast. Some cyanobacteria (N2) into biologically necessary compounds

Figure 6.1. Stromatolites—3.5-billion-year-old cyanobacterial fossils. Stromatolite layers


were produced as calcium carbonate precipitated over the growing mat of bacterial filaments.
The minerals, along with grains of sediment precipitating from the water, were trapped
within the sticky layer of mucilage that surrounds the bacterial colonies. The colonies con-
tinued to grow upward through the sediment to form new layers. [Pictures courtesy of Isao
Inouye (University of Tsukuba), Mark Schneegurt (Wichita State University), and Cyanosite
(www.cyanosite.bio.purdue.edu).]
RHY6 2/6/04 3:55 PM Page 145

INTRODUCTION 145

b
Figure 6.2. (a) Colony of Microcystis sp. Cyanobacteria are quite small and usually unicellular,
although they often grow in colonies large enough to see. [Picture courtesy of Isao Inouye (Uni-
versity of Tsukuba), Mark Schneegurt (Wichita State University), and Cyanosite (www.cyanosite.
bio.purdue.edu).] (b) Oscillatoria filament. Many cyanobacterial species are filamentous. [Pic-
tures courtesy of Roger Burks (University of California at Riverside), Mark Schneegurt (Wichita
State University), and Cyanosite (www.cyanosite.bio.purdue.edu).]
RHY6 2/6/04 3:55 PM Page 146

146 CIRCADIAN RHYTHMS IN CYANOBACTERIA

through a process called nitrogen fixation. nitrogenase sensitivity to oxygen by devel-


At the heart of the nitrogen fixation process oping specialized cells called heterocysts for
is the enzyme nitrogenase that catalyzes the fixing nitrogen, thereby spatially separating
following reaction: nitrogen fixation from photosynthesis (Fig.
6.3). Vegetative cells participate in photo-
N2 + 8H+ + 8e- Æ 2NH3 + H2 synthesis, produce oxygen, and do not fix
nitrogen. Nitrogen fixation takes place in
the heterocysts that switch off photosyn-
Nitrogen Fixation Versus Photosynthesis
thesis when the nitrogenase enzyme is
Nitrogen fixation and photosynthesis need expressed. Heterocysts supply fixed nitro-
to be separated from each other. The nitro- gen compounds to the vegetative cells and
gen fixing enzyme nitrogenase is extremely survive on sugar supplied by them.
sensitive to oxygen and is inactive in the Spatial segregation of photosynthesis
presence of even small amounts of oxygen, and nitrogen fixation is not possible or
creating a design problem for these photo- practical in unicellular or nonheterocystous
synthetic diazotrophs. In addition, nitroge- species. An alternative strategy, temporal
nase is a rather slow enzyme, reducing only separation, has been utilized by these
three molecules of nitrogen gas per second. cyanobacteria.
Because of its weak activity, nitrogen fixing
cells need to maintain large amounts of the
Temporal Separation
enzyme. As much as 5% of the protein in
these cells may be nitrogenase. Unicellular and nonheterocystous cyan-
Some filamentous nitrogen fixing cyano- obacteria temporally separate photosynthe-
bacteria have overcome the dilemma of sis and nitrogen fixation. In 1985,

Figure 6.3. Filaments of Anabaena and Nostoc showing heterocysts. Heterocysts are spe-
cialized cells in which nitrogen fixation takes place, spatially separating it from photosyn-
thesis. [Pictures courtesy of Roger Burks (University of California at Riverside), Mark
Schneegurt (Wichita State University), and Cyanosite (www.cyanosite.bio.purdue.edu).]
RHY6 2/6/04 3:55 PM Page 147

INTRODUCTION 147

nitrogenase activity in the nonhetero- lacking obvious cellular organization,


cystous, filamentous cyanobacterium Oscil- possess a circadian clock.
latoria sp. was shown to display daily
rhythmicity, with a peak at night. These
Circadian Control of Carbohydrate
rhythms persisted even after the organism
Storage
was transferred to continuous light (LL).
Subsequently, in 1986, Mitsu and co- Carbohydrate storage in Cyanothece occurs
workers described the daily oscillations of in a circadian fashion. Cyanothece sp., a
nitrogenase activity in the unicellular marine, unicellular diazotrophic organism,
marine cyanobacterium, Synechococcus. studied by Sherman and co-workers, tem-
They demonstrated that nitrogen fixation porally separates photosynthesis and nitro-
and photosynthesis were temporally sepa- gen fixation. When this cyanobacterium is
rated. Photosynthesis took place during the grown under conditions of alternating light
day or light period, while nitrogen fixation and dark (LD), photosynthetic oxygen evo-
occurred in the dark. They further showed lution is limited to the light phase and the
that carbohydrate synthesis, respiratory fixation of nitrogen occurs during discrete
oxygen uptake and cell cycle exhibited daily periods in the dark phase. Nitrogen fixation
rhythms in constant light (LL). is an energy dependent process, and as
Robust oscillations of nitrogenase activ- photosynthesis cannot directly supply the
ity in light–dark (LD) conditions were also energy and reducing power required in the
reported for a freshwater species of Syne- dark, this organism relies on stored carbo-
chococcus RF1, isolated from rice fields. hydrate granules to provide the requisite
Rhythms of photosynthesis and nitrogen energy. The carbohydrates, which are stored
fixation were demonstrated to continue in as granules between the photosynthetic
constant light and were entrained by prior membranes, accumulate toward the end of
light–dark cycles. These scientists recog- the light phase, with the number of granules
nized that these daily rhythms were remi- and amount of carbohydrate peaking prior
niscent of eukaryotic circadian rhythms, to the onset of nitrogen fixation.The number
further demonstrated temperature com- of granules and level of carbohydrates is
pensation of the circadian period in greatly reduced at the end of the dark phase.
Synechococcus. Temperature-compensated When Cyanothece is grown under condi-
rhythms of nitrogen fixation and amino tions of constant light, nitrogen fixation
acid uptake have also been reported. In activity peaks during the subjective night
the marine cyanobacterium, Synechococ- and, despite the continuous light, photosyn-
cus WH7803, rhythms of cell division thetic activity is reduced during this phase.
that are temperature-compensated over In addition, the cells still appear to use
the 16–22°C range have been reported. In stored carbohydrates to fuel nitrogen fixa-
another marine cyanobacterium, Syne- tion even though light energy is availa-
chococcus sp. Miami BG43511, rhythms of ble. Carbohydrate conversion to energy
nitrogenase activity, photosynthesis, and requires oxygen in a process called respira-
cell division maintain a 24-hour period over tion. It has been suggested that the use of
the 24–30°C temperature range. carbohydrates, as a fuel source during nitro-
In cyanobacteria circadian rhythms, we gen fixation, lowers the oxygen concentra-
see the three criteria for circadian clock tion within the cell, thereby protecting the
function satisfied: (1) oscillation of rhythms nitrogenase enzyme from inactivation (see
under constant conditions, (2) phase reset- Fig. 6.4 for a model of circadian pattern).
ting, and (3) temperature compensation. Many species of cyanobacteria do not
This indicates that cyanobacteria, although grow without light. Cyanothece can how-
RHY6 2/6/04 3:55 PM Page 148

148 CIRCADIAN RHYTHMS IN CYANOBACTERIA

ZT12 at homologous sites, can receive DNA by


Lights off conjugation from Escherichia coli, can
Carbon express reporter genes, and has a small
and energy
Nitrogen
genome. Studies of rhythms in cyanobacte-
storage ria have relied on the use of a luciferase
fixation
Photosynthesis and reporter that provides a rapid, quantitative
and cell growth respiration assay for circadian function.

ZT0 Bioluminescence Assays for Circadian


Lights on
Regulation
Figure 6.4. Model of circadian pattern in Bioluminescence produced by a luciferase
Cyanothece, showing the temporal separa-
tion of photosynthesis and nitrogen fixation.
reporter is a convenient parameter to
monitor as an assay for circadian function.
Bacterial luciferase catalyzes the oxidation
ever be grown under conditions of continu- of reduced flavin mononucleotide (FMN)
ous darkness (DD) when provided with and a long-chain aldehyde by molecular
glycerol, a source of carbon. Photosynthetic oxygen to yield FMN, the corresponding
activity is reduced during this period. Nitro- acid, water, and light. The aldehyde is not
gen fixation occurs within the subjective essential to the reaction, but it has an acti-
dark phase. Carbohydrate granules are vating effect on the luminescence:
accumulated during the subjective light
phase and dissipated during the period of FMNH2 + n-decanal + O2 Æ
nitrogen fixation. The molecular mecha- FMN + n-decanoic acid + H2O + light
nisms of Cyanothece circadian oscillations
are relatively unknown and a molecular Bacterial colonies expressing the luciferase
clock has yet to be found. gene glow. Researchers exploited this
property of luciferase, together with its
䊏 MOLECULAR BASIS OF THE CLOCK short half-life and real-time reporting,
to monitor circadian regulation of gene
IN CYANOBACTERIA
expression. A promoterless segment of
the Vibrio harveyi luciferase structural
Synechococcus elongatus
gene, luxAB, was introduced downstream
Synechococcus elongatus is a model system of the promoter of the psbA1 gene (Fig.
for molecular genetic dissection of the 6.5a). psbAI is a photosynthetic gene that
cyanobacterial circadian clock. The encodes D1, a major protein component of
research groups of Kondo, Johnson, and the photosystem II reaction center. The
Golden have developed cyanobacteria as a psbA1 mRNA is expressed at high levels
prokaryotic model for molecular analysis of and cycles rhythmically in constant light
circadian rhythms. For their study they following entrainment to a LD cycle. The
chose Synechococcus elongatus (previ- psbAI promoter–luciferase reporter fusion
ously known as Synechococcus sp. strain construct was integrated at a specific target
PCC7942), a nondiazotrophic freshwater site in the chromosome of Synechococcus
strain for which genetic transformation is elongatus to generate the reporter strain
well established. Although this strain does AMC149. The reporter strain exhibited
not fix nitrogen, it has many advantages robust circadian bioluminescence rhythms,
for genetic analyses. It is transformable by under both light–dark and constant-light
linear or circular DNA, which recombines conditions, when n-decanal, the aldehyde
RHY6 2/6/04 3:55 PM Page 149

MOLECULAR BASIS OF THE CLOCK IN CYANOBACTERIA 149

(a)
NS psbA1 luxAB NS

Luciferase
Decanal

Bioluminescence

Luciferase reporter strain

Figure 6.5. The use of a luciferase reporter to measure circadian gene expression in
cyanobacteria. (a) Luciferase reporter construct, showing the luciferase gene, lux AB down-
stream of the psbA1 promoter. (b) Circadian rhythm of bioluminescence from the AMC149
reporter strain in continuous light. The reporter strain was cultured at 30°C under a 12 : 12
L : D cycle. Shown are two traces from cultures previously entrained to L : D cycles 12 hours
out of phase with each other. [Reprinted with permission from Kondo et al. (1993). Copyright
1993 National Academy of Sciences, USA.]

substrate, was provided (Fig. 6.5b). This compensation of the periods. The luciferase
rhythm satisfied the fundamental criteria reporter has successfully been used to find
for circadian rhythms: (1) persistence of circadian gene expression in other cyano-
rhythms in constant light, (2) entrainability bacterial species such as Anabaena and
to light–dark cues, and (3) temperature Synechocystis.
RHY6 2/6/04 3:55 PM Page 150

150 CIRCADIAN RHYTHMS IN CYANOBACTERIA

Gene Expression were distinguished. Class 1 genes were


defined as those whose expression peaks at
In addition to providing an assay for the end of the day, and class 2 genes were
circadian rhythms in cyanobacteria, the those that peak at the end of the night. The
luciferase reporter allowed for automated photosynthetic gene psbAI is a class 1 gene,
detection of genes that showed circadian peaking at dusk (Fig. 6.6). PurF, which
regulation. This was done through a screen encodes glutamine PRPP amidotrans-
of thousands of colonies that represented ferase, a key regulatory enzyme in the de
different insertion sites of the luciferase novo purine synthetic pathway, is a class 2
reporter. The luciferase gene set (luxAB) gene. Transcriptional activity peaks at dawn
was inserted into the Synechococcus elon- and is minimal at dusk (Fig. 6.6).
gatus genome so as to achieve random On the basis of results from the random
insertions of the luxAB gene set through- promoter trap experiments, a model for the
out the chromosome. Of approximately global circadian control of gene expression
30,000 transformed clones, 800 clones was formulated (Fig. 6.7). The model incor-
whose luminescence was bright enough to porates both nonspecific circadian control
be easily monitored were selected. Exami- and circadian regulation by specific cis
nation of the luminescence expression pat- elements and trans factors. As such a large
terns of these 800 glowing clones revealed, number of genes appear to be influenced by
to the researcher’s surprise, that all the the clock in cyanobacteria, it seems unlikely
clones displayed clear circadian rhythmic- that each of them is controlled by a specific
ity. These rhythms exhibited a range of regulatory factor. Clearly, some global
waveforms, amplitudes, and phase relation- factors could be involved as well. Thus,
ships. Two predominant phase relationships according to this model, there would be one

Figure 6.6. Promoter activity of class 1 and class 2 genes as assayed with the luxAB lumi-
nescent reporter. The upper trace is from the class 1 gene psbAI; the bottom trace is from the
class 2 gene purF. Data were recorded in constant light (LL), but the times of expected or sub-
jective night are shown by the gray bars along the abscissa. [Reprinted from Johnson and
Golden (1999) wiht permission from the Annual Review of Microbiology, Volume 53. © 1999
by Annual Reviews, www.annualreviews.org.]
RHY6 2/6/04 3:55 PM Page 151

MOLECULAR BASIS OF THE CLOCK IN CYANOBACTERIA 151

Light
Activity of trans-factor A psbAI

Phototransduction
Activity of trans-factor B purF
Circadian
Photoreceptor
clockwork

Rhythmic activation of
transcription (not gene-specific)
Rhythm of
nonspecific
factor

Figure 6.7. Model for global circadian control of gene expression. Coding regions of clock-
controlled genes are depicted as rectangles with adjacent promoter regions. Some ensembles
of genes may be controlled by rhythmic activity of specific trans factors that coordinately reg-
ulate groups of genes. These trans factors could be activated at different phases. Other genes
may be regulated by global but nonspecific factors that oscillate. [Modified from Johnson et
al. (1996).]

or more types of class 1–specific cis ele- displaying a wide range of circadian pheno-
ments turned on during the day by a class types were isolated. The phenotypes
1–specific trans factor (trans factor A in Fig. included arrhythmia, altered waveforms
6.7). A different set of class 2–specific cis and atypical periods (ranging between 14
elements would be turned on at night by a and 60 hours). Most of these mutants grow
class 2–specific trans factor (trans factor B), as well as wild type and show no obvious
and so on. The model is supported by the phenotype other than circadian abnormali-
following observation. The altered expres- ties. Of these 100 or so mutants about 30
sion of the rpoD2 gene significantly lowers were rescued by the introduction of a wild-
the amplitude of the luminescence rhythm type genomic Synechococcus elongatus
driven by some promoters (e.g., psbAI) DNA library. A gene cluster composed of 3
but not of luminescence rhythms driven novel open reading frames (ORFs) was
by others (e.g., purF). The rpoD2 gene is a identified in the rescuing DNA fragments.
member of the sigma factor gene family This gene cluster was named kai (cycle or
and encodes a sigma70-like transcription rotation in Japanese),with the ORFs termed
factor. kaiA, kaiB, and kaiC (Fig. 6.8).
Nineteen mutations were initially
mapped by genetic rescue and DNA
kai Gene Cluster in
sequencing to the three kai genes (Table
Cyanobacterial Clock
6.1). Fourteen mutations mapped to kaiC,
Using an exhaustive ethylmethanesulfonate three to kaiA, and two to kaiB. All are
(EMS) mutagenesis screen and the luci- missense mutations arising from single-
ferase reporter strain, about 100 mutants nucleotide exchanges.
RHY6 2/6/04 3:55 PM Page 152

152 CIRCADIAN RHYTHMS IN CYANOBACTERIA

kaiA promoter kaiBC promoter

A B C

Transcription

Translation

KaiA KaiB

KaiC

The kai genes A, B, and C produce kaiA, kaiB, and kaiC proteins

Figure 6.8. Schematic representation of the kai gene cluster. The kai gene cluster is tran-
scribed by 2 promoters, PkaiA and PkaiBC. PkaiA produces monocistronic kaiA mRNA, which
gives rise to the 284 aa KaiA protein; PkaiBC generates the dicistronic kaiBC mRNA, which
produces KaiB, a 102 aa protein, and a 519 aa KaiC protein.

䊏TABLE 6.1. Mutations Mapped to the kai Genes


Allele Name Strain Name Period Phenotype Mutation
kaiA1 A33a 33 hours G2280A E103K
kaiA2 A30a 30 hours G2719A R249H
kaiA3 ALAa Arrhythmic G2793A E274K
kaiB1 B21a 21 hours C2948T L11F
kaiB2 B22a 22 hours C3137T R74W
kaiC1 C22a 22 hours C3535T A87V
kaiC2 C29a 29 hours C3745T S157C
kaiC3 C16a 16 hours C3918T R215C
kaiC4 C28a 28 hours C3981T P236S
kaiC5 cLAa Arrhythmic C4017T P248L
kaiC6 CLAd Low amplitude C4018T P248L
kaiC7 C40a 40 hours G4033A R253H
kaiC8 C37a 37 hours G4094A M273I
kaiC9 C21a 21 hours G4237A R321Q
kaiC10 C27a 27 hours A4500G T409A
kaiC11 C44a 44 hours G4536A G421R
kaiC12 C60a 60 hours T4599C Y442H
kaiC13 CLAb Arrhythmic G4654A G460E
kaiC14 CLAc Arrhythmic T495A
RHY6 2/6/04 3:55 PM Page 153

MOLECULAR BASIS OF THE CLOCK IN CYANOBACTERIA 153

kai Gene Deletion or Inactivation


Deletion of the entire kai gene cluster or
inactivation of individual kai genes abol-
ishes circadian rhythmicity. Cyanobacteria
in which the entire kai gene cluster is inac-
tivated grow as well as wild-type cells and
emit bioluminescence, but are completely
arrhythmic (Fig. 6.9). The cluster is thus
essential for circadian rhythmicity but not
for growth. Reintroduction of the kai gene
cluster into another site in the genome of
the deletion mutant restores rhythmicity to
the cells (Fig. 6.9). Thus, the kai gene cluster
contains the entire DNA sequence infor-
mation required for kai function. In addi-
tion, strains carrying an inactivated kaiA,
kaiB, or kaiC are arrhythmic (Fig. 6.9),
indicating that each of the three kai genes
is critical for the circadian rhythm of
Synechococcus.

kai Clock Proteins


The kai gene cluster of Synechococcus is
transcribed by two promoters, PkaiA and
PkaiBC (Fig. 6.8). The kaiA promoter pro-
duces a monocistronic kaiA mRNA while
the kaiBC promoter generates a dicistronic
kaiBC mRNA. Both kaiA and kaiBC tran-
scripts cycle in abundance. Overexpression Figure 6.9. Arrhythmia of bioluminescence
of kaiC represses the pkaiBC promoter caused by inactivation of the kai genes.
acting as a negative-feedback regulator. (a) WT cells; (b) kaiABC-deleted cells
Thus KaiC functions as the key component (DkaiABC) in which the entire kai gene
cluster was deleted by replacement with the
of the circadian oscillatory loop. Overex- W fragment; (c) kaiABC-transposed cells
pression of kaiA enhances PkaiBC tran- (DkaiABC + kaiABC) in which the entire WT
scription, thereby providing the positive, cluster was transposed to NSII in the genome
interlocking component described for other of DkaiABC cells; (d) kaiA-inactivated cells in
systems in previous chapters. Therefore, which the kaiA gene was inactivated by a
nonsense mutation; (e) kaiB-inactivated cells
KaiA functions as the positive element in in which the kaiB gene was inactivated by a
the kaiBC feedback loop, and KaiC acts as nonsense mutation; (f) kaiC-disrupted cells
the negative element to inhibit its own tran- in which the kaiC ORF was replaced with the
scription. KaiB does not as yet have a spe- W fragment. [Reprinted with permission
cific role assigned to it. from Ishiura et al. (1998). Copyright (1998)
American Association for the Advancement
The predicted proteins KaiA, KaiB, and of Science.]
KaiC are composed of 284, 102, and 519
amino acids, respectively. The Kai proteins
have neither DNA binding motifs nor any
RHY6 2/6/04 3:55 PM Page 154

154 CIRCADIAN RHYTHMS IN CYANOBACTERIA

other obvious functional motifs such as the teins. In fact, KaiC was shown to possess an
PAS domain. However, the primary struc- ATP binding activity that is important for
ture of KaiC is interesting in that it has a circadian rhythms. Two DXXG sequences
tandem repeat. The first half of the KaiC that are highly conserved in GTP binding
sequence (1–260) called the CI domain, proteins are also found in the KaiC CI
shares about 42% sequence similarity to domain. Genetic disruption of a nucleotide
the second half, the CII domain (261–519) binding loop in KaiC strongly impairs the
(Fig. 6.10). KaiC also has two ATP or GTP kaiBC expression profile. Substitution of a
binding motifs ( P loops or Walker’s motif), conserved lysine (K52), in P loop 1 by his-
whose consensus is GXXXXGKT/S, and tidine results in complete loss of the kaiBC
two catalytic carboxylate glutamate circadian rhythm. A similar substitution
residues that are found in ATP binding pro- of lysine 294 in P loop 2 results in an

KaiA

MLSQIAICIW VESTAILQDC QRALSADRYQ LQVCESGEML LEYAQTHRDQ 50


IDCLILVAAN PSFRAVVQQL CFEGVVVPAI VVGDRDSEDP DEPAKEQLYH 100
SAELHLGIHQ LEQLPYQVDA ALAEFLRLAP VETMADHIML MGANHDPELS 150
SQQRDLAQRL QERLGYLGVY YKRDPDRFLR NLPAYESQKL HQAMQTSYRE 200
IVLSYFSPNS NLNQSIDNFV NMAFFADVPV TKVVEIHMEL MDEFAKKLRV 250
EGRSEDILLD YRLTLIDVIA HLCEMYRRSI PRET

KaiB

MSPRKTYILK LYVAGNTPNS VRALKTLKNI LEVEFQGVYA LKVIDVLKNP 50


QLAEEDKILA TPTLAKVLPL PVRRIIGDLS DREKVLIGLD LLYGELQDSD 100
DF

KaiC
MTSAEMTSPN NNSEHQAIAK MRTMIEGFDD ISHGGLPIGR STLVSGTSGT 50
GKTLFSIQFL YNGIIEFDEP GVFVTFEETP QDIIKNARSF GWDLAKLVDE 100
GKLFILDASP DPEGQEVVGG FDLSALIERI NYAIQKYRAR RVSIDSVTSV 150
FQQYDASSVV RRELFRLVAR LKQIGATTVM TTERIEEYGP IARYGVEEFV 200
SDNVVILRNV LEGERRRRTL EILKLRGTSH MKGEYPFTIT DHGINIFPLG 250
AMRLTQRSSN VRVSSGVVRL DEMCGGGFFK DSIILATGAT GTGKTLLVSR 300
FVENACANKE RAILFAYEES RAQLLRNAYS WGMDFEEMER QNLLKIVCAY 350
PESAGLEDHL QIIKSEINDF KPARIAIDSL SALARGVSNN AFRQFVIGVT 400
GYAKQEEITG LFTNTSDQFM GAHSITDSHI STITDTIILL QYVEIRGEMS 450
RAINVFKMRG SWHDKAIREF MISDKGPDIK DSFRNFERII SGSPTRITVD 500
EKSELSRIVR GVQEKGPES

Figure 6.10. Amino acid sequences of the Kai proteins. KaiA consists of 284 amino acids
with a molecular mass of 32.6 kD and a calculated isoelectric point (pI) of 4.69. KaiB consists
of 102 amino acids, is 11.4 kD and has a pI of 7.11. KaiC consists of 519 residues, is 58 kD and
has a pI of 5.74. The Walker’s motif A in KaiC is in bold, imperfect motif Bs are underlined,
DXXG motifs are bold and underlined, and CKABD domains are highlighted in gray. Amino acid
abbreviations are as follows: A, Alanine; C, Cysteine; D, Aspartate; E, Glutamate; F, Phenyl-
alanine; G, Glycine; H, Histidine; I, Isoleucine; K, Lysine; M, Methionine; N, Asparagine; p,
Proline; Q, Glutamine; R, Arginine; S, Serine; T, Threonine; V, Valine; W, Tryptophan; and Y,
Tyrosine.
RHY6 2/6/04 3:55 PM Page 155

MOLECULAR BASIS OF THE CLOCK IN CYANOBACTERIA 155

extremely long period phenotype with nance energy transfer (BRET), a technique
lowered amplitude. The K52H substitution that assays protein-protein interactions,
also significantly reduces the ATP binding has also indicated that KaiB polypeptides
activity of KaiC. In addition, replacement interact.
of threonines in both loops by alanine BRET is a naturally occurring phenom-
residues disrupts the rhythm completely. enon resulting from the nonradiative
Like their mRNAs, KaiB and KaiC pro- energy transfer between a bioluminescent
teins display circadian oscillations under donor and a fluorescent acceptor that are in
constant conditions (LL).The KaiA protein close molecular proximity. For instance, in
shows little, if any, circadian variation. the sea pansy Renilla reniformis, the lumi-
nescence that results from the catalysis of
coelenterazine by Renilla luciferase (Rluc)
KaiC: The Central Clock Component
is transferred to Renilla green fluorescent
KaiC is clearly the central component of protein (GFP), which in turn fluoresces
the cyanobacterial clock. Most of the when the two proteins dimerize. The BRET
mutants displaying altered circadian phe- assay has been developed using the Renilla
notypes map to kaiC. The KaiC protein luciferase and enhanced yellow fluorescent
continues to be rhythmically expressed protein (EYFP) tagged to the proteins of
under conditions of constant darkness and interest. Escherichia coli cells expressing
metabolic shutdown. The rhythm of KaiC constructs of kaiB-Rluc and kaiB-
abundance appears to be essential for cir- Eyfp genes, produce a luminescence
cadian timekeeping since induced expres- that results from the interaction of the
sion of KaiC within the physiological range tagged gene products, indicating that KaiB
results in phase resetting. Together with the homodimerizes.
activity of KaiC in repressing the kaiBC KaiC enhances the interaction of KaiA
promoter, these findings indicate that KaiC with KaiB in vitro and in yeast cells, sug-
is the canonical clock protein that nega- gesting the ability of these three Kai pro-
tively regulates itself and whose oscillations teins to form a multimeric complex. Yeast
drive overt rhythms. two hybrid studies and in vitro interaction
assays indicate that KaiC possesses two
KaiA binding domains, CKABD1 and CKABD2.
Interaction among Kai Proteins
The interaction between KaiA and KaiC
A clue to the mechanism of the cyanobac- appears to be crucial for circadian rhyth-
terial clock comes from the finding that the micity as known rhythm disrupting muta-
Kai proteins interact. In vitro binding tions in the respective binding domains
assays, yeast two hybrid assays and a reso- alter the CKABD–KaiA interactions. Rela-
nance energy transfer assay indicate that tively new data provide further evidence
KaiA, KaiB, and KaiC proteins interact of the key interaction between KaiA
both homotypically and heterotypically. and KaiC—biochemical and genetic
The tandem similar domains of KaiC, CI, data strongly suggest that the C-terminal
and CII, interact individually with KaiA, domain of KaiA stimulates KaiC phospho-
KaiB, and KaiC in vitro. A long-period rylation, which appears to be important for
mutation in kaiA results in an altered circadian timing. The N-terminal domain
heterotypic interaction between KaiA and of KaiA is a pseudoreceiver domain and
KaiB (a dramatically enhanced KaiA–KaiB is thought to be a timing input device
interaction in vitro), suggesting that that regulates KaiA stimulation of KaiC
protein–protein contact is important to the autophosphorylation. It has been proposed
clock mechanism. Bioluminescence reso- that KaiA and KaiC function cooperatively
RHY6 2/6/04 3:55 PM Page 156

156 CIRCADIAN RHYTHMS IN CYANOBACTERIA

Figure 6.11. Alignment of 11 new deduced KaiC homologs with the KaiC sequence
of Synechococcus PCC7942 and Synechocystis PCC688. The completely conserved P loop is
underlined.

in both positive- and negative-feedback kaiC Gene Distribution


processes in the cyanobacterial clock by
controlling the state of KaiC phosphoryla- Proteins of the kai system show no
tion in a circadian manner. sequence similarity to the clock proteins of
How the interactions among the Kai eukaryotic model organisms. However, a
proteins contribute to the cyanobacterial homolog to kaiABC is present in the com-
circadian clock is not as clearly under- pleted genome sequences of Synechocystis
stood as in Drosophila, where interactions and other cyanobacteria. In addition, Miao
between CLK, CYC, PER, and TIM are and co-workers demonstrated that kaiC-
central to the circadian clock. Formation of related sequences are widely distributed
the CLK–CYC complex activates tran- among phylogenetically diverse strains of
scription of per and tim. PER and TIM cyanobacteria. A PCR-based survey, using
proteins are synthesized in the cytoplasm, degenerate primers designed to identify
heterodimerize, and move into the nucleus. genes of the kai cluster, was used. Over 40
The PER-TIM complex negatively regu- strains of cyanobacteria, representing the
lates per and tim transcription by inhibit- five different sections of the phylum, were
ing CLK-CYC activity. While the process determined to possess kaiC homologous
may not be identical in cyanobacteria, it sequences. Deduced sequences of putative
appears that physical associations between kaiC genes from the 40 strains showed 64%
the Kai proteins contribute to circadian identity with a 198-residue region of the
rhythmicity. Synechococcus elongatus KaiC protein.
RHY6 2/6/04 3:55 PM Page 157

MOLECULAR BASIS OF THE CLOCK IN CYANOBACTERIA 157

Figure 6.11. (Continued)

Most importantly, the P-loop motif that clearly linked to the phase of the oscillation.
possesses ATP binding activity essential for KaiA overexpression enhances the pkaiBC
circadian oscillatory function is completely promoter activity, suggesting a positive-
conserved (100% identity) in all strains feedback mechanism. This transcription/
examined (Fig. 6.11). That the kaiC gene is translation feedback model for the cyanobac-
conserved over a wide range of cyano- terial clock is schematically similar to those
bacterial strains suggests that the clock proposed for Drosophila, Neurospora, and
system may be universal in this prokaryotic mammals.While this model shares many fea-
organism. tures with the eukaryotic clock models, it has
fewer known players. It is likely that, as in
Drosophila and mammals, there are tran-
Circadian Organization in Synechococcus as
scription factors involved in feedback
Model for Cyanobacterial Clock
regulation.The role and identity of these tran-
A transcription/translation feedback model scription factors are currently not known.
has been proposed for the circadian oscillator
of the cyanobacterium, Synechococcus (Fig.
SasA as a Circadian Amplifier
6.12). The kai genes are rhythmically
expressed and the proteins KaiB and KaiC A histidine kinase protein gene, sasA,
oscillate. Continuous overexpression of KaiC has been identified in a yeast two-hybrid
represses the pkaiBC promoter, implying a screen for KaiC associating proteins. sasA
negative feedback mechanism. Overexpres- encodes a Synechococcus sensory histidine
sion of KaiC for a few hours resets the phase kinase. The sensory histidine kinase and
of the rhythm.The level of KaiC expression is its cognate response regulator constitute
RHY6 2/6/04 3:55 PM Page 158

158 CIRCADIAN RHYTHMS IN CYANOBACTERIA

CpmA
Overt rhythms
RpoD2

+
- A B C
CikA KaiA PkaiA PkaiBC
KaiC
CR-1 KaiB
Light Transcription

Pex

Translation

KaiB KaiA
Kai proteins interact

KaiC

Figure 6.12. A transcription/translation-based molecular feedback model for the cyanobac-


terial circadian clock. KaiC negatively regulates its own (kaiBC) expression to generate a
molecular feedback loop, while KaiA activates kaiBC expression as a positive element to make
the loop oscillate. CikA is a novel phytochrome related histidine kinase that may be a circa-
dian photoreceptor. CR1 and PEX proteins may be associated with input pathways, while
CpmA and RpoD2 are components of the output pathway.

phosphorylation-based signal transduction trolled by “transmitter” domains of one or


proteins that make up a two-component more histidine kinases, which may act to
regulatory system. The simplest two-com- bring about its phosphorylation or dephos-
ponent signal transduction systems have phorylation through their kinase or phos-
two protein components: a “sensor” often phatase activities respectively. The signal
located in the cytoplasmic membrane, that constituted by the phosphorylation of the
monitors some environmental parameter; receiver may be translated several ways
and a cytoplasmic “response regulator” that depending on the particular system and, as
mediates changes in gene expression or mentioned above, may include regulation
enzymatic activity in response to sensor of gene transcription or enzymatic activity.
signals. This system uses the phosphoryla- Transmitters have an autokinase activity
tion state of a “receiver” (in the response that transfers phosphoryl groups from ATP
regulator) to propagate a signal. The phos- to a histidine residue, from which they are
phorylation state of the receiver is con- transferred to the receiver. The amino ter-
RHY6 2/6/04 3:55 PM Page 159

MOLECULAR BASIS OF THE CLOCK IN CYANOBACTERIA 159

minal end of the sensory kinase protein is overexpressed or disrupted. Genetic dis-
thought to be the signal input domain. It is ruption of the sasA gene dramatically
this domain of SasA (residues 1–97) that reduced the magnitude of kaiA and kaiBC
interacts with KaiC and shares sequence expression and shortened the period length
similarity to KaiB (Fig. 6.13). The two of the kaiBC oscillation (Figs. 6.14a–d). His-
polypeptides share 60% similarity, and each tidine 162, a putative autophosphorylation
is able to bind to KaiC. The binding site on site on SasA, appears to be necessary for
KaiC appears to be the tandem repeat kaiBC expression. Replacement of this con-
composed of CI and CII domains. Either CI served residue with a glutamine reduced
or CII domain of KaiC is sufficient for the kaiBC promoter activity and lowered the
interaction with SasA. amplitude of the rhythm, indicating that
normal robust rhythmicity in Synechococ-
cus requires autophosphorylation of histi-
dine 162 on SasA.
Effect of sasA Disruption on
A role for SasA was supported by
Circadian Regulation
studies in which it was constitutively over-
The functional relevance of SasA to the expressed. Continuous overexpression of
cyanobacterial circadian clock was exam- sasA completely suppresses the kaiBC
ined by analyzing cyanobacterial transfor- promoter, eliminating circadian rhythms.
mants in which the sasA gene was However, the effect is not immediate. After

(a)
1 162 387
H

(b)
10 20 30 40 50 60
SasA QALAQPLLLQLFV-DTRPLSQHIVQRVKNILAAVEATVPISLQVINVADQPQLVEYYRL
.. . .:.:.: . : : . .. .::::: :: : ..::.: .:::.: ..
KaiB MSPRKTYILKLYVAGNTPNSVRALKTLKNILAEVEQGVYA-LKVIDVLKNPQLAEEDKI
10 20 30 40 50

70 80 90 100
SasA VVTPALVKIGP-GSRQVLSGIDLTDQLANQLPQWLV
..::.:.:. : :.... .. ... :
KaiB LATPTLAKVLPLPVRRIIGDLSDREKVLIGLDLLYG
60 70 80 90

Figure 6.13. (a) Schematic representation of SasA. Putative histidyl residue for autophos-
phorylation (His 162) and conserved C-terminal kinase domain are indicated by shadowing on
the SasA molecule. (b) Alignment of SasA with KaiB showing sequence similarity. Conserved
residues are shaded. [Reprinted from Iwasaki et al. (2000) with permission from Elsevier
Science.]
RHY6 2/6/04 3:55 PM Page 160

160 CIRCADIAN RHYTHMS IN CYANOBACTERIA

(a)

Bioluminescence (counts-30 s-1-colony-1)

(b)
Bioluminescence (counts-30 s-1-colony-1)

(c)

Figure 6.14. Disruption and overexpression of sasA affects circadian kai gene expression. (a)
Low amplitude bioluminescence rhythms seen in the absence of sasA. Bioluminescence
rhythms of the PkaiA and PkaiBC reporters, in both wild type and sasA lacking backgrounds,
are shown. For clarity, the bioluminescence profile of the PkaiBC reporter in the strain lacking
sasA is also shown in magnified scale (both solid lines in this panel correspond to the profile
in a sasA null. (b) The kaiBC expression profile in the H162Q mutant, in which the putative
autophosphorylation residue in sasA was substituted with Gln. Bioluminescence from wild-
type (. . .) and mutant lines (solid line) was monitored in LL. (c) Continuous overexpression of
sasA disrupts circadian rhythms. Bioluminescence of a PkaiBC reporter in a Ptrc vector or a
Ptrc::sasA (for overexpression of SasA) construct was monitored. Cells were grown on agar
plates in LL, exposed to darkness to synchronize their clocks, returned to LL and treated with
IPTG or water at hour 24 after dark exposure (arrows indicate addition of water or IPTG).
RHY6 2/6/04 3:55 PM Page 161

MOLECULAR BASIS OF THE CLOCK IN CYANOBACTERIA 161

SasA is overexpressed, a robust kaiBC


expression cycle occurs before expression
shuts down (Fig. 6.14e). In contrast, kaiC
overexpression immediately suppresses
kaiBC expression. Transient overexpres-
sion of sasA advances or delays the phase
of kaiBC expression rhythm.
Together these studies indicate that
SasA is not an essential component of the
basic timing loop but acts as an amplifier of
the oscillator to generate robust rhythms.
Since the expression of most genes in Syne-
chococcus is under circadian clock control,
the effect of sasA disruption on the expres-
sion pattern of several other genes was also
examined. Disruption of sasA attenuated
expression of all genes tested (Table 6.2 and
Fig. 6.15). It should be noted that growth in
sasA-disrupted strains is also severely hin-
dered in light–dark cycles (Fig. 6.16),
although there are no obvious morpholog-
ical differences between the wild-type and
sasA mutant strains under these conditions.
This suggests that under natural diurnal
conditions, sasA is essential for normal
growth.
Sensory kinases function in a phospho-
ryl relay system that begins with the
autophosphorylation of the kinase and the
transfer of a phosphoryl group. The relay
terminates by modifying function of a DNA
binding, transcription regulating, response Figure 6.15. Effect of sasA disruption on
regulator, or other output domain on a the expression of other circadian genes. Bio-
luminescence rhythms comparing wild-type
response regulator. A SasA-mediated (open circles) to sasA-disrupted (filled circles)
reporter strains. The promoter driving luxAB
is indicated for each panel. (a) Three reporter
䊏TABLE 6.2. Altered Gene strains with an arrhythmic circadian pheno-
type in a sasA-disrupted background are
Expression Patterns in a sasA shown. Note that the PopcA rhythm has a class
Null Background 2 (180° offset) phase angle. (b) Two addi-
tional reporter strains that maintain de-
Gene/Promoter Expression
tectable rhythmic expression patterns are
kaiA; kaiBC Very low amplitude, short shown. [Reprinted from Iwasaki et al. (2000)
period bioluminescence with permission from Elsevier Science.]
psbAII; cikA Arrhythmic, constitutive
expression
rpoD2; ndhD Period, amplitude or phase
angle of expression
rhythm altered
RHY6 2/6/04 3:55 PM Page 162

162 CIRCADIAN RHYTHMS IN CYANOBACTERIA

(a)

(b)

Figure 6.16. sasA strains show slow growth in LD but not in LL or temperature cycle condi-
tions. (a) Colonies of wild-type, kaiABC-deficient, or sasA-disrupted strains were allowed to
form on solid media for 10 days under continuous illumination (LL) or 12 hours light/12 hours
dark (12L/12D) conditions at the indicated light intensity. (b) Growth phenotypes of wild type
and strains lacking sasA, kaiABC, or sphSR on agar plates grown under LL (50 mE m-2 s-1) and
12L (50 mE m-2 s-1)/12D at 30°C, or LL (50 mE m-2 s-1) with 12-hour 30°C/12-hour 37°C cycles. These
strains carried a PkaiBC::luxAB reporter. [Reprinted from Iwasaki et al. (2000) with permission
from Elsevier Science.]

phosphoryl relay would therefore include a fied for SasA, all the data support the
SasA cognate response regulator, which hypothesis that a two-component signaling
may plausibly function to activate kaiBC system may be involved. The role for this
gene transcription. While such a cognate postulated response regulator in the
response regulator has not yet been identi- cyanobacterial circadian oscillator would
RHY6 2/6/04 3:55 PM Page 163

INPUT/OUTPUT PATHWAYS OF CYANOBACTERIAL CIRCADIAN SYSTEM 163

be similar to that described for the PAS KaiC could presumably alter its light sensi-
domain containing transcription factors, tivity to further regulate SasA’s activity.
CLK/CYC, and CLOCK/BMAL1 in
Drosophila and mammals, respectively, and 䊏 INPUT/OUTPUT PATHWAYS
WC1/WC2 in neurospora. OF CYANOBACTERIAL
CIRCADIAN SYSTEM
Model for Role of SasA in Synechococcus
Circadian System Input
On the basis of their experimental data, The circadian clock is heuristically consid-
Iwasaki and co-workers have proposed a ered to consist of three components: input
model for SasA-mediated modulation of pathways, which relay environmental infor-
kaiBC expression (Fig. 6.17). SasA and mation to a circadian pacemaker; a circa-
KaiC function as both positive and negative dian pacemaker, which generates the daily
elements in feedback control of kai oscillations; and output pathways through
gene expression. During the subjective day, which the pacemaker regulates various
total KaiC concentration is low, but consid- observable rhythms. While circadian organ-
erable amounts of SasA-KaiC complex ization in reality is more of a feedback
exist in the cell. This complex functions as web/network, this model is still useful to
a positive element by stimulating SasA assign roles to molecular and cellular
autophosphorylation and subsequent acti- components.
vation of a cognate response regulator that Circadian clocks are entrained to envi-
directly or indirectly enhances kaiBC ronmental changes in light and tempera-
expression. SasA may act as a negative ture. Until relatively recently, very little
element when it inactivates its cognate information existed on the molecular
response regulator with its phosphoryl- biology of input pathways to the cyanobac-
protein phosphatase activity. This terial clock. In Synechococcus sp. RF1,
would occur when SasA does not bind experiments suggest that a break in photo-
KaiC or when KaiB binds the SasA-KaiC synthetic activity by darkness is responsible
complex. for photic entrainment. A Synechococcus
Additionally, two alternatives for SasA sp. RF1 rhythm mutant, designated CR1, is
function in cyanobacterial circadian organ- not fully entrainable to light–dark cycles but
ization have also been proposed (Fig. 6.17). is to temperature cycles. CR1 may be the
The first proposes that SasA functions as first cyanobacterial photic–input mutant.
the first output from the circadian timing Other candidates for input factors are pex
mechanism, transmitting timing signals to and cikA. Period extender (pex), is a newly
all clock-controlled processes. This would identified gene that is involved in adjusting
encompass feedback on kai expression the period but does not appear necessary
and on light sensitivity of the oscillator for robust oscillations. Disruption of the pex
and metabolic adaptation to natural growth gene in wild-type cells shortens the period
conditions. The second alternative suggests by 1 hour, while overexpression of pex
that SasA may act as a circadian amplifier lengthens the period by 3 hours, with a con-
to modulate a photic input pathway into the comitant decrease in amplitude. Overex-
oscillator. The amplitude of the circadian pression of pex in various cyanobacterial
oscillation in a sasA mutant is more sensi- clock mutants causes arrhythmicity. Pex
tive to light fluence than the wild-type encodes a 148–amino acid protein and is
strain, indicating that SasA senses light thought to function as a modifier of the cir-
directly or indirectly. Binding of SasA to cadian clock in Synechococcus.
RHY6 2/6/04 3:55 PM Page 164

164 CIRCADIAN RHYTHMS IN CYANOBACTERIA

Figure 6.17. Models for SasA function in the circadian system. (a) A model for the function
of the SasA-KaiC complex in kaiBC expression. In the early morning, when total KaiC abun-
dance is low, KaiC may bind SasA, stimulating its autophosphorylation and thereby activat-
ing its cognate response regulator (RR) through phosphoryl transfer from His (H) to Asp (D).
Activated RR is proposed to directly or indirectly activate expression of kaiBC genes from PkaiBC
(left). As the KaiBC concentration rises in the evening, the KaiC-SasA complex may bind addi-
tional KaiC or KaiB to form a larger complex(es), which reduces SasA autophosphorylation.
SasA may then act as a phosphoryl protein phosphatase, inhibiting RR-mediated stimulation
of PkaiBC activity (right). (b,c) SasA may function as the first output of the clock to regulate
downstream clock-controlled processes (b) or as an element of redundant input pathways (c).
In both models, SasA and clock protein KaiC form an outer feedback loop to amplify the
KaiABC-based basic timing process by modulating kai gene expression as depicted in (a). In
model (b), KaiC-SasA complex controls its cognate response regulator and thereby directly
controls downstream gene expression. In model (c), SasA controls the robustness of the cir-
cadian oscillation in a light-dependent manner, although SasA is not essential to entrain the
clock. SasA may sense light signals directly or indirectly, and the binding of SasA to KaiC would
alter SasA’s enzymatic activity. [Reprinted from Iwasaki et al. (2000) with permission from
Elsevier Science.]

cikA ground indicates that the product of this


gene is not necessary for circadian oscilla-
cikA, circadian input kinase, is a clock asso- tor function. Inactivation of cikA shortens
ciated gene that lies on the input pathway the circadian period of gene expression
and provides phase-setting information to rhythms by approximately 2 hours and
the Synechococcus clock. Persistence of changes the phase of a subset of rhythms.
robust rhythms in the cikA genetic back- Wild-type Synechococcus strains main-
RHY6 2/6/04 3:55 PM Page 165

INPUT/OUTPUT PATHWAYS OF CYANOBACTERIAL CIRCADIAN SYSTEM 165

tained in continuous light respond to a dark Output


pulse by changing the phase of subsequent
Currently, two genes, rpoD2 and cpmA,
peaks by 10–12 hours. cikA-inactivated
have been identified as components of the
reporter strains are unable to reset the
output pathway. A mutation in the rpoD2
phase of the clock in response to a similar
gene lowers the amplitude of circadian
5-hour dark pulse, suggesting that cikA
oscillations of psbAI and ndhD gene
functions in an input pathway to the circa-
expression but has no effects on that of
dian oscillator.
many others, including purF. The RpoD2
CikA belongs to the family of bacterio-
gene is a member of the sigma factor family
phytochromes, has a conserved histidine
and encodes a sigma70-like transcription
kinase motif, and has a carboxyl terminus
factor. Disruption of the circadian phase
similar to the receiver domains of response
modifier A (cpmA) gene alters the phase of
regulators. The similarity of the CikA
the expression rhythm of certain genes,
amino-terminal sequence to those of Ara-
including psbAII, and kaiA, but has no ef-
bidopsis and Deinococcus photoreceptors
fect on psbAI, purF, kaiBC, rpoD2, or
raises the possibility that CikA is a pho-
ndhD. CpmA was identified from a trans-
toreceptor (Fig. 6.18).

(a)

N CB H N D/F G RR C

(b)

CikA 184 DLSEILTTTIREIRQLLNADRAIIYQFKPQCLDAGLDQRW—-PLYIPSQSY--ITYEDRRNE-A


PhyE 216 DIGALCDTVVEDVQRLTGYDRVMVYQFHE-------------------DDHGEVVSEIRRSD-L
RcaE 113 QLKEILHTTVTEVQRILQADRVLIYHVLP-------------------DGTGKTISESVLPDY-
Cph1 152 NLRDFYDVIVEEVRRMTGFDRVMLYRFDE-------------------NNHGDVIAEDKRDD-M

CikA 243 LLSVIDPLVQPGLLITTEEWQRFQQGETLLIDSVG-----------------------FYKERL


PhyE 261 EPYLGLHYPATDIPQA—-ARFLFKQNRVRMICDCNATPVKVVQSEELKRP--LCLVNSTLRAPH
RcaE 157 PTLMDLEFPQEVFPQ--EYQQLYAQGRVRAIADVHDP----------------------TAGLA
Cph1 196 EPYLGLHYPESDIPQP--ARRLFIHNPIRVIPDVYGVAVPLTPAVNPSTNRAVDLTESILRSAY

CikA 284 PEQYSFYERVQVRSVCKIPILVQGR------IWGLLVAHQCQQDHRWQPRERDILQHLAEH


PhyE 321 GCHTQYMANMGSVASLALAIVVKGK--DSSKLWGLVVGHHC---SPRYVPFPLRYACEFLMQ
RcaE 197 ECLVEFVDQFHIKAKLIVPIVQNLNANSQNQLWGLLIAHQCDS-VRQWVDFELELMQQLADQ
Cph1 258 HCHLTYLKNMGVGASLTISLIKDGH------LWGLIACHHQ---TPKVIPFELRKACEFFGR

Figure 6.18. (a) Graphic representation of the 754 amino acid CikA protein indicating the
chromophore binding (CB) domain of phytochromes at the amino terminus, the H, N, D/F, and
G boxes of histidine protein kinases and the receiver domain (RR) of response regulators at
the carboxyl end. (b) Comparison of chromophore binding domains of CikA, PhyE from Ara-
bidopsis thaliana, Synechocystis Cph1, and RcaE from Fremyella diplosiphon. Residues con-
served in all sequences are in boldface type, residues conserved in some sequences are
highlighted in gray. Numbers at the beginning of each line indicate position in the respec-
tive protein sequence. [Modified from Schmitz et al. (2000): Science 289, 765–768.]
RHY6 2/6/04 3:55 PM Page 166

166 CIRCADIAN RHYTHMS IN CYANOBACTERIA

poson-generated mutant of Synechococcus glows. Kondo and Golden demonstrated


PCC7942 that maps to a 780-bp (base pair) that cultures of Synechococcus elongatus
open reading frame. CpmA homologs have that divided twice or more per day dis-
been identified in Synechocystis sp. strain played circadian rhythms of biolumines-
PCC6803 and in three species of Archaea. cence and levels of mRNA. Mori and
All these organisms also have a putative Johnson found that cell division was pro-
kaiC homolog. This suggests that kaiC may hibited for several hours in the circadian
also encode a central clock component in cycle. In actively growing continuous cul-
other prokaryotes and that cpmA acts as a tures in light–dark, cells divided in the light
circadian output factor in those organisms but not in the dark. In constant light a cir-
as well. cadian pattern of division continued even
though the doubling time of the culture was
faster than once every 24 hours (Fig. 6.19).
Control of Cell Division by the
Cell division occurred in the late subjective
Circadian Clock
night and the subjective day. Cells slowed or
Until fairly recently, it was widely accepted stopped dividing early in the subjective
that circadian rhythms were exclusively night. What this meant was that although
found in eukaryotes. This was a plausible cells were dividing more than twice every
idea, since it was assumed that an endoge- 24 hours, the timing of division was under
nous timekeeper with a period close to 24 the control of a circadian oscillator. Syne-
hours would not be very useful to organ- chococcus cells are therefore able to keep
isms that divide more rapidly than once track of two temporal circuits independent
very 24 hours, as do many prokaryotes. The of each other.
discovery of the circadian oscillation of The independence of the circadian
nitrogen fixation in 1986 dispelled this idea. timing circuit from the cell division cycle
The subsequent development of molecular has also been demonstrated by overex-
markers allowed researchers to observe the pressing the cell septum protein FtsZ in S.
relationship between the cell division cycle elongatus. When the FtsZ protein is over-
and the circadian cycle in actively dividing expressed, cell division but not cell growth
cultures. It was found that cell division in is suppressed. This results in green filamen-
Synechococcus is actually gated by the cir- tous cells, and not unicells, being formed
cadian clock; thus it is a circadian output. (Fig. 6.19). This inhibition of cell division
Cultures of Synechococcus elongatus does not affect gene expression rhythms
grow with doubling times as rapid as one (Fig. 6.19), implying that the periodicity of
division per 6–10 hours. In order to investi- the circadian oscillations is stable and inde-
gate circadian rhythms in these cells two pendent of the division state.
research groups concurrently examined
gene expression in Synechococcus elongatus
using the AMC149 strain that carries a 䊏 ADAPTIVE SIGNIFICANCE
luciferase reporter. As described earlier, OF CYANOBACTERIAL
AMC149 contains a PpsbAI : luxAB trans-
CIRCADIAN CLOCK
lational fusion integrated into a neutral site
of the wild-type strain. The psbAI gene
Cyanobacteria Fitness
encodes the photosynthetic protein D1.
When the luciferase gene luxAB is correctly Resonating circadian clocks confer en-
oriented to the promoter PpsbAI, luciferase hanced fitness in cyanobacteria. From an
is expressed and the cyanobacterial colony evolutionary perspective, it is believed that
RHY6 2/6/04 3:56 PM Page 167

ADAPTIVE SIGNIFICANCE OF CYANOBACTERIAL CIRCADIAN CLOCK 167

Number of cells

Control
1000

Luminiscence (cps)
DT = 11.8 h 0
FitZ over-express
1000

0
0 24 48 72 96 120
Time in LL (days) Time in LL (hours)

Figure 6.19. Circadian rhythm of cell division in continuous cultures of rapidly growing S.
elongatus cells and luminescence rhythms in dividing versus nondividing cells. (a) Growth
curve of log-phase culture. (b–e) Cell division of growing bacteria is stopped by overexpres-
sion of FtsZ. Morphology of control cells (b), cells in which FtsZ is overexpressed (c). Lumi-
nescence rhythms in control cells (d) and in FtsZ overexpressing cells (e). [Reprinted from Mori
and Johnson (2001) with permission from Elsevier Science.]

circadian clocks improve survival by aiding light–dark cycle. Light–dark cycles that pro-
adaptation to the environment. While it is vided equal amounts of light and dark but
argued that circadian regulation enables a with varying periodicity were tested. In all
more predictive control, the merit of circa- cases examined, the strain whose period
dian control is not obvious, as clock mutants most closely matched the period of the
in several species often survive as well as light–dark cycle outgrew its competitors
wild-type organisms in the laboratory. (Fig. 6.20). It has been suggested that there
The value of circadian programming to are two possible reasons for this observa-
reproductive fitness in cyanobacteria was tion: (1) clocks allow optimal utilization of
tested by using wild-type and mutant limiting resources such as CO2, light and
Synechococcus strains that exhibited differ- nutrients—clearly the strain whose period
ent periods under constant conditions. The matches that of the environment can best
strains tested all grew in pure culture at exploit these resources; and (2) cyanobac-
essentially the same rate under light–dark teria rhythmically secrete inhibitory factors
conditions and under conditions of con- that dampen the growth of other strains.
stant light, so there did not appear to be any While there are no physiologic data to
advantage to having a particular circadian support either possibility, it is clear that the
period. However when the different strains phase relationships between biological
were mixed together and grown in compe- rhythms and environmental cycles are
tition with each other a pattern emerged important and having an endogenous
that depended on the endogenous period of period that approximates that of the exter-
each strain and on the period of the nal cycle confers a selective advantage.
RHY6 2/6/04 3:56 PM Page 168

168 CIRCADIAN RHYTHMS IN CYANOBACTERIA

Log-phase culture
(a) (grown in constant light) (b)
100

Fraction of strain in population


Strain with consonance
between FRP and LD cycle
Strain 1 Strain 2
In constant light
Mix

Grow together Strain with nonoptimal


in competition FRP to the LD cycle
0
Plate aliquots at various timepoints Number of generations
to determine population distributions

(c)

wt

wt
Optimal
growth

C28a C28a

12 hour light 12 hours dark 15 hours light 15 hour dark

Figure 6.20. Competition of circadian mutant strains in different light–dark cycles. (a) Dif-
ferent strains of cyanobacteria were mixed together in batch cultures and grown in compe-
tition under different light–dark cycles. At 8-day intervals, the cultures were diluted with fresh
medium. At various times during the competition, aliquots were plated as single colonies, and
the luminescence rhythms of individual colonies were monitored to determine the frequency
distribution of the different circadian phenotypes. (b) The strain whose endogenous free-
running period most closely matched that of the environmental light–dark cycle was able to
outcompete strains with a nonoptimal period. In constant light (nonselective conditions), all
the strains were able to maintain their initial fraction in the population. (c) Phase of a lumi-
nescence rhythm for wild-type (= wt) and the mutant exhibiting a 30-hour period (= C28a) in
a light–dark cycle of 12 hours light/12 hours dark (left) versus 15 hours light/15 hours dark
(right). [From Johnson (2001) with permission from the Annual Review of Physiology, volume
63. © 2001 by Annual Reviews, www.annualreviews.org.]

Circadian Rhythms in Other Prokaryotes tosynthetic cyanobacteria. This raises the


question of whether a circadian clock may
As discussed earlier, it was previously provide similar value in nonphotosynthetic
believed that prokaryotic organization and prokaryotes. There had been a report that
rapid growth lifestyle were not compatible E. coli might have a circadian oscillator, but
with circadian rhythms. It has since been that report was based on an old study with
established that the circadian clock is an poor temperature control, and it appears
integral part of the cyanobacterial organi- that the daily trends observed were a result
zation. Further, circadian programming of diurnal temperature cycles. Nevertheless,
clearly has an adaptive value for the pho- researchers think that there is a good pos-
RHY6 2/6/04 3:56 PM Page 169

CONCLUSIONS 169

sibility that circadian clocks that enhance clocks. The fact that many chloroplast
adaptation and fitness will be found in encoded genes in Chlamydomonas and in
other eubacteria. higher plants are expressed in a circadian
manner raises the likelihood that kai genes
could have been introduced into ancestral
Evolution of the Circadian Clock
plant cells together with photosynthesis
The physiological properties of circadian genes. However, no homolog of kai has
rhythms are so remarkably similar in a wide been found in plant genomes thus far
range of genera that it was generally sequenced. Finally, a genomic survey asserts
accepted that the circadian oscillatory that kaiC is a member of the bacterial
mechanism was developed once and was RecA/DnaB family. RecA is an ATP-
evolutionarily conserved. Clock models dependent DNA recombinase, and DnaB is
of similar structure have been proposed the replication fork helicase in bacteria. It
for cyanobacteria, fungi, and animals. is hypothesized that the ancestral kaiC was
However, following the cloning of the clock a single-domain RecA-like protein that
genes, it has become apparent that key pro- originated in eubacteria and was laterally
teins, encoded by these genes, do not share transferred to Archaea, where gene dupli-
sequence similarity. The clock systems in cation and fusion occurred. The double-
cyanobacteria, fungi, animals, and probably domain gene was subsequently transferred
plants appear to have developed independ- from the Archaea to cyanobacteria after
ently. Natural selection by the alternating the main eubacterial lineages had been
environment has led varied organisms to established.
evolve similar clock mechanisms like the The discovery of genes encoding pro-
negative feedback of gene expression. In teins similar to eukaryotic photoreceptors
each group of organisms, the feedback loop (cryptochromes and phytochromes) in
might have developed a mechanism for Synechocystis and the existence of CikA,
temperature-compensated 24-hour periods a member of the bacteriophytochrome
and evolved into the circadian clock. family, in Synechococcus further suggests
The cyanobacterial kai genes are inter- an evolutionary link between the cyanobac-
esting from an evolutionary perspective for terial clock and those of higher organisms.
several reasons. First, the kai genes are gen-
erally well conserved among cyanobacter-
ial species and potential homologs have 䊏 CONCLUSIONS
also been identified in Archaea. Genes
similar to kaiB and kaiC have been found Although developed recently (as of 2003),
in Archaea and in at least two other line- the cyanobacteria model for circadian
ages of photosynthetic Eubacteria. The rhythms has already made important con-
amino acid identity between the cyanobac- tributions to our understanding of circadian
terial and Archaeabacterial proteins range biology. While components of this molecu-
between 25.7 and 34.2%. More importantly, lar clock are not conserved in animal
the P loop motif, GXXXXGK(T/S), a systems, the mechanisms certainly are, and
GTP/ATP binding region, is highly con- it is likely that the components will be
served. To date a circadian clock has not similar to plant circadian proteins. In addi-
been identified in Archaea. Next, since tion, cyanobacteria provide a powerful tool
cyanobacteria are believed to be ancestral to study the relationship between the circa-
to chloroplasts there is the distinct possibil- dian cycle and basic cellular processes such
ity that there may be a phylogenetic link as the cell cycle. Many other aspects of the
between cyanobacterial and plant circadian circadian control of cell metabolism are
RHY6 2/6/04 3:56 PM Page 170

170 CIRCADIAN RHYTHMS IN CYANOBACTERIA

likely to emerge from studies of the Kondo, T, Strayer, CA, Kulkarni, RD, Taylor, W,
cyanobacteria clock. Ishiura, M (1993): Circadian rhythms in
prokaryotes: Luciferase as a reporter of cir-
cadian gene expression in cyanobacteria.
Proc Natl Acad Sci USA 90: 5672–5676.
FURTHER READING Lakin-Thomas, PL (2000): Circadian rhythms:
New functions for old clock genes. Trends
Ishiura, M, Kutsuna, S, Aoki, S, Iwasaki, H, Genet 16: 135–142.
Andersson, CR, Tanabe, A, Golden, SS, Liu, Y, Golden, SS, Kondo, T, Ishiura, M,
Johnson, CH, Kondo, T (1998): Expression of Johnson, CH (1995): Bacterial luciferase as a
a gene cluster kaiABC as a circadian feed- reporter of circadian gene expression
back process in cyanobacteria. Science 281: in cyanobacteria. J Bacteriol 177: 2080–2086.
1519–1523. Lorne, J, Scheffer, J, Lee, A, Painter, M, Miao,
Iwasaki, H, Williams, SB, Kitayama, Y, Ishiura, VP (2000): Genes controlling circadian
M, Golden, SS, Kondo, T (2000): A KaiC- rhythms are widely distributed in cyano-
interacting sensory histidine kinase, SasA, bacteria FEMS. Microbiol Lett 189: 129–
necessary to sustain robust circadian oscilla- 133.
tion in cyanobacteria. Cell 101: 223–233. Mori, T, Binder, B, Johnson, H (1996): Circadian
Iwasaki, H, Kondo, T (2000): The current state gating of cell division in cyanobacteria
and problems of circadian clock studies in growing with average doubling times less
cyanobacteria. Plant Cell Physiol 41: than 24 hours. Proc Natl Acad Sci USA 93:
1013–1020. 10183–10188.
Johnson, CH, Golden, SS, Ishiura, M, Kondo, T Mori, T, Johnson, CH (2001): Circadian pro-
(1996): Circadian clocks in prokaryotes. Mol gramming in cyanobacteria. Semin Cell Dev
Microbiol 21: 5–11. Biol 12: 271–278.
Johnson, CH, Golden, SS (1999): Circadian pro- Schmitz, O, Katayama, M, Williams, SB, Kondo,
grams in cyanobacteria, adaptiveness and T, Golden, SS (2000): CikA, a bacteriophy-
mechanism. Annu Rev Micrbiol 53: 389–409. tochrome that resets the cyanobacterial cir-
Johnson, CH (2001): Endogenous timekeepers cadian clock. Science 289: 765–768.
in photosynthetic organisms. Annu Rev Schneegurt, MA, Sherman, DM, Nayar, S,
Physiol 63: 695–728. Sherman, LA, Mitchell, CA (1994): Oscil-
Kondo, T, Mori, T, Lebedeva, NV, Aoki, S, lating behavior of carbohydrate granule
Ishiura, M, Golden, SS (1997): Circadian formation and dinitrogen fixation in the
rhythms in rapidly dividing cyanobacteria. cyanobacterium Cyanothece sp. strain ATCC
Science 275: 224–227. 51142. J Bacteriol 176: 1586–1597.
RHY7 2/6/04 3:57 PM Page 171

7
MOLECULAR ANALYSIS OF
CIRCADIAN RHYTHMS
IN NEUROSPORA
Amita Sehgal

Neurospora crassa, commonly known as the rhythm and to characterize it at many levels
“bread mold,” has been exploited as a including determining its response to light
genetic system to study many different and temperature. Since then, there has been
aspects of biology. Its potential use for the no looking back as researchers have used
study of clocks was first noted in 1953 with first genetics and then molecular tools to
the description of a rhythmic growth uncover the complexities of the Neu-
pattern in a Neurospora strain lacking the rospora circadian system.
amino acid proline (thereby called a proline
auxotroph). A few years later, Pittendrigh,
also known for his contributions to 䊏 OVERT RHYTHMS IN NEUROSPORA
Drosophila circadian biology, reported that
the rhythm was circadian. He also found Production of Asexual Spores (Conidia)
that the period was unchanged at different
temperatures, thereby satisfying the crite- Like other filamentous fungi, Neurospora
rion of temperature compensation spread over the surface of the medium in
expected of a circadian rhythm (see the form of undifferentiated filaments
Chapter 1). Sargent and colleagues went on (mycelia). A developmental switch, that is
to develop assays for the Neurospora under circadian control, results in the pro-

Molecular Biology of Circadian Rhythms. Edited by Amita Sehgal


ISBN 0-471-41824-2 Copyright © 2004 John Wiley & Sons, Inc.

171
RHY7 2/6/04 3:57 PM Page 172

172 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS IN NEUROSPORA

duction of aerial hyphae (mycelia growing sion, provides researchers with new assays
perpendicular to the medium) that elabo- that could either replace or supplement the
rate asexual spores (conidia). The switch race tube.
usually occurs toward the end of the sub-
jective night, and the differentiation
process ends around the middle of the sub- 䊏 MOLECULAR BASIS OF THE
jective day, at which point surface filaments
NEUROSPORA CLOCK
dominate the culture again. This rhythm of
conidiation, as it is called, is easily moni-
The frq Gene as a Clock Component
tored in race tubes, which are basically glass
tubes containing a solid medium on which As in Drosophila, progress in the Neu-
the fungus grows (Fig. 7.1). The position of rospora circadian field has relied largely on
the growth front is marked at regular inter- the use of genetics. The first circadian
vals and a constant linear growth rate is rhythm mutations in Neurospora were
assumed. At 25°C these rhythmic growth found by Jerry Feldman in 1971, about the
patterns occur with a periodicity of ~21.6 same time that Ron Konopka reported
hours. the Drosophila period mutants. Many of the
Through molecular analysis, it is now original mutant alleles mapped to the fre-
becoming clear that aspects of Neurospora quency ( frq) gene, which thus became to
metabolism and function other than coni- Neurospora circadian biology what per was
diation are under circadian control. Thus, a to Drosophila. frq mutants affect circadian
number of output cycling genes have been rhythms in Neurospora in many different
isolated that are not required for conidia- ways, ranging from altered periodicity to
tion, but play an important role in other arrhythmicity under normal, race tube
processes. One such process, that is most assay conditions (Fig. 7.2). Many of them
likely regulated by the clock, is the response also affect temperature compensation.
to stress. The identification of other The frq gene was isolated in Jay
rhythms, including those of gene expres- Dunlap’s laboratory and shown to encode
a protein of unknown function. Although
frq shares a glycine-threonine/glycine-
serine repeat region with per, this is not
indicative of a common origin of these two
genes, which are otherwise very dissimilar.
Side Nor did this repeat provide any clues to the
Point of biochemical function of frq (note that the
One circadian cycle
inoculation presence of this repeat in per was initially
Top thought to indicate a proteoglycanlike
nature of the protein, but that notion was
24 hours of subsequently dispelled). The molecular
growth
lesions underlying mutant frq phenotypes
Figure 7.1. The race tube assay for measur- were identified and found to be largely mis-
ing circadian rhythms in Neurospora. Conidi- sense mutations resulting in single amino
ation, characterized by the production of a acid subsitutions (Fig. 7.3).
cluster of aerial hyphae, can be directly visu- As in the case of per, it was the regula-
alized in the race tube. The endogenous tion of frq that turned out to be the most
rhythm is <24 hours, as is evident from the
pattern in the tube. [Reprinted from Bell- intriguing aspect of this gene. Thus, frq
Pedersen et al. (1996) with permission from RNA and protein levels also cycle with a
the Indian Academy of Sciences.] circadian rhythm, and the protein nega-
RHY7 2/6/04 3:57 PM Page 173

MOLECULAR BASIS OF THE NEUROSPORA CLOCK 173

frq+

frq 2

frq7

frq 9

Figure 7.2. Effects of frq mutants on the conidiation rhythm. frq2 is a short-period mutant
as seen by the advancing of its phase relative to wild type each day. frq7 lengthens period,
as demonstrated by the progressive delay in its phase relative to wild type. frq9 is a null that
lacks a circadian rhythm of conidiation. [Reprinted from Bell-Pedersen et al. (1996) with
permission from the Indian Academy of Sciences.]

Upstream ORFs FRQ open reading frame

1 kbp

TG/SG repeat proline rich


basic serine rich/acidic acidic

ATG#1 ATG#3
frq3 frq 7,8 frq1 frq 9 frq 2,4,6
glu364->lys gly459->asp gly482->ser 1 bp deletion ala895->thr
(24.0.hours) (29.0.hours) (16.5.hours) (loss of function) (19.0.hours)

Figure 7.3. Molecular basis of different frq mutants. Major domains and translation initia-
tion sites in frq are depicted schematically. The locations as well as amino acid changes asso-
ciated with mutations that shorten and lengthen period are indicated. The frameshift
mutation in frq9 truncates the protein. (ORF—open reading frame; TG—threonine glycine;
SG—serine glycine.) [Reprinted with permission from the Annual Review of Genetics, Volume
30, 1996, by Annual Reviews (www.annualreviews.org).]

tively regulates the synthesis of its own agents has allowed dissection of the Neu-
mRNA, resulting in a feedback loop analo- rospora molecular cycle at a level that is dif-
gous to the ones we have read about in the ficult in other organisms. To examine each
preceding chapters. Constitutive expression aspect of the feedback loop independently,
of high levels of frq eliminates the overt a frq transgene driven by a quinic acid
rhythm and acute induction of frq resets the inducible promoter, was introduced into a
clock. Together, these data demonstrate the frq9 (frq-null) background (Fig. 7.4). Treat-
importance of frq cycling to the Neu- ment of this strain with quinic acid resulted
rospora circadian system. in rapid induction of frq RNA followed by
the accumulation of FRQ protein. To
address the time course of feedback, levels
Post-translational Regulation of FRQ
of endogenous frq9 RNA were assayed. It
The ability to manipulate gene expression was found that feedback, as assayed by a
together with the use of pharmacological decrease in frq9 levels, was achieved in 3–6
RHY7 2/6/04 3:57 PM Page 174

174 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS IN NEUROSPORA

Figure 7.4. Time course of feedback inhibition. frq was expressed under control of a quinic
acid–inducible promoter in a frq9 background. The time course of feedback on frq9 RNA was
examined by inducing expression of transgenic frq with quinic acid and monitoring levels of
the endogenous frq9 transcript (dotted line). As indicated, negative feedback is achieved
rapidly and stays in effect for several hours after the quinic acid is removed. The extended
period of feedback correlates with the amount of time the transgenic FRQ protein (solid line)
stays around. [Adapted from Merrow et al. (1997).]

hours and could be effected by as few as 10 A second kinase that phosphorylates


molecules of FRQ per nucleus. Interest- FRQ and was found to copurify with it is
ingly, the peak of FRQ protein normally lags casein kinase II (CKII). Null mutants of this
behind the peak of its mRNA by 4–6 hours, kinase affect general growth and develop-
which corresponds to the time taken to ment of Neurospora, thereby precluding
achieve negative feedback in this experi- analysis of the conidiation rhythm.
ment. Recovery from feedback, again as However, in the absence of this kinase, frq
indicated by levels of frq9 RNA, takes much RNA and protein fail to cycle. Both are
longer. Thus, frq9 RNA levels do not return expressed at high levels, supporting the idea
to normal for 14–18 hours.The FRQ protein that hypophosphorylated FRQ is resistant
stays around for ~14 hours and, although to degradation and, in addition, indicating
levels have decreased by this point, they are that phosphorylation is important for neg-
still sufficient to mediate repression. ative feedback, specifically, for reducing
As described earlier for the Drosophila levels of frq mRNA.
proteins, FRQ is progressively phosphory- FRQ can also be phosphorylated by
lated as it accumulates. Phosphorylation casein kinase I a and b. Recall that in
regulates turnover and is also important for Drosophila and mammals, CKIe plays an
other aspects of FRQ function. Blocking important role in the central feedback loop.
phosphorylation with kinase inhibitors In Neurospora, mutants of CK1 show some
results in decreased FRQ turnover and effects on rhythms, but its function in the
lengthens circadian period. Biochemical clock is not clear yet.
experiments in Y. Liu’s laboratory led to
the identification of a calcium/calmodulin-
wc-1 and wc-2 Genes as Positive
dependent kinase, CaMK1, as being
Regulators of frq
responsible for half of the FRQ kinase
activity. Strains lacking this kinase have The WC-1 and WC-2 proteins were first
very modest effects on phase and period, identified as clock-related components on
but do appear to alter light responses of the the basis of their requirement for the induc-
conidiation rhythm. tion of frq by light (further discussed
RHY7 2/6/04 3:57 PM Page 175

MOLECULAR BASIS OF THE NEUROSPORA CLOCK 175

below). Work done in the Macino labora- found in molecules implicated in photore-
tory, where the two wc genes were cloned, ception, most notably in the plant photore-
showed that these proteins mediate all ceptor NPH1.
photic responses in Neurospora, in particu- FRQ dimers interact directly with WC-2
lar responses to blue light. The Loros and and WC-1 and, in doing so, inhibit the tran-
Dunlap laboratories subsequently demon- scriptional activity of the WC complex
strated that not only are both genes (WCC). Analysis of these interactions in
required for the frq light response, they also the CKII mutant background have shed
operate in constant darkness as integral some light on the role of phosphorylation
components of the circadian feedback loop. in this process. It turns out that the defect
The two proteins, WC-1 and WC-2, in negative feedback in this mutant is not
are PAS-domain-containing transcription due to reduced association of FRQ with the
factors that act together as a complex WCC. Increased amounts of the WC pro-
to activate expression of frq. Thus, they teins coimmunoprecipitate with FRQ in
are the Neurospora equivalents of the CKII mutant Neurospora, suggesting that it
CLK/BMAL1 complex found in is the hypophosphorylated form of FRQ
Drosophila and mammals and they func- that preferentially associates with the
tion similarly to activate expression of the WCC. Thus, it appears that association with
autoregulating, so-called “negative” com- the WCC and repression of its activity are
ponent. In Drosophila, these negative com- two separable functions of FRQ—the
ponents are encoded by the per and tim former involves predominantly hypophos-
genes, in mammals they are PER and CRY phorylated FRQ, while the latter requires
and in Neurospora it is frq. In addition phosphorylation by CKII. Degradation of
to PAS domains, each of the two WC pro- FRQ in the latter part of the night allows
teins contains a transcriptional activation WCC-mediated transcription of frq mRNA
domain and a zinc finger DNA binding to resume.
domain (Fig. 7.5). In this respect, WC-1 and As one might predict, loss-of-function
WC-2 differ from their animal counter- mutations in any of these three genes have
parts, which fall in the category of basic dramatic effects on overt rhythmicity.While
helix–loop helix (bHLH) transcription the null mutations lead to loss of rhythmic-
factors rather than zinc finger factors. In ity under standard conditions, some alleles
addition, WC-1 contains an LOV (for of frq and wc-2 alter circadian period.
“light,” “oxygen,” and “voltage”) domain, Period altering mutations have not yet been
which is similar to a PAS domain and is reported for wc-1.

AD LOV PAS PAS NT Zn AD


WC1

WC2

Figure 7.5. Schematic representation of the WC-1 and WC-2 proteins (AD—acidic domain;
NT—nuclear transport signal; Zn—zinc finger). WC-1 contains two PAS repeats and a LOV
domain, while WC-2 has only one PAS domain. [Adapted from Ballario et al. (1998): Roles in
dimerization and blue light photoresponse of the PAS and LOV domains of Neurospora crassa
white collar proteins. Mol Microbiol 29: 719–729 and from Lee et al. (2000).]
RHY7 2/6/04 3:57 PM Page 176

176 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS IN NEUROSPORA

Function of WC-1 indicates that in Neurospora, as in the other


systems we have read about in previous
WC-1 expression cycles and is regulated by
chapters, there are interlocked molecular
FRQ. The WC-1 protein shows 48% simi-
loops at the heart of the clock mechanism
larity to the mammalian BMAL1 protein.
(Fig. 7.7).
This degree of homology in proteins from
To determine whether FRQ regulates
two such distant organisms is striking and
WC-1 at the level of synthesis or degrada-
attests to the evolutionary conservation of
tion, Lee et al. grew Neurospora in medium
circadian mechanisms. wc-1 RNA is
containing cycloheximide, which blocks
induced by light, but it does not cycle under
new protein synthesis, in the presence of
freerunning conditions. However, the
light as well as in the dark. They found that,
protein cycles, although with a phase that is
under these conditions, levels of WC-1 were
different from what is observed for FRQ.
independent of FRQ levels. FRQ levels
Thus, levels of WC-1 peak at night while
were substantially higher in the light but
FRQ peaks during the day (Fig. 7.6). The
were degraded at the same rate in light as
phase is consistent with WC-1’s role as a
well as dark. WC-1, on the other hand,
transcriptional activator of frq, given that
showed more rapid turnover in the light as
the expression of frq RNA precedes the
compared to the dark, indicating that its
expression of FRQ by several hours. WC-1
turnover is independently controlled. The
is also regulated by posttranslational mech-
overall levels also did not support a role for
anisms. In response to light, it is phospho-
FRQ in WC-1 turnover. Thus, FRQ controls
rylated and degraded, suggesting a causal
WC-1 synthesis through an unknown post-
relationship between the two processes.
transcriptional mechanism.
In frq-null mutants, levels of WC-1 are
low suggesting that FRQ is a positive regu-
lator of WC-1. In fact, induction of FRQ,
Function of WC-2
using an inducible promoter, results in
upregulation of WC-1 levels without alter- Levels of WC-2 do not cycle. The protein is
ing wc-1 transcript levels. The expression of expressed constitutively in nuclei in abun-
WC-1 under these conditions lags behind dant nonlimiting quantities. The primary
FRQ expression by ~8 hours, consistent role of WC-2 in freerunning clock function
with the relative profiles of these two pro- is apparently to mediate interactions
teins under normal wild-type conditions. between WC-1 and FRQ. FRQ and WC-1
This cross-regulation of FRQ and WC-1 do not interact in the absence of WC-2.

Figure 7.6. Molecular oscillations of frq and WC. WC-1 and frq are the two known oscillat-
ing components of the Neurospora clock. While frq RNA as well as protein cycle, WC-1 cycles
only at the level of the protein with a phase different from that of FRQ. The oscillations are
shown relative to the light–dark cycle (depicted by the bars at the bottom).
RHY7 2/6/04 3:57 PM Page 177

MOLECULAR BASIS OF THE NEUROSPORA CLOCK 177

or short, is observed. Given reports postu-


lating a role for redox activity in clock
entrainment and perhaps also in clock
output, it is easy to see how thioredoxin
reductase might be involved in the circa-
dian system. Other metabolic mutants have
small effects on period that probably do not
result from direct effects on the clock. For
instance, the arg-13 mutation affects an
amino acid carrier in the mitochondrial
membrane. It is likely that the overall effect
on cellular metabolism produces a small
Figure 7.7. The molecular clock in neuro- change in circadian periodicity.
spora. FRQ negatively regulates its transcrip- The most intriguing interactions between
tion by blocking activity of the WC complex metabolic processes and clock function are
in the nucleus. At the same time it positively
regulates expression of the WC-1 protein, seen with lipid-deficient strains cel-1 and
resulting in an interlocked loop. [Adapted chol-1 (Table 7.1).The cel-1 mutant is defec-
from Lee et al. (2000).] tive in fatty acid synthesis and so displays a
reduced growth rate unless exogenous fatty
acids are provided. In the absence of satu-
Thus, nuclear entry of FRQ leads to its rated fatty acids or in the presence of unsat-
WC-2-dependent interaction with WC-1, urated fatty acids, the conidiation rhythm in
thereby inhibiting the activity of the cel-1 strains is poorly temperature compen-
WC complex. Degradation of FRQ relieves sated, such that the periods are longer at
this repression, allowing the WC-1/WC-2 lower temperatures. In addition, the pres-
heterodimer to resume transcription of the ence of unsaturated fatty acids also length-
frq gene. ens the period at ambient temperature
(22°C). Interestingly, the period lengthening
defects are also observed when cel-1
Rhythms in frq-null Revealed by
mutants are assayed in a frq-null back-
Metabolic Defects
ground (Table 7.1). However, the frq;cel-1
Analysis of circadian rhythms in Neurospora rhythms do not entrain to light.
is complicated by aspects of development The chol-1 strain requires choline for its
and physiology that either interfere with cir- growth. In the absence of choline it displays
cadian rhythms or else display rhythms abnormal, swollen morphology, reduced
themselves.The presence of these rhythms in growth rate, and altered circadian rhythms
frq-null strains has also challenged the that range from arrhythmia to extremely
notion that frq is the central oscillator. long periodicity. The addition of choline to
Some of the metabolic mutations that the medium increases the growth rate and
affect circadian periodicity will likely turn also reduces the severity of the circadian
out to affect functioning of clock proteins. phenotype. As in the case of cel-1, long-
For instance, the cys-9 mutation, which period rhythms displayed by chol-1 in the
occurs in the gene encoding thioredoxin absence of the nutritional supplement
reductase, has dramatic effects on period (choline) are also observed in frq-null back-
length. The phenotype is initially a period grounds, but are not entrained by light
that is 5 hours shorter than the wild type, (Table 7.1).
but after a few days in freerunning condi- Given that the lipid mutations affect
tions irregular periodicity, that can be long growth in general, it is difficult to dissociate
RHY7 2/6/04 3:57 PM Page 178

178 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS IN NEUROSPORA

䊏TABLE 7.1. Experiments Using Null Alleles (Either frq9 or frq10) as frq
Mutants
Lipid Supplemented
Mutant Lipid-Deficient Conditions Conditions Light Entrainability
cel-1 Loss of temperature Wild type Yes
compensation; long
periods in the presence
of unsaturated fatty acids
chol-1 Long period Wild type Yes
frq; cel-1 Long period Arrhythmic No
frq; chol-1 Long period, loss of Arrhythmic No
temperature
compensation
Source: Based on data from Lakin-Thomas and Brody (2000).

the effect on circadian rhythms from the rospora. As in the other organisms
general effect on growth. It is possible that described earlier, light effects changes in the
they unmask an underlying low-amplitude levels of a clock component, namely, frq.
rhythm in growth rate that is present in Levels of frq RNA and thereby that of FRQ
all strains. The reduction of growth rate protein are increased by light exposure in
increases the amplitude of the rhythm. In both the early part of the night as well as
support of this idea, choline-starved wild- toward the end of the night.As frq levels are
type strains will phenocopy the chol-1 normally high during the day, what this does
strain to produce a long-period rhythm. is to reset the clock to a “day” timepoint,
Since the background in this case is frq+, with the early night exposure resetting the
this rhythm can be entrained by light. The clock to dusk and the late-night exposure
oscillator activity observed under choline resetting to dawn; thus, the phase is delayed
starvation conditions may correspond to or advanced, respectively (Fig. 7.8). In this
the oscillator revealed by temperature model, Neurospora is reset primarily by a
cycles in frq9 strains. It is important to dawnlike cue (lights on) and not by a dusk-
remember that the growth rhythms, whose like cue. The discovery, in 2003, of an anti-
periods can vary greatly, do not meet the sense transcript encoded by the frq locus has
criteria for “circadian rhythms” (entrain- added to our understanding of the mecha-
ability by light, temperature compensation, nisms involved. The antisense transcript is
etc.), nor are they known to be controlled also induced by light and serves to modulate
by the kind of intracellular loops that lie at resetting of the clock.
the heart of circadian control. The increase in frq (sense mRNA) levels
in response to light occurs at the level of
䊏 RESPONSE OF NEUROSPORA transcription. In fact, through deletion
CLOCK TO LIGHT AND analysis, two light-response elements
(LREs) have been identified in the frq pro-
TEMPERATURE
moter. Each LRE by itself can mediate a
limited light response. Thus, deletion of the
Photic Resetting of the Clock by frq
distal LRE reduces the response by ~50%,
The molecular events that reset the clock in while deletion of the proximal one (i.e., the
response to light were first described in Neu- one closer to the transcription start site)
RHY7 2/6/04 3:57 PM Page 179

RESPONSE OF NEUROSPORA CLOCK TO LIGHT AND TEMPERATURE 179

occurred in both dark and light, the DNA-


protein complex displayed an altered
mobility in the presence of light. More
importantly, the formation of the light
complex occurred at doses and wavelengths
of light required for circadian responses.
This light response was reconstituted with
in vitro–translated proteins to demonstrate
Figure 7.8. Resetting of the clock in that the EMSA complex was composed
response to light. The circadian photorecep-
tor is the product of the wc-1 gene, although only of the WC proteins and that the
VVD, and possibly phytochrome and cryp- shift from the “dark” to the “light” form
tochrome, play a modulatory role. WC-1 required flavin adenine dinucleotide
recruits its partner, WC-2, and activates tran- (FAD). Finally, treatment of WC-1 alone
scription of frq. [Adapted from Froehlich et
with light produced the “light” complex,
al. (2002).] Since WC-1 itself and other clock-
controlled genes (ccgs) are also upregulated indicating that this was the photoreceptor
by light, it is shown to activate transcription responsible for the shift.
of these genes also. Sequence analysis of the two LREs
reveals an imperfect repeat of the consensus
sequence, CGATN-CCGCT. The GATN
reduces light-induced frq induction by sequence is necessary for the binding of WC,
~70%. However, deletion of both com- so the presence of two of these sequences in
pletely eliminates the light response. In each LRE supports the idea that each LRE
addition, the LREs can confer light is bound by each of the two WC proteins.
inducibility onto a reporter gene, establish- Mutants lacking WC-1 activity are circadian-
ing their role in driving light-induced gene blind, indicating that this is the major pho-
expression. toreceptor. The VVD protein (see text
below) also binds a flavin via a LOV domain
and uses light information to modulate the
Mediation of frq Light Response by the
primary action of WC-1. Finally, two phy-
WC-1 Photoreceptor
tochromes and a crypotochrome have also
As mentioned above, WC-1 contains a been found in the Neurospora genome, but
LOV domain, which had been found in the role of these in the circadian light
photoreceptors in other organisms and was response is unclear at present.
known to bind flavin mononucleotide. On
the basis of the sensitivity of Neurospora
The vvd Gene
rhythms to blue light, flavin-based photore-
ceptors had been predicted. Consistent with The vivid (vvd) gene product gates the cir-
this idea, mutations in the LOV domain of cadian response to light. Work done in
WC-1 were shown, by the Macino labora- several species indicates that the circadian
tory, to render Neurospora blind to blue response to light is gated such that it is
light. More recently, the Loros and Dunlap restricted to definitive periods of time. One
laboratories demonstrated, through an of the most striking examples of this is the
elegant series of experiments, that WC-1 is “dead zone” that occurs in the phase
a blue-light photoreceptor that mediates response curve of many organisms. This
circadian responses to light. dead zone, which is usually found in PRCs
The WC proteins were found to bind generated with low intensity light pulses,
the frq LREs in electrophoretic mobility corresponds to a time in the middle of the
shift assays (EMSAs). While this binding day when light exposure does not produce
RHY7 2/6/04 3:57 PM Page 180

180 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS IN NEUROSPORA

a shift in the rhythm. In Neurospora, gating vvd does, however, affect input to the
is also indicated by other phenomena. clock. In wild-type Neurospora induction of
Light-induced processes occur transiently, the frq and vvd RNA is gated such that it is
even under constant light conditions, greater at certain times of day. In the
suggesting a mechanism that serves to limit absence of the VVD protein, the amplitude
these responses. of the gating is reduced, especially for frq
The light response is regulated, at least and particularly in response to low-fluence
in part, by the product of the vvd gene. vvd light. In addition, VVD affects the phase-
encodes a PAS-domain-containing protein response curve. The dead zone is reduced,
that regulates its own expression through the overall light response is greater and the
negative feedback (Fig. 7.9). Since VVD is shift from delays to advances is delayed in
a small protein, the PAS domain actually vvd null mutants. This effect of VVD on
encompasses almost the entire protein. photic input most likely involves interac-
vvd RNA is induced by light in a WCC- tions with the WC proteins.
dependent manner and then downregu-
lated through a process that requires the
Temperature Regulation by FRQ
VVD protein. While this light response
drives rhythmic expression of vvd in a The FRQ protein affects the range of tem-
light–dark cycle, the oscillations dampen by perature over which the clock functions. As
the second day of constant darkness. Nev- described in earlier chapters, temperature
ertheless, vvd is clock-controlled because its interacts with the clock in multiple ways.
expression on the first day of constant While the period is compensated against
darkness is affected by frq mutants. A long effects of changes in ambient temperature,
period frq allele delays the vvd peak while the phase is reset by temperature signals,
the null mutant results in reduced vvd and the very occurrence of rhythmicity is
levels on the very first day of constant dark- restricted to a specific temperature range.
ness. frq expression and cycling, on the frq appears to participate in mediating all
other hand, are unaffected in a vvd null these different effects of temperature.
indicating that vvd is clock-controlled, but Although the mechanisms underlying
does not itself affect the clock. temperature compensation are not known,

Figure 7.9. Regulation of photic input by the vvd gene. vvd is induced by light in a WC-
dependent fashion. The protein product then reduces activity of the WC complex, thereby
regulating the intensity of the light response. [Adapted from Heintzen et al. (2001).]
RHY7 2/6/04 3:57 PM Page 181

RESPONSE OF NEUROSPORA CLOCK TO LIGHT AND TEMPERATURE 181

some of the frq mutants are deficient in this levels and the decrease was somewhat
property, indicating a role for frq in this dependent on the starting point, that is the
process. prior phase. While the frq RNA levels
At higher temperatures levels of FRQ showed an acute response to temperature
are higher at all times of day. Thus, such that they increased with the shift to the
the protein basically oscillates around a higher temperature and decreased at the
higher baseline level. An increase of tem- lower temperature, these effects did not last
perature automatically resets the clock longer than a few hours. Thus, the adjust-
without any change in the levels of frq ment to different temperatures involves
RNA or protein because the FRQ level cor- regulation of FRQ largely at a posttran-
responds to a different time of day at the scriptional level.
new temperature (Fig. 7.10). Since the Interestingly, temperature can dominate
trough level at 28°C is higher than the peak over light in the entrainment of the Neu-
level at 21°C, Liu et al. predicted that a rospora clock: (1) temperature cycles
stepup from 21 to 28°C would always reset produce greater synchrony of the conidia-
the clock to CT0 (the low point of FRQ tion rhythm, and (2) when Neurospora
expression). They did, in fact, find this to be were placed under conditions of competing
the case. Conversely, a stepdown of tem- light and temperature cycles, they entrained
perature reset the phase to CT12, although to temperature, provided the amplitude of
this resetting was not as strong as that pro- the temperature cycle was great enough.
duced by the stepup, most likely because Normally, conidiation occurs during the
the stepdown required a decrease of FRQ dark phase of a light–dark cycle and the
low-temperature phase of a temperature
cycle. When light and temperature cycles
were superimposed such that the low-
temperature phase coincided with the light
phase, conidiation was observed during the
light phase. Thus, at least in Neurospora,
Oscillation of frq at
Levels of frq RNA

light may not be the dominant entraining


a higher temperature
signal at all times in nature (Fig. 7.11).
Finally, regulation of frq defines the
limits of rhythmicity for the Neurospora
oscillator. The use of two different transla-
tion initiation sites, the choice of which is
controlled at a posttranscriptional level,
Oscillation of frq at
a lower temperature
results in two forms of FRQ that are dif-
ferentially regulated at different tempera-
CT0 CT12 tures. At higher temperatures the longer
form is preferentially made while at lower
Figure 7.10. Resetting of the circadian temperatures, the shorter form dominates.
clock by temperature. At higher temperature
FRQ oscillates around a higher level, such Each is required for rhythmicity at its
that even the trough levels at 28°C are respective end of the temperature range.
higher than peak levels at 21°C. It follows While either one, by itself, will rescue
then that a temperature shift up at all times rhythms of a frq-null strain, the restored
of day resets the clock to the trough time- rhythms are restricted to a narrower range
point (CT0) A temperature shift down (not
shown) always resets to the peak timepoint of temperature. Normally, Neurospora
(CT12). [Adapted from Loros and Dunlap clock functions can be observed from 18 to
(2001).] 34°C. Strains carrying only the short form
RHY7 2/6/04 3:57 PM Page 182

182 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS IN NEUROSPORA

Figure 7.11. Temperature dominates over light in entrainment of the conidiation rhythm.
In a light–dark cycle conidiation occurs during the dark phase. In the temperature cycle shown
here it occurs during the low-temperature phase. When both cycles are imposed simultane-
ously such that the light phase coincides with the low-temperature phase, conidiation occurs
in the light/low temperature phase, indicating that temperature is a more potent entraining
stimulus.

are rhythmic at lower temperatures, but tion technology and then by differential
become arrhythmic above 27°C while those screening of libraries made from “morning”
carrying the long form retain rhythmicity at or “evening” RNA. More recently, with the
higher temperatures, but are arrhythmic advent of genomics technology, efforts have
below 20°C (Fig. 7.12). focused on high-throughput screens using
microarrays.
As a result of these methods, several
䊏 OUTPUTS CONTROLLED BY CCGs have been identified, and many of
THE NEUROSPORA CLOCK them turn out to be regulated not only by
the circadian clock but also by light. Clearly,
As discussed in previous chapters, outputs many of these will turn out to be involved
of the clock are molecular, cellular, physio- in the remodeling process required for the
logical, or behavioral rhythms that are production of aerial hyphae and conidia.
manifested only when a functional clock is For instance, the eas gene encodes
present. In most cases the molecular and hydrophobin, a hydrophobic protein that
cellular rhythms drive physiological coats conidia and prevents dehydration
processes to occur at specific times of day. (note that the formation of these conidia
In Neurospora the dominant and best involves exposure of fungal filaments to the
studied overt rhythm is that of conidiation. air after contact with a wet medium). Other
However, it is now apparent that many CCGs encode enzymes, many of which are
other aspects of fungal physiology are involved in metabolism. Levels of glycer-
under circadian control: lipid metabolism, aldehyde-3-phosphate dehydrogenase and
activity of various enzymes, and possibly of a carotenoid biosynthetic enzyme, ger-
also the response to environmental stresses. anylgeranyl pyrophosphate synthase, cycle
The classical approach toward dissecting under constant conditions. The cycling of
output pathways in Neurospora has con- trehalose synthase appears to be required
sisted of molecular screens to identify genes for the conidiation rhythm, but may also
that cycle under control of the clock. The play a role in conferring circadian regula-
term clock-controlled-gene (CCG) was, tion to the stress response.
in fact, coined by Neurospora researchers,
Loros and Dunlap in 1989, and the first such
Promoter Elements in CCGs
screens were done in this organism.
Morning- and evening-specific RNAs were Careful dissection of promoter regions of
isolated first through subtractive hybridiza- frq as well as of some of the CCGs, in par-
RHY7 2/6/04 3:57 PM Page 183

CONCLUSIONS 183

AUG1
AUG3
AUG2

Time in DD (hours)

Figure 7.12. Different forms of FRQ mediate rhythmicity at different temperatures. The dif-
ferent forms of FRQ are produced through the use of alternate initiation codons that are
~100 amino acids apart (AUG1 or AUG3). The choice of the codon is made at a posttran-
scriptional level. While both can restore rhythmicity in frq-null mutants at 25°C, only the short
form (YL34) restores rhythms at low temperatures and only the long form (JCC101) restores
rhythms at higher temperatures. [Reprinted from Liu et al. (1997), copyright 1997, with
permission from Elsevier Science.]

ticular eas, have defined elements required can also confer circadian regulation on
for clock control and distinguished these heterologous genes.
from others required for developmental or
light control. As discussed above, in case of
frq, two regions (LREs) are required for 䊏 CONCLUSIONS
the light response. Both regions are bound
by the WCC in EMSA assays, but only one, Research in Neurospora has clearly been
the distal LRE, is required to confer clock invaluable in testing classical hypotheses
control. In addition, a 68-bp region in the about circadian rhythms, identifying molec-
eas gene is required for clock control and ular mechanisms of clock function, and
RHY7 2/6/04 3:57 PM Page 184

184 MOLECULAR ANALYSIS OF CIRCADIAN RHYTHMS IN NEUROSPORA

unraveling the importance of circadian reg- The PAS protein VIVID defines a clock-
ulation in basic cellular physiology. The dis- associated feedback loop that represses light
covery of frq, second only to that of per in input, modulates gating, and regulates clock
Drosophila, was a major landmark in the resetting. Cell 104: 453–464.
Lakin-Thomas, PL, Brody, S (2000): Circadian
circadian field. More recent data that ques-
rhythms in Neurospora crassa: Lipid defi-
tion the all-important role of frq only serve
ciecies restore robust rhythmicity to null
to highlight the use of Neurospora for frequency and white-collar mutants. Proc Natl
probing the generation of rhythmicity at a Acad Sci (USA) 97: 256–261.
deeper level. While the feedback loops Lee, K, Loros, JJ, Dunlap, JC (2000): Intercon-
reported here and in other chapters are an nected feedback loops in the neurospora cir-
integral part of the circadian timekeeping cadian system. Science 289: 107–110.
mechanism, rhythmicity is so ingrained in Liu, Y, Garceau, NY, Loros, JJ, Dunlap, JC
organisms that aspects must persist even (1997): Thermally regulated translational
when these loops fail. control of FRQ mediates aspects of temper-
ature responses in the neurospora circadian
clock. Cell 89: 477–486.
Liu, Y, Merrow, M, Loros, JJ, Dunlap, JC (1998):
FURTHER READING How temperature changes reset a circadian
oscillator. Science 281: 825–829.
Ballario, P, Macini, G (1997): White collar Loros, JJ, Dunlap, JC (2001): Genetic and molec-
proteins: PASsing the light signal in Neu- ular analysis of circadian rhythms in Neu-
rospora crassa. Trends Microbiol 5: 458– rospora. Annu Rev Physiol 63: 757–794.
462. Merrow, MW, Garceau, NY, Dunlap, JC (1997):
Bell-Pedersen, D, Garceau, N, Loros, JJ (1996): Dissection of a circadian oscillation into dis-
Circadian rhythms in fungi. J Genet 75: crete domains. Proc Natl Acad Sci (USA) 94:
387–401. 3877–3882.
Dunlap, JC (1996): Genetics and molecular Pittendrigh, CS, Bruce, VG, Rosenzweig, NS,
analysis of circadian rhythms. Annu Rev Rubin, ML (1959): A biological clock in Neu-
Genet 30: 579–601. rospora. Nature 184: 169–170.
Feldman, JF, Hoyle, M (1971): Isolation of circa- Sargent, ML, Briggs, WR, Woodward, DO
dian clock mutants of Neurospora crassa. (1966): The circadian nature of a rhythm
Genetics 75: 605–613. expressed by an invertase strain of
Froehlich, AC, Liu, Y, Loros, JJ, Dunlap, JC Neurospora crassa. Plant Physiol 41: 1343–
(2002): White Collar-1, a circadian blue light 1349.
photoreceptor, binding to the frequency pro- Yang, Y, Cheng, P, Liu, Y (2002): Regulation of
moter. Science 297: 815–819. the Neurospora circadian clock by Casein
Heintzen, C, Loros, JJ, Dunlap, JC (2001): kinase II. Genes Dev 16: 994–1006.
RHY8 2/6/04 3:56 PM Page 185

8
PHYSIOLOGICAL AND MOLECULAR
CHARACTERISTICS OF PLANT
CIRCADIAN CLOCKS
Jose A. Jarillo, Juan Capel, and Anthony R. Cashmore

䊏 INTRODUCTION Advances in the identification and char-


acterization of components of the plant cir-
The circadian clock represents a fascinating cadian system have been made largely
adaptation to life on Earth. Organisms have through genetic studies in Arabidopsis.
endogenous biological clocks enabling Approximately 20 genes have been shown
them to tell the time of day. Such circadian to affect the function of the Arabidopsis
timing mechanisms were first described in circadian clock, contrasting with the smaller
photosynthetic organisms. These organisms number of genes known to regulate the
depend on sunlight for their energy, so the circadian clock of insects, mammals or
requirement to adapt to daily and seasonal fungi. As in the mammalian circadian
fluctuations in light provides strong selec- clock, several clock-associated genes from
tive pressure for a capacity to determine Arabidopsis have overlapping functions.
time. Whereas many physiological and bio- The complexity of phototransduction
chemical processes in plants have been pathways in plants may contribute to the
found to be regulated in a circadian large number of genes implicated in clock
manner, a detailed mechanism underly- function.
ing the circadian oscillator remains to be In this chapter, we review the most
elucidated. recent progress in the field, highlighting

Molecular Biology of Circadian Rhythms. Edited by Amita Sehgal


ISBN 0-471-41824-2 Copyright © 2004 John Wiley & Sons, Inc.

185
RHY8 2/6/04 3:56 PM Page 186

186 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

work on the central clock mechanism itself Presumably, control of these processes by
and on the photopigments involved in the the clock allows plants to anticipate and
entrainment of the central clock. prepare for changes in the environment that
occur at dusk and dawn. The effect of dis-
rupting the clock on photoperiodism indi-
䊏 BEGINNING OF PLANT cates that the circadian clock provides a
CIRCADIAN BIOLOGY timing mechanism for the measurement of
daylength. This allows plants to follow the
Plants played a fundamental role in the dis- changing of the seasons as the Earth orbits
covery that organisms possess an accurate the Sun, and as such the clock is involved in
timing mechanism allowing them to syn- the regulation of processes sensitive to pho-
chronize their physiology with the daily toperiod like the timing of flowering.
environmental cycle. As early as 1729, the
French astronomer de Mairan observed
that the daily leaf movements of Mimosa 䊏 PLANT PHOTORECEPTORS
pudica persisted for several days after the
plants were placed in constant darkness. As in other organisms, the circadian system
Plant circadian rhythms continued to in plants consists of input pathways that
intrigue scientists from Linnaeus to provide temporal information from the
Darwin, but little progress ensued until two environment to the clock, the central oscil-
centuries later when Erwin Bunning identi- lator mechanism itself, and a set of pathways
fied the first plant clock mutants in bean. through which the temporal information
Bunning proposed that plants relied on provided by the clock is used to generate
their endogenous circadian clock to overt rhythms in several processes (Fig. 8.1).
monitor the duration of day and night. In addition to its role in entraining the
In 1920, Garner and Allard found that a clock, light is a crucial developmental regu-
tobacco mutant, Maryland Mammoth, grew lator throughout the life history of plants,
profusely to ~5 m in height, but failed to from the regulation of seed germination
flower in the prevailing summer conditions. through seedling establishment to the
However, the plants flowered in the green- timing of flowering. Plants have conse-
house during the winter. By covering plants quently evolved an array of photoreceptors
grown during the long days of summer with capable of detecting light over a large range
a light-tight tent late in the afternoon, arti- of fluence rates and wavelengths. Plant pho-
ficial short days were then provided; these toreceptors fall into three main classes: the
short days also caused plants to flower. phytochromes, which absorb primarily in
Garner and Allard coined the term “pho- the red and far-red region of the spectrum,
toperiodism” for this phenomenon, and and the cryptochromes and phototropins,
plant researchers established the involve- both of which absorb in the blue and ultra-
ment of the circadian clock in controlling violet A (UVA) region of the spectrum. The
these timed events. majority of the most recent work on plant
The pervasive influence of the circadian photoreception has been carried out in Ara-
clock in plants is reflected in the variety of bidopsis thaliana.
processes employed as circadian markers
by researchers. Over the years, overt
Mediation of Response to Red/Far-Red
rhythms have been measured in processes
Light by Phytochromes
such as stem elongation, root pressure,
stomatal opening, cell membrane potential, The phytochrome photoreceptors are
chloroplast movements, and CO2 exchange. homodimeric chromoproteins; each subunit
RHY8 2/6/04 3:57 PM Page 187

PLANT PHOTORECEPTORS 187

Input Central Output Overt Clock - controlled


pathways oscillator pathways rhythms processes
Gene
Ca2+ expression
2+
Ca2+ Ca

Cytosolic
CRY calcium levels
P
Protein
PHY phosphorylation

light

Chloroplast
movement Stomatal opening
thermal
energy
Hypocotyl Leaf
length movement

temperature
receptor?

Imbibition
Hormone production
and resposiveness
Flowering (auxin, ethylene)

Figure 8.1. Model of a simple plant circadian system. The system consists of a set of entrain-
ment pathways (inputs), a putative central oscillator, and sets of output pathways. Entrain-
ing stimuli include red and blue light perceived by phytochromes and cryptochromes,
temperature (by a putative temperature receptor), and possibly imbibition. The central
oscillator is illustrated as a loop including positive and negative elements that give a self-
sustaining oscillation with a period close to 24 hours. Multiple output pathways are drawn
as each regulating an overt rhythm with a distinct phase. Plant clocks control different bio-
logical processes, including the expression of several genes, cytosolic Ca2+ concentration, phos-
phorylation of some proteins, chloroplast movement, stomatal opening, hypocotyl
elongation, cotyledon and leaf movement, and hormone production and responsiveness. The
clock is also crucial for synchronizing developmental processes such as flowering time. Indeed,
mutations in almost all the putative clock-associated genes cause altered photoperiodic
control of flowering.

of the dimer is composed of a linear classified in two groups: (1) type I (PHYA)
tetrapyrrole chromophore that is covalently is light-labile and (2) type II (PHYB-
bound to an apoprotein (Fig. 8.2). Phy- PHYE) are light-stable. At the molecular
tochromes can exist as one of two spectrally and cellular levels Phy responses include
distinct forms, a red-light absorbing form development of the chloroplast, inhibition
(Pr), and a far-red-light absorbing form or promotion of cell growth, ion fluxes, and
(Pfr) that are interconvertible by the gene expression responses. The carboxy-
absorption of red (R) light and far-red (FR) terminal moiety of all phytochromes con-
light, respectively. In higher plants, the Phy tains two PAS repeats and a histidine-
apoproteins are encoded by a small gene kinase-related (HKR) domain (Fig. 8.2).
family, with the holoprotein derived from PAS domains are generally involved in
each isoform having both distinct and over- protein–protein interaction and ligand
lapping functions in light perception. Ara- binding and have been found in many
bidopsis contains five phytochromes (A–E) central clock proteins in other systems.
RHY8 2/6/04 3:57 PM Page 188

188 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

absorbance
Pr 0,8
PAS domains 0,6
PfB B
A 0,4
HKR
Ser 7 Ser 598 0,2

P
300 400 500 600 700 800

Red Far -red

Pfr

absorbance
0,8
Ser 7 Ser 598 0,6
P P ATP P 0,4
PS 0,2

Desensitization ATP ADP


300 400 500 600 700 800
ADP
PIF3 PIF4 Wavelength (nm)
Signaling

Figure 8.2. A phytochrome signaling model. Phytochromes exist in two different forms, the
red-light absorbing (Pr) and the far-red-light (Pfr) absorbing forms, which have different
absorption spectra. The red-light-stimulated phytochrome kinase activity may initiate light sig-
naling by phosphorylating different phytochrome substrates (PSs), such as PKS1 and/or by spe-
cific interactions with downstream elements of the phytochrome signaling pathway like PIF3
and PIF4. PHYA and PHYB regulate multiple aspects of seedling deetiolation, including cell
expansion and plastid development via PIF3. PIF4, in contrast, is proposed to act specifically in
the PHYB pathway to regulate a subset of genes involved in cell expansion. The phytochrome
response might also be downregulated by phosphorylation of the serine-rich amino-terminal
region of phytochrome. PfB phytochromobilin, the phytochrome chromophore.

Mediation of Responses to Blue Light versely, CRY2 performs an important role


by Cryptochromes and Phototropins in the regulation of flowering time. Expres-
sion of the C-terminal domain of Ara-
Two cryptochromes are present in Ara- bidopsis CRY in transgenic seedlings
bidopsis: CRY1 and CRY2 (Fig. 8.3). They results in a constitutive light response. This
have been identified in several plant latent activity of the C-terminal domain is
species, including ferns and algae, and apparently repressed in the dark in the
appear to be ubiquitous throughout the intact molecule. Irradiation with blue light
plant kingdom. The N-terminal domain of relieves this repression, presumably
plant cryptochromes shares strong homol- through a redox reaction involving the
ogy with the chromophore binding domain flavin bound to the N-terminal photolyase-
of DNA photolyases. Thus, they bind FAD like domain. A hypothetical model of CRY
and pterin chromophores; however, they signaling is shown in Fig. 8.4.
lack photolyase activity. In contrast to The phototropin family of blue-light
CRY1, CRY2 is rapidly degraded in photoreceptors in Arabidopsis has two
response to high-intensity blue light. The members: PHOT1 and PHOT2 (Fig. 8.3).
major photoreceptor mediating blue-light Mutants deficient in PHOT1 fail to bend and
inhibition of the elongation of hypocotyl grow toward unidirectional low fluence blue
(the stem of the seedling between the light.PHOT2 controls chloroplast relocation
cotyledons and the radical), is CRY1. Con- induced by blue light. Recombinant pho-
RHY8 2/6/04 3:57 PM Page 189

PLANT PHOTORECEPTORS 189

Deaza
flavin

Pterin FAD
E. coli photolyase

Pterin FAD
Arabidopsis cry1

Pterin FAD

Arabidopsis cry2

FMN FMN

Arabidopsis phot1 LOV LOV Kinase

FMN FMN

Arabidopsis phot2 LOV LOV Kinase

Figure 8.3. Protein structures of E. coli type I photolyase, and Arabidopsis cryptochromes
and phototropins.

Darkness Blue
Inactive light
CRY CRY
Flavin F HY5 F
e-
CCT

LAF1
CIP HY5
CCT

26S
COP1 proteasome LAF1
Zn Coil G Inactive CIP
COP1

Figure 8.4. A hypothetical model of cryptochrome signaling. Arabidopsis cryptochromes


contain a conserved N-terminal flavin-binding domain and a distinct C-terminal extension
(CCT). The CCT interacts with the C-terminal WD-40 repeat domain of COP1. In darkness, COP1
is active and promotes the degradation of different interacting proteins via a 26S protea-
some-mediated process. CRY blue-light activation may cause a change in the redox state
and/or the conformation of the CCT. The activated CCT interacts with COP1, abrogating the
COP1 degradation of different substrates (HY5, LAF1, CIP).

totropins noncovalently bind the chro- tion, and blue-light regulation of stoma-
mophore flavin mononucleotide (FMN) and tal opening are severely impaired in a phot1
undergo autophosphorylation in response phot2 double mutant.All three of these phys-
to blue-light irradiation. Hypocotyl pho- iological responses serve to improve the effi-
totropism, light-induced chloroplast reloca- ciency of photosynthesis.
RHY8 2/6/04 3:57 PM Page 190

190 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

䊏 PHYTOCHROME–CRYPTOCHROME known to act. Despite the fact that PHYA


INTERACTION IN A CIRCADIAN and CRY1 act in this range, they fail to
INPUT PATHWAY compensate for loss of each other, suggest-
ing that they are both required to mediate
low-fluence-rate blue light to the clock.
The identification of timing mutants and Indeed, observations indicate that both
the related clock-associated genes was PHYA and CRY1 are also required for low-
facilitated by the development of technolo- fluence-rate red-light input to the clock,
gies that exploited the circadian oscillation implicating CRY1 as a signal transduction
of transcription of specific genes; these pro- component downstream of PHYA in light
moters were used to drive reporter genes input to the clock. The interaction between
whose expression is amenable to auto- PHY and CRY is supported by functional
mated quantitation. The chlorophyll A/B interactions between PHYB and CRY2 and
binding protein 2 (CAB2) gene encodes between PHYD and CRY1. PHYB inter-
a component of the photosynthetic ma- acts directly with CRY2 as observed in
chinery and shows a strong rhythm of tran- coimmunoprecipitation experiments using
scription, peaking during the early morning. transgenic plants overexpressing CRY2.
In an assay that measured the rhythm in Through fluorescence resonance energy
bioluminiscence from the luciferase (luc) transfer microscopy, it was shown that
reporter gene linked to the CAB2 gene pro- PHYB and CRY2 interact in nuclear
moter, both the phytochromes and the speckles that are formed in a light-
cryptochromes were demonstrated to be dependent fashion.
involved in light input to the clock in plants. Arabidopsis cry1cry2 double mutants
However, there was no perturbation of cir- still show robust rhythmicity, indicating that
cadian function in a phot1 mutant, possibly cryptochromes do not form a part of the
indicating that phototropins do not mediate central circadian oscillator in plants as they
input to the circadian clock. In addition, do in mammals. Interestingly, a quadruple
both red and blue light were effective in photoreceptor mutant of cry1, cry2, phyA,
shortening CAB2 expression period length, and phyB still keeps track of time and
further implicating Phy and CRY plant retains circadian rhythmicity. The mutant
photoreceptors. Mutants lacking phyB was nearly blind for developmental
show a deficiency in responses to high responses but perceived light cues for
fluence rates of red light for shortening of entraining the circadian clock.
period length, whereas mutants lacking in All five PHY and two CRY genes show
phyA are deficient in the response to both diurnal variation, with peaks of expression
red and blue light of low fluence rates. at different timepoints during the light
Analysis of mutants deficient in one or period. On transfer to constant conditions
both cryptochrome photoreceptors reveals (light or dark), circadian regulation is main-
a redundancy in the action of CRY1 and tained for all genes, with the exception of
CRY2 in blue light. At intermediate fluence PHYC; nevertheless, mRNA accumulation
rates of blue light, CRY1 and CRY2 redun- follows a circadian rhythm for all seven
dantly act to maintain circadian period genes under constant light. Similarly, both a
length, whereas at higher fluence rates of suppressor of phyA 1 (spa1) and a compo-
blue light that would degrade CRY2, CRY1 nent of the phototropic response, root pho-
acts alone in circadian photoperception. totropic 2 (rpt2), are also clock-regulated.
Significantly, the cry1 mutant is also defi- PHOT1 gene expression is also clock-
cient in responses to low fluence rates of controlled; however, circadian function is
blue light in the range in which PHYA is unaffected in phot1 mutants, suggesting
RHY8 2/6/04 3:57 PM Page 191

OTHER COMPONENTS OF LIGHT SIGNALING IN PLANTS 191

that PHOT1 is solely an output component. A role for redox potential in resetting
Because phytochromes and cryptochromes the clock mechanism has been proposed in
mediate light input to the clock, and are mammals. DNA binding activity of both
themselves regulated by the clock, they are NPAS2芰BMAL1 and CLK芰BMAL1, the
both clock input and output components components utilized by the cellular clock in
creating feedback loops. Clock control of mammals, are exquisitely sensitive to the
these light signaling components may cell’s redox state (see Chapter 4). A similar
account for the circadian gating of photic role for redox potential as an input to the
signals. clock may also exist in plants as it is known
that the cellular redox state changes over
circadian time in plants.
䊏 RHYTHM ENTRAINMENT
IN PLANTS BY OTHER INPUTS
䊏 OTHER COMPONENTS OF LIGHT
Temperature, as well as light, is an effective SIGNALING IN PLANTS
entraining stimulus and has been demon-
strated to entrain rhythms in CO2 assi- Two Arabidopsis mutants that undergo pho-
milation in crassulacean acid metabolism tomorphogenic seedling development in
(CAM) plants. Cyclic temperature treat- darkness, constitutive photomorphogenic 1
ments of dark-grown pea seedlings induce (COP1) and deetiolated 1 (DET1), display
a rise in specific transcript levels of altered circadian rhythms. Both mutants
light-regulated genes related to photomor- have a shortened circadian period of
phogenesis. Rhythms in light harvesting CAB2芰luc in continuous dark and light
complex B (LHCB) and catalase 3 (CAT3) conditions. Since DET1 and COP1 are
transcription are also entrained by temper- thought to mediate signaling from both phy-
atures cycles in Arabidopsis and low tochromes and cryptochromes, this aber-
temperatures reset the phase of LHCB rantly short period may reflect an imbalance
and cold-circadian rhythm-RNA binding 2 between these different signaling pathways.
(CCR2) oscillations. In tomato, a chilling- Alternatively, and because these mutations
sensitive species, cold pulses stop the clock. cause inappropriate expression of several
Several studies indicate that a circadian gene sets, their effects may not be
clock also affects seed germination. A cir- specific to light. COP1 has three different
cadian oscillator is running in etiolated protein–protein interaction motifs, a RING
dark-grown seedlings that have not seen finger, a coiled coil domain, and a seven
the light or have not been exposed to tem- WD-40 motif domain. Several RING finger-
perature cycles. The amplitude of the acute containing proteins have recently been
induction of both LHCB and CAT2 mRNA implicated in the ubiquitination of specific
abundance by light varies according to the substrates, and the subsequent degradation
timing of the onset of illumination, which by the proteasome. COP1 interacts directly
indicates that a circadian clock running in with HY5, a b-ZIP factor that binds to pro-
etiolated seedlings gates the induction of moters and regulates expression of light-
LHCB and CAT2 by light. The timing of regulated genes. COP1 targets HY5 for
hydration of the dry seed may also serve as degradation by the proteasome in the dark.
a entraining stimulus. The acute induction A direct physical interaction between COP1
of CAT2 mRNA varies with time after and SPA1 has also been demonstrated sug-
imbibition, indicating that this event pro- gesting that SPA1 may function to link the
vides a signal capable of resetting the cir- phytochrome A–specific branch of the light
cadian clock. signaling to COP1. Moreover, both cryp-
RHY8 2/6/04 3:57 PM Page 192

192 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

tochromes CRY1 and CRY2 bind to COP1 controlled processes so far examined. The
and apparently repress COP1 activity period of cycling in transcription and
through direct protein–protein interaction. mRNA abundance of two differently
COP1 also binds to the C-terminal domain phased genes (CAB2 and CCR2) are both
of PHYB in yeast two-hybrid assays, shorter than wild type by 2–3 hours in the
although the relevance of this interaction to toc1 background. CAB2 expression peaks
phytochrome signaling remains to be estab- about 4–6 hours after dawn, whereas peak
lished; as in contrast to the cryptochromes, CCR2 mRNA abundance occurs 5–6 hours
the isolated C-terminal domains of the phy- later. This different phasing suggests that
tochromes do not appear to be active in vivo. different signaling pathways lead from the
oscillator to control of each of these two
genes, and that TOC1 acts upstream of the
䊏 COMPONENTS OF THE CENTRAL divergence.
OSCILLATOR IN PLANTS Toc1-1 also shortens the cab2芰luc
rhythm induced by a red-light pulse in eti-
Much is known about how plants perceive olated seedlings and causes shortened cir-
light, providing many tools for the explo- cadian rhythms in the absence of light input
ration of light interactions with the plant to the clock. Moreover, leaf movement and
clock. Components of the plant central stomatal conductance rhythms in toc1-1 are
oscillator are being identified, although proportionately shorter than in the wild
no homologs of clock genes described type. TOC1 also controls photoperiodic
in other organisms have been found in flowering responses through its effects on
arabidopsis. the clock, and the mutation results in early
flowering.
The TOC1 gene encodes a nuclear
Timing of CAB1 (TOC1)
protein containing an atypical response
Toc1 mutation affects different clock- regulator receiver domain and two motifs
controlled processes. Using the cab2芰luc that suggest a role in transcriptional regu-
luminiscence reporter system mentioned lation: a basic motif conserved within the
above, mutants were identified in which the CONSTANS family of transcription factors
peak of free-running cycling bioluminis- (the CCT motif) and an acidic domain.
cence was altered. More than 20 mutants In the motif that is similar to the receiver
were recovered displaying period lengths domain of the response regulators from two
ranging from 21 to 27 hours (wild type = component signal transduction systems
24.5 hours). The best characterized of these (see Chapter 6), the conserved Asp residue
mutants, toc1-1, runs with a period of 21 that undergoes phosphorylation in other
hours in continuous white light. The mor- characterized response regulators is sub-
phology of the mutant is wild type, consis- stituted in TOC1. Consistent with this
tent with the normal phenotypes observed finding, TOC1 and other pseudoreceivers
for period length mutants in other organ- are unable to undergo phosphorylation in
isms. The simplest observation suggests that vitro, and the function of these putative
as in other organisms the clock is not based domains remains unclear. TOC1 is itself
on metabolic processes that are fundamen- circadianly regulated and participates
tal to the maintenance of the organism but in a feedback loop to control its own
rather, arises from interactions between expression. In fact, TOC1 mRNA cycles
processes that are at least in part specific to with a shortened period in the toc1-1
the clockwork itself. mutant. TOC1 mRNA oscillations dam-
The toc1-1 mutation affects all clock- pen rapidly in darkness, whereas for other
RHY8 2/6/04 3:57 PM Page 193

COMPONENTS OF THE CENTRAL OSCILLATOR IN PLANTS 193

genes (CCR2 and CAT3), expression tochromes appears to be sufficient to syn-


rhythms persist in the dark. In transgenic chronize the APRR circadian waves.
TOC1-overexpressing plants, the free-
running robust circadian rhythm of CAB2
Function of LHY and CCA1 in
was dampened and the circadian profiles of
Autoregulatory Loops
potential clock-associated genes CCA1,
LHY, GI, and CCR2 were all markedly Late elongated hypocotyl (LHY) and circa-
altered, implicating TOC1 as a player dian clock-associated 1 (CCA1), two other
within, or close, to the central oscillator. molecular components controlling the
It is of interest that in Synechococcus plant circadian system, possess a Myb-
elongatus, the bacteriophytochrome CikA related DNA-binding domain. LHY and
(circadian input kinase), which provides CCA1 fulfill some of the required criteria
light input to the cyanobacterial clock and for oscillator components. Both genes show
is important for resetting the clock (see robust circadian oscillations of transcript
Chapter 6), also carries an unusual receiver and protein levels in plants kept in contin-
motif that, like the motif in TOC1, lacks the uous light. They also fulfill the criterion that
expected aspartyl residue needed for the component controls its own levels by
phosphoryltransfer in response regulator feedback inhibition of its synthesis. In con-
proteins. Whether this similarity between stant light, overexpression of either CCA1
TOC1 and CikA is indicative of a bio- or LHY results in repression of each of the
chemical function that cyanobacterial and two genes’ endogenous expression. In addi-
plant mechanisms share has yet to be deter- tion, overexpression of CCA1 and LHY
mined. CikA also lacks the conserved Cys stops all overt rhythmicity that has been
residue expected as a bilin ligand for phy- measured, including leaf movement,
tochromes, although the similarity of CikA hypocotyl elongation, and the expression of
to phytochromes may provide a potential genes that peak with different phases.
evolutionary parallel between cyanobacte- However, in a cca1 null mutant, robust
rial and eukaryotic circadian systems. rhythmicity of output gene expression is
maintained, albeit with a shorter period
than in wild-type plants. These findings
The APRR1/TOC1 Family
indicate that although CCA1 is clearly an
Arabidopsis possesses at least five important element of clock function, LHY
members belonging to the APRR family of oscillations might be able to compensate
pseudoresponse regulators. One of the for the loss of CCA1. Indeed, by construct-
members, APRR1, is identical to TOC1. All ing lhy cca1 double mutants, it has been
these APRR proteins are transcribed with demonstrated that LHY and CCA1 are
a circadian rhythm. Furthermore, in a given partially redundant genes required to main-
24-hour period, the APRR mRNAs accu- tain circadian rhythms. For the first 24–48
mulate sequentially after dawn, separated hours in continuous light, the lhy cca1
by 2–3-hour intervals, suggesting that the mutant showed further shortening of the
APRR family of proteins may all be asso- period of rhythmicity of several clock-
ciated in some way with the Arabidopsis controlled genes. However, by 2 days in
circadian clock. Certain light stimuli are constant light these rhythms and those of
crucial determinants to induce the robust leaf movements damped out completely.
circadian waves, and the first light-induced Thus circadian rhythms cannot be sustained
APRR9 appears to be primarily responsible without both LHY and CCA1, and the
for this light response of the circadian results are consistent with them being com-
waves. A red-light pulse perceived by phy- ponents of the central oscillator.
RHY8 2/6/04 3:57 PM Page 194

194 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

But how can the plant show any oscilla- 䊏 OTHER CLOCK-ASSOCIATED GENES
tions in the absence of LHY and CCA1? A
family of several DNA binding proteins, the ZGT Gene
REVEILLE (RVE) proteins, which have
MYB-like domains similar to those of A clock- and light-regulated tobacco gene
LHY and CCA1, has been identified. Their ZGT, links the circadian oscillator to
mRNAs show rhythmic expression, and the LHCB expression. ZGT transcripts have
constitutive expression of RVE1 and RVE2 alternate forms that are differentially
leads to arrhythmic CAB2芰luc expression expressed in different tissues. ZGT is
in continuous light. The homology with expressed rhythmically in light–dark cycles
LHY and CCA1 is particularly striking in and in constant light. Constitutive expres-
the MYB domain, suggesting that they may sion of ZGT sustains the expression of the
bind similar DNA sequences. These clock-controlled light harvesting complex
proteins might partially substitute LHY LHCB1*1 gene in constant darkness, when
and CCA1, but not be able to sustain the it would normally dampen, but does not
rhythms. affect LHCB1*1 expression in constant
A reciprocal regulation between TOC1 light. ZGT expression is induced rapidly by
and LHY/CCA1 within the Arabidopsis cir- light and its overexpression increases the
cadian clock has been described. LHY and sensitivity of the circadian oscillator to brief
CCA1 negatively regulate TOC1, such that light pulses. The ZGT promoter includes a
its expression is strongly reduced in plants G-box motif, found in many light-regulated
that constitutively overexpress either LHY promoters in plants, and evening element
or CCA1. Indeed, both MYB proteins bind motifs that are correlated with circadian
to a region in the TOC1 promoter that con- control of plant genes.
tains a sequence (the evening element) that
is critical for its circadian regulation. The
genetic interaction between TOC1 and
Phytochrome Interacting Factors (PIF)
LHY/CCA1 is reciprocal, as TOC1 appears The phytochrome interacting factor (PIF)
to participate in the positive regulation of proteins bind to and regulate putative clock
LHY and CCA1 expression. Both the LHY components. Light input to the clock via
and CCA1 messages cycle with a shorter phytochrome is likely to be mediated
period length in toc1-1 and toc1-2 alleles.The through an interaction with PIF3. PIF3 is a
interactive regulation between TOC1 and novel basic helixp–loop helix (bHLH)
CCA1/LHY may define a basic framework protein containing a PAS domain, neces-
for the clock mechanism in arabidopsis. sary for normal photoinduced signal trans-
CCA1 also acts as a positive regulator of duction. PHYA-PIF3 and PHYB-PIF3
CAB gene expression, which suggests that interactions are known to occur. A brief
these MYB factors may simultaneously irradiation with red light induces the rapid
regulate genes that are phased to different binding of the activated phytochrome to
times of the day. The specification of circa- PIF3, whereas a pulse of far-red light
dian phase may entail differential binding releases activated phytochrome from the
by different members of this protein family complex. PIF3 binds specifically to a G-box
at distinct circadian phases. Alternatively, DNA-sequence motif present in CCA1,
phase specification may involve differential LHY, and SPA1 promoters and possibly to
phosphorylation of family members at dis- other targets in the clock. Indeed, the
tinct circadian phases or different interact- induction of CCA1 and LHY was reduced
ing partners recruited to the promoters that in transgenic plants expressing PIF3 anti-
modulate CCA1/LHY function. sense RNA. PHYB binds reversibly to
RHY8 2/6/04 3:57 PM Page 195

OTHER CLOCK-ASSOCIATED GENES 195

G-box-bound PIF3 on light-triggered con- oscillator progression past a light-sensitive


version of the photoreceptor to its biologi- phase in the subjective evening.
cally active conformer. ELF3 encodes a novel nuclear protein
HFR1 is an atypical bHLH protein that that may function as a transcriptional regu-
acts in the phytochrome A signal transduc- lator. The ELF3 transcript level is regulated
tion pathway. HFR1 did not bind directly to in a circadian manner, and this cycling
either PHYA or PHYB. However, HFR1 requires ELF3 protein. Constitutive
did bind to PIF3, suggesting heterodimer- expression of LHY does not abolish the cir-
ization, and both the HFR1/PIF3 complex cadian rhythm of ELF3 gene expression.
and PIF3 homodimer bound preferentially Light and the circadian clock regulate
to the Pfr form of both phytochromes. Thus, ELF3 protein accumulation in nuclei. Fur-
HFR1 may function to modulate PHYA thermore, yeast two-hybrid assays and in
signaling via heterodimerization with PIF3. vitro binding experiments demonstrate that
On the other hand, PIF4, another bHLH ELF3 interacts with PHYB. Genetic analy-
protein, appears to interact preferentially ses suggest that ELF3 requires PHYB func-
with PHYB in its Pfr form. PIF4 does not tion in early morphogenesis but not for the
contain a canonical PAS domain; however, regulation of flowering time. In addition,
it has a region similar to the putative ELF3 modulates resetting of the clock.
PAS-like domain of PIF3 located close to Plants overexpressing ELF3 have an
the bHLH region. PIF4 overexpression increased period length in both constant
resulted in red-light hyposensitivity, while blue and red light; furthermore, etiolated
loss-of-function mutations at PIF4 resulted ELF3-overexpressing seedlings exhibit a
in red-light hypersensitivity. PIF4 may decreased acute CAB2 response after a
function as a nuclear negative regulator of red-light pulse, whereas the null mutant is
PHYB signaling, sequestering the active hypersensitive to acute induction. These
form of PHYB in a nonproductive findings suggest that ELF3 negatively
complex, thereby reducing the photorecep- regulates light input to the clock and its
tor’s effective activity elsewhere. outputs. The action of ELF3 is phase-
dependent. Absence of ELF3 activity
causes a significant alteration of the phase
Early Flowering 3 (ELF3)
response curve during subjective night,
ELF3 modulates resetting of the circadian and constitutive overexpresssion of ELF3
clock. Elf3 mutants are aphotoperiodic and results in decreased sensitivity to the reset-
exhibit light-conditional arrhythmia. Elf3-7 ting stimulus. Indeed, the phase of ELF3
mutant retains oscillator function in the function correlates with its peak expression
light but blunts circadian gating of CAB levels in the subjective night. It is possible,
gene activation, indicating that deregulated then, that ELF3 action represents a mech-
phototransduction may mask rhythmicity. anism by which the oscillator modulates
Furthermore, elf3 mutations confer the light resetting.
resetting pattern of SD photoperiodism,
indicating that gating of phototransduction
Plant Genes Affecting Clock Function
may control resetting. Temperature com-
and Flowering Time
pensation can bypass the requirement for
normal ELF3 function for the oscillator GIGANTEA (GI) is a circadian clock-
and partially restore rhythmic CAB expres- controlled gene that plays a role in the pho-
sion. Therefore, ELF3 specifically affects toperiodic control of flowering in Ara-
light input to the oscillator, similar to its bidopsis. GI transcripts levels show
function in gating CAB activation, allowing circadian oscillations in both light and dark
RHY8 2/6/04 3:57 PM Page 196

196 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

conditions, with peak expression in the dence that early PHY signaling events are
evening. Moreover, in plants overexpress- nuclear localized.
ing either CCA1 or LHY, oscillations of GI ZEITLUPE (ZTL)/ADAGIO 1
mRNA are disrupted. Thus, GI expression (ADO1) and FLAVIN-BINDING KELCH
is under control of the clock. Consistent REPEAT F-BOX 1 (FKF1)/ADO3 genes
with this idea, GI mRNA rhythms are also have also been implicated in both circadian
perturbed in elf3 mutants, suggesting that clock function and the regulation of flower-
GI acts downstream of ELF3. The absence ing time in response to daylength. Mutations
of ELF3 causes arrhythmic and upregu- in ZTL or FKF1 delay flowering under long
lated expression of GI plants under con- but not short days and affect clock-
tinuous light and, although to different controlled gene expression. In the ZTL
extents, in long and short photoperiods. mutant, the circadian-clock-controlled
Complicating this scenario, however, is the peaks in gene expression occur approxi-
finding that gi mutants exhibit an altered mately 3 hours later than in the wild type,
period and reduced amplitude of CCA1 and the circadian rhythmicity of cotyledon
and LHY expression, suggesting that GI movement is also altered (Fig. 8.5). The
functions in input to the clock. GI is fkf1 mutant has a weaker effect on circa-
nucleus-localized, consistent with its pro- dian clock regulation, reducing the level
posed role in PHYB signaling, given evi- of expression of clock-controlled genes,

Z0 Z6 Z12 Z18 Z24 Z30 Z36 Z42 Z48 Z54 Z60 Z66 Z72

60
Relative pixel position

50

40
Columbia
ado1
30

20

10

0
Z0 Z12 Z24 Z36 Z48 Z60 Z72
Relative pixel position

Figure 8.5. Cotyledon tip movement in the ado1 mutant. Cotyledon movement was recorded
for wild-type (Columbia) (left part of each panel) and mutant (ado1) (right part of each panel)
seedlings grown under a 12-hour photoperiod and then transferred to constant white light.
The position in pixels of one cotyledon tip per seedling for 72 hours was recorded. The peri-
odicity of cotyledon movement under continuous white light for the ado1 mutant was about
6 hours longer than that for wild type.
RHY8 2/6/04 3:57 PM Page 197

OTHER CLOCK-ASSOCIATED GENES 197

without altering the level or timing of the LOV domains have been implicated in
peak itself. FKF1 mRNA is itself clearly cir- binding flavin chromophores, which sug-
cadian clock regulated, peaking toward the gests that these proteins might act as blue-
end of the day, whereas ZTL mRNA is not. light photoreceptors. Phenotypic
Transgenic plants overexpressing the characterization of the fkf1 and ztl mutants
ZTL gene have elongated hypocotyls and suggest roles for these proteins in the
petioles with elongated cells and exhibit response to light. The second domain, the F
distinct cotyledon movement during the box, is implicated in targeting proteins for
day. Moreover, these plants flower later degradation via the ubiquitin system and
than do wild-type plants when they are the kelch domain may serve as a
grown under long-day, but not under short- protein–protein interaction domain that
day conditions. GFP-ZTL fusion protein is recruits specific proteins for degradation.
observed in nuclei and in the cytosol, Candidates target proteins for these F-box
indicating that ZTL is a nucleocytoplasmic proteins are circadian clock components
protein that influences flowering time in the and CRY1 and PHYB, which physically
long day pathway of arabidopsis. interact with ZTL/ADO1 in both yeast
A circadian role has also been reported two-hybrid and in vitro binding studies.
for LKP2/ADO2, the other member of the FLC (flowering locus C) encodes a
family. Its mRNA expression is not regu- MADS-box protein that functions as an
lated by the circadian clock and is detected autonomous pathway flowering repressor.
in all tissues examined. Overexpression of Loss-of-function mutations confer early
LKP2 results in arrhythmic leaf movement flowering and shorten the circadian period
and CAB2芰luc expression in constant and FLC may account for ANDANTE
light, and arrhythmic CCR2芰luc expresion (AND) QTL (quantitative trait locus), that
in both constant light and constant dark. affects period of circadian leaf movements
These findings suggest that LKP2 may func- in continuous light, based on its map posi-
tion within or close to the circadian clock. tion. Overexpression of the flowering locus
ZTL/ADO1, LKP2/ADO2, and FKF1/ M (FLM) gene, which is closely related to
ADO3 encode closely related proteins that FLC, produces comparable effects of late
have a combination of a LOV domain, flowering to FLC, acting as an inhibitor of
an F box and six kelch repeats (Fig. 8.6). flowering. Expression of the flowering time

LOV/PAS F-box Kelch


ZTL /ADO1
1 609

LKP2 /ADO2
1 612

FKF1/ADO3
1 619

Flavin ? Skp1p? Target proteins?


PAS protein?

Figure 8.6. Domain structure of the ZTL family of proteins. Schematic alignment of ZTL/ADO1
with paralogs LKP2/ADO2 and FKF1/ADO3. The PAS-like LOV domain, the F box and six kelch
repeats are shown, together with their potential interaction targets.
RHY8 2/6/04 3:57 PM Page 198

198 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

gene Agamous-like 20 (AGL20) is antago- The circadian leaf movement of Mimosa


nistically regulated by FLC and CON- pudica L. is regulated through the action of
STANS through a 113-bp promoter leaf opening substances and leaf-closing
sequence. A CarG box (a DNA element substances. The balance of concentration
that binds a MADS box protein) within the between the two leaf movement factors is
AGL20 promoter sequence is bound speci- inverted during the day. A glycoside-type
fically by the FLC protein in vitro. leaf movement factor is hydrolyzed by
Other identified QTLs such as ESP- b-glucosidase, the activity of which is regu-
RESSO, NON TROPPO, and RALENT- lated by the biological clock. Circadian
ANDO may account for new genes that oscillations in cotyledon position in Ara-
regulate the Arabidopsis circadian system in bidopsis arise from variations in elongation
nature, one of which may be the flowering rates of abaxial (dorsal side of the leaf) and
time GIGANTEA, discussed above. adaxial (ventral side of the leaf) cells of
petioles. These cotyledon movements are
easily monitored by video imaging and
An Arabidopsis Homolog of
have been exploited as the basis for the
TIMELESS (TIM)
identification of natural alleles that con-
TIM is an essential gene for circadian func- tribute quantitatively to the circadian
tion in drosophila. Mammalian timeless period length in Arabidopsis.
genes appear to be not the true orthologs There are also rhythms reflecting growth
of dTIM, but are the likely orthologues of rate, for example, cell elongation. Inflores-
a newly described fly gene, timeout. The cence stems of Arabidopsis exhibit a
predicted Arabidopsis TIM protein con- circadian oscillation in elongation rate cor-
tains TIM homology domains and a puta- related with indol-3-acetic acid (IAA)
tive bipartite nuclear localization signal. levels in rosette leaves, although IAA levels
Arabidopsis tim mutants appear unaffected in the inflorescence stem do not oscillate. In
in circadian properties. Chenopodium stems the circadian rhythms
in elongation rate were correlated with a
rhythm in oleic acid content.
䊏 OUTPUT RHYTHMS The circadian clock also controls the
elongation of the arabidopsis hypocotyl.
One advantage of plants as model clock Light signaling leads to a cessation of
systems is the myriad rhythmic outputs of hypocotyl elongation. Mutations that
plant clocks. The clock gates different overt reduce photoperception and signaling yield
rhythms to distinct times of the day. In most an elongated hypocotyl phenotype;
cases the connections between the clock and conversely, overexpression of elements
the output rhythm are not known; however, of the phototransduction pathways reduces
oscillations of molecules/processes relevant hypocotyl elongation relative to the wild
for various outputs have been reported. type. The hypocotyl elongation defect may
also result from a primary dysfunction in
the circadian system with a resulting failure
Diverse Mechanisms Underlying
to impose a daily period of growth arrest.
Growth Rhythms
The growth rhythms include the pulvinar
Stomatal Aperture, Gas Exchange, and
leaf movement systems, in which the cells in
CO2 Assimilation
the extensor and flexor regions of the pul-
vinus swell in antiphase to drive circadian Oscillations of metabolic enzymes con-
oscillations in leaf position. tribute to stomatal aperture, gas exchange,
RHY8 2/6/04 3:57 PM Page 199

OUTPUT RHYTHMS 199

and CO2 assimilation.The stomatal pores of cell signaling and in the circadian regula-
higher plants allow for gaseous exchange in tion of stomatal aperture and gas exchange.
and out of leaves. These pores are situated Ca2+ may play a role in the entrainment of
in the epidermis, where they are sur- the circadian oscillator as well as in the reg-
rounded by a pair of guard cells controlling ulation of clock-controlled gene expres-
their opening. Circadian rhythms in sion. Free cytosolic and chloroplastic Ca2+
stomatal aperture are correlated with a levels, monitored by aequorin luminis-
rearrangement of the cytoskeleton of the cence, oscillate with a circadian rhythm in
guard cells. On the other hand, Arabidopsis tobacco and Arabidopsis. The light-to-dark
exhibits a circadian rhythm in the rate of transitions stimulates a spike in chloroplast
CO2 assimilation and in beans this rhythm stromal Ca2+ levels, although whether this
includes circadian regulation of the Calvin signals the circadian clock is not known.
cycle reactions. It has been demonstrated that nuclear
Flattened stems of cactus can survive calcium does not exhibit circadian rhytmic-
after detachment from the plant for several ity and that calcium rhythms of different
months without water. Their stomata are cell types oscillate with different circadian
closed all the time and the CO2 released by phases. This may represent different under-
respiration is refixed into malate. This lying cellular control mechanisms and have
process, which has been called crassulacean significant implications for understanding
acid metabolism (CAM) idling, allows the how Ca2+ mediates signal transduction in
plant to survive for prolonged periods of plant cells.
time while losing remarkably little water. External applications of either Ca2+ or
Recent observations have provided evi- Ca2+ ionophores phase-shift the leaflet
dence for the tonoplast functioning as the movement rhythm of Robinia. Different
master switch for the circadian regulation circadian oscillators control Ca2+ fluxes and
of this process. However, phospho- Lhcb gene expression. Free calcium is
enolpyruvate carboxylase (PEPC) kinase responsible for driving the rhythm of Lhcb
gene transcription is under circadian expression. These rhythms free-run with
control and hence the regulation of CAM different periods in tobacco seedlings in
may also be regulated through PEPC. constant conditions, providing evidence for
Some studies have demonstrated circa- separate circadian pacemarkers controlling
dian regulation of sucrose phosphate molecular events in plants.
synthase activity in tomato by a protein
phophatase and circadian regulation of
sucrose metabolism.
Oscillation of Enzymes Required for
Hormone Biosynthesis
Production of ethylene is known to vary in
Circadian Rhythm in Levels of Calcium
a circadian fashion in several species,
Ca2+ is a ubiquitous second messenger including barley, wheat, and rye. In
implicated in phytochrome-mediated as sorghum, this rhythmic ethylene produc-
well as UVA-blue and UVB light signal tion is correlated with rhythms in the activ-
transduction pathways. Pulses of blue light ity of 1-aminocyclopropane-1-carboxylic
induce transient spikes in cytosolic but not acid (ACC) oxidase, the enzyme that con-
in chloroplastic Ca2+, and these transients trols the conversion of ACC to ethylene.
are implicated in signalling downstream of A diurnal oscillation observed in gib-
PHOT1 in the phototropic response. berellic acid (GA) levels in sorghum may
Calcium also plays a critical role in guard be circadian. There is also a circadian
RHY8 2/6/04 3:57 PM Page 200

200 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

oscillation in rosette leaf auxin levels in include the Rubisco activase (RCA), and
Arabidopsis. The rhythm in inflorescence in CAM plants, PEPC kinase. The peak
stem elongation in Arabidopsis in the expression of PEPC kinase mRNA occurs
absence of rhythmic IAA levels might at midnight, and correlates with kinase
reflect rhythmic responsiveness to auxin. In activity, reflecting the requirement of the
potato, feedback control and diurnal enzyme at that time. The expression of
regulation of gibberellin 20-oxidase, a key a single NADPH-protochlorophyllide oxi-
regulatory enzyme in the GA-biosynthetic doreductase gene in cucumber, implicated
pathway has been reported. in the light-dependent step in the biosyn-
Regulation by light–dark cycles of thesis of chlorophyll, is also positively reg-
enzymes implicated in abscisic acid (ABA) ulated by diurnal and circadian rhythms.
biosynthesis in tomato [zeaxanthine epo-
xidase (ZEP) and 9-cis-epoxycarotenoid
Circadian Control of Gene Expression
dioxygenase (NCED)], has been shown.
ZEP mRNA oscillates with a phase very Circadian control of gene expression
similar to LHCII mRNA, and the oscilla- extends to virtually all aspects of plant
tions continue in a 48-hour dark period. metabolism. The expression of two serine
Genes that resemble response regulators, hydroxymethyltransferase (SHM) genes,
which are implicated in propagation of phy- encoding mitochondrial components of the
tohormone responses (e.g., ethylene and photorespiratory pathway, is also circadian-
cytokinin), are also regulated by the circa- regulated. This, together with the circadian
dian clock. regulation of nuclear genes encoding both
chloroplastic (RBCS and RCA) and perox-
isomal components of the photorespiratory
Rhythmic Expression of Genes Involved
pathway, suggests an integrated temporal
in Photosynthesis
regulation of this process. On the other
Genes regulated by circadian output path- hand, the transcripts of two closely related
ways have been shown to peak in expres- putative cold-induced glycine-rich RNA
sion at different times of the day. Examples binding genes, CCR1 and CCR2, have a
include genes involved in photosynthesis, high level of expression in the afternoon,
oxidative stress, cold response, and cell wall with a peak expression 8–12 hours after
production. Circadian rhythms in gene dawn in the wild type. Overexpression of
expression were first reported in plants with CCR2 in arabidopsis strongly depresses the
the observation of a circadian oscillation in cycling of the endogenous CCR2 transcript.
mRNA abundance of LHCB/CAB gene However, CCR2 overexpression has no
expression. effect on the period or the levels of other
In most species, the photosynthetic unrelated circadian-regulated transcript
genes CAB and RUBISCO anticipate dawn such as CAB3 or CAT3. CCR2 gene expres-
by rising to a high level of expression in sion lies most likely downstream of the
early morning. There are species-specific effects of TOC1 on the clock, and may
differences, such as in the expression of define a slave oscillator that is still subject
genes encoding the small subunit of to temporal control from the master oscil-
RUBISCO (RBCs). This is not rhythmic lator, but may itself control an undefined
in all species and, where rhythmicity is subset of clock-controlled outputs.
observed, the circadian regulation can be ATGER3 germinlike cell wall promoter
transcriptional or posttranscriptional. confers circadian-regulated transcription
Other genes related to photosynthesis with peak expression at the beginning of
are also expressed rhythmically. These the night. Nonphotosynthetic enzymes
RHY8 2/6/04 3:57 PM Page 201

OUTPUT RHYTHMS 201

under the control of the circadian clock representing 8200 different genes, more
include catalases, which are primarily than 450 genes displayed were found to
involved in eliminating toxic H2O2 from the show circadian changes in steady-
cell. Catalase genes are expressed at differ- state mRNA levels. These included genes
ent times; CAT2 peaks in the morning and involved in photosynthetic light harvesting,
CAT3 peaks in the afternoon, 12 hours out synthesis of protective phenolic com-
of phase with each other. This difference pounds, lipid modification, carbon meta-
probably reflects different roles for each in bolism and partitioning, nitrogen and
plant metabolism. sulfur assimilation, cell elongation and
Circadian and light regulation of nitrate flowering, and genes coding for photore-
reductase has been shown in Arabidopsis. ceptors. Through analysis of the promoter
Gene expression of S-adenosylmethionine regions of the oscillating genes, a novel
decarboxylase (SAMDC), a key enzyme promoter element, the evening element
involved in the biosynthesis of polyamines, (AAAATATC), which confers circadian
from Pharbitis nil and Tritordeum, is under rhythmicity, was identified. The functional
the control of the circadian clock. A circa- role of this conserved motif was confirmed
dian and senescence-enhanced expression by mutational analysis. Mutation of the full-
of a tobacco cysteine protease gene has length evening element in CCR2 promoter
been identified, and a role in the amino acid caused a decrease in rhythmicity, as did
remobilization in senescing leaves has been deletion of this region.
proposed. In Arabidopsis, the expression of In another study, 11% of the Arabidop-
RD19a, that encodes a drought-inducible sis genes showed diurnal expression and
cysteine proteinase, is also circadian-clock- approximately 2% cycled with a circadian
regulated. rhythm. By clustering microarray data from
additional nonrelated experiments, groups
of genes regulated by the circadian clock
Identification of Clock-Controlled Genes
were identified. Genes within a group were
by Differential Screens
found to be coregulated, such as those
Two clock-controlled genes were isolated involved in photosynthesis and those
from Pharbitis nil through a differential required for starch metabolism. In Ara-
screen for mRNAs increased in abundance bidopsis, starch is synthesized in the plastids
in photoperiods that induce flowering. One during the day and broken down during
of these is PnZIP, a novel mesophyll- the night, thus providing plants with
specific gene that is regulated by phy- sugars. These experiments show that am-
tochrome and encodes a protein with a ylase is expressed in the leaves late in
leucine zipper motif. The other one, the day. Another example of genes found
PnC401, is also clock-regulated and its tran- in these studies are genes encoding pro-
script accumulates during flower-inductive teins involved in nitrogen metabolism.
darkness. Fluctuations in levels of PnC401 Nitrate reductase shows strong circadian
mRNA are regulated by phytochrome and regulation, whereas nitrite reductase is
by the circadian clock and are associated light-regulated.
with photoperiodic events that include The selective advantage that plants gain
induction of flowering. by having genes under circadian regulation
Several circadian clock-regulated genes is presumed to be the ability to anticipate
have been identified in Arabidopsis by diurnal changes in the environment, such as
fluorescent differential display, including variations in light, UV radiation, and tem-
CAB, CAB2, carbonic anhydrase 2 (CAH2), perature. The fact that several photosyn-
RBCS, and CCA1. Using gene chip arrays thetic genes, genes involved in the synthesis
RHY8 2/6/04 3:57 PM Page 202

202 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

of phenolic “sunscreens” and in chilling tol- mRNA undergoes circadian oscillations


erance are under circadian control is con- and the waveform depends on the
sistent with this hypothesis. In addition, the daylength; under SD, the CO peak is largely
clock may provide a temporal framework confined to darkness, whereas under induc-
for internal coordination of physiological tive LD, levels of CO are high during most
activities. This could account for the circa- of the light period. In the gi3 mutant, CO
dian rhythmicity of genes involved in oscillates with a lower amplitude, and in the
carbon partitioning and in the energy- lhy mutant, CO is expressed at a reduced
intensive processes of nitrogen and sulfur level with a different phase.The gi late flow-
assimilation. ering phenotype is corrected by overex-
pressing CO in the mutant background.
Conversely, CO oscillates at higher levels in
CONSTANS (CO)
the early flowering elf3 mutant.
CONSTANS mediates the interaction It is possible that the clock exerts its
between the circadian clock and the con- control on photoperiodic response by
trol of flowering. Flowering time in many setting the CO rhythm. CO, in turn,
species is environmentally-regulated by will promote flowering via downstream
photoperiod, light quality, and vernaliza- genes such as FLOWERING LOCUS T
tion. This ability to perceive changes in (FT). It seems likely that at its peak phase,
daylength, photoperiodism, is essential for CO activates FT, presumably through its
organisms to recognize seasonal changes zinc finger DNA binding domain. The pho-
and is associated with the circadian clock toperiod dependent phase of CO cycling
system. In 1935, Erwin Bunning hypothe- is consistent with the view that the CO
sized that the circadian clock was required rhythm is the molecular basis for differen-
for photoperiod sensitivity in flowering and tial light sensitivity. Thus, the perception of
physiological and genetic data from ara- the photoperiod may be mediated by
bidopsis have corroborated his insight. adjustments in the phase of the circadian
Some plants, including Arabidopsis, cycle that arise on reentrainment to a dif-
flower earlier when growth conditions ferent light–dark cycle. The nature of the
mimic the long days of summer. Most of the rhythm underlying the floral response is not
more recently identified circadian mutants known, but candidate molecules are being
exhibit alterations in flowering time, and identified.
some previously identified flowering A CONSTANS ortholog (PnCO) from
mutants also exhibit circadian defects. For the short-day plant Pharbitis nil also plays
example, toc1 and elf3 show defects in a role in flowering. The level of PnCO is
flowering time. Both LHY and CCA1 photoperiodically regulated and shows a
overexpressing mutants are late flowering. circadian pattern of expression. PnCO is
Mutations at the GI locus result in late able to complement the co mutant of Ara-
flowering, whereas null mutations in the bidopsis. The cloning of Hd1, a major QTL
floral repressor FLC confer earliness in controlling heading date (flowing time) in
flowering. rice, showed that it encodes a GATA1-type
The Zn finger transcription factor CON- protein with high similarity to CONSTANS
STANS (CO), is thought to promote in the zinc finger domain and in the
flowering in long-day (LD) photoperiod C-terminal region. Thus, similar genes are
conditions because co mutant plants have involved in controlling the daylength
delayed flowering in LD, but flower at the response of flowering in short-day (SD) rice
same time as wild type under short-day and long-day (LD) Arabidopsis plants.
(SD) conditions. The abundance of CO Unlike CO in Arabidopsis, HD1 appears to
RHY8 2/6/04 3:57 PM Page 203

OUTPUT RHYTHMS 203

promote heading under short days and to are depicted in Figure 8.7 and listed as
inhibit it under long days. This kind of follows:
research on rice may help to elucidate the
difference in genetic control of photope- 1. Light input to the clock via phy-
riod sensitivity between SD and LD plants. tochrome may occur through a
In Arabidopsis, CO target genes play dis- complex with PIF3 and PIF4. PIF3
tinct roles in reproductive development. binds to a G-box motif in CCA1,
Two of them, AGL20 and FT, are required LHY, and SPA1 promoters and pos-
for CO to promote flowering; AGL20 sibly to other targets in the clock.
encodes a MADS-box transcription factor, PIF4 may function as a negative reg-
whose expression responds to long pho- ulator of PHYB signaling. HFR1
toperiods, consistent with the role that seems to modulate PHYA signaling
CO plays in the promotion of flowering. via heterodimerization with PIF3.
FT encodes a putative phosphatidyl- 2. ZGT is a novel clock- and light-
ethanolamine binding and nucleotide regulated tobacco gene that links
binding protein, with similarity to Raf the circadian oscillator to LHCB
kinase inhibitor protein. The AGL20 and expression.
FT genes are also regulated by a second 3. COP1 may function as a repressor
flowering time pathway that acts independ- protein that targets different sub-
ently of CO, and their functions are geneti- strates for degradation by the pro-
cally redundant. teasome. SPA1 may link the
Two Arabidopsis CO-related proteins, phytochrome A–specific branch of
CONSTANS-like 1 (COL1) and CON- light signaling to COP1. Photoacti-
STANS-like 2 (COL2), are predicted to vated CRY1 and CRY2 and PHYB
encode proteins with 67% amino acid iden- also bind to COP1, repressing its
tity overall to the CO protein.All three pro- activity.
teins contain two adjacent N-terminal zinc 4. At the core of the oscillator, feedback
finger motifs that are 85% identical loops generated by reciprocal regula-
between the proteins. They also share high tion between the MYB-related
identity in a C-terminal basic region con- transcription factors CCA1 and LHY,
taining a putative nuclear localization and possibly RVEs, and the family of
signal (the CCT motif). COL1 and COL2 pseudoresponse regulators APRR,
transcripts levels are regulated by the cir- which includes TOC1, are responsible
cadian clock with a transcript peak around for generating circadian rhythms. The
dawn. Transgenic plants misexpressing MYB factors may function as activa-
COL1 and COL2 show little effect on flow- tors of genes peaking at dawn and
ering time. However, overexpression of repressors of genes peaking at dusk.
COL1 shortens the period of two distinct CCA1 and LHY are phosphorylated
circadian rhythms: CAB expression and by CK2, which may make them sub-
leaf movement. The fact that these circa- strates for the SCF complex and
dian defects are fluence-rate-dependent target them for degradation by the
may suggest an effect on a light input proteasome.
pathway. Collectively, all these data suggest 5. ZTL/ADO1 and related proteins
that there is an intimate interaction might act as novel blue-light photore-
between the circadian clock and photoperi- ceptors, using the LOV domain as a
odic timing. flavin binding site. Through the F-box
All possible molecular interactions domain, they may target proteins for
described within the plant circadian clock degradation via the ubiquitin system,
RHY8 2/6/04 3:57 PM Page 204

204 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

PIF4 ?

PIF3 CK2
CRY2 CCA1
LHY
COP1 RVEs
PHYB
COP1
ZGT CAB
Gi Other
morning genes
PHYC,D,E TOC1/ CCA1- P
other APRRs LHY- P
ELF3 RVE- P CAT3, CCR2, GI
SPA1
Other evening genes

COP1 COP1
SCF complex ?
HFR1
CRY1 PHYA ? Ubiquitin?
PIF3

TIM ?

output
? FLC

CRY1 CO
output
ADO1 FT AGL-20
ADO2 PHOT1
PHYB ADO3 RPT2
PHOT2 ? Flowering

Figure 8.7. Molecular interactions within the plant circadian clock. Both photoreceptors,
phytochromes and cryptochromes, mediate the effects of light–dark cycles on the phase and
period of the circadian clock. The effects of light on the clock are also modulated by the clock
itself through its regulation of both positive and negative light signaling components such
as GI and ELF3.

and via the kelch domain they may periodic changes in the environment.
recruit specific proteins for degrada- Output pathways come from the
tion, playing some role in protein oscillator to input components
turnover of clock-associated compo- known to be regulated by the clock at
nents and interacting with CRY1 and transcriptional, mRNA abundance, or
PHYB. protein abundance levels. Because
6. Arabidopsis contains a TIMELESS phytochromes and cryptochromes
homolog gene, although its role in the mediate light input to the clock, they
circadian oscillator remains to be are both clock input and output
elucidated. components, creating outer feedback
7. Clock-controlled expression of genes loops.
involved in the photosynthetic 8. Flowering is triggered by a change in
process, cell elongation, light signal- photoperiod. In Arabidopsis, CO pro-
ing pathways, and flowering-time reg- motes flowering in response to long
ulation provides the organism with photoperiods, mediating the control
the ability to anticipate and adapt to of flowering by the circadian clock.
RHY8 2/6/04 3:57 PM Page 205

MECHANISMS THAT CONFER CIRCADIAN REGULATION 205

CO can directly increase expression (278 and 119 bp) have also been defined
of the FT and AGL-20 genes, pro- for two tomato LHCA genes. A minimal
moting the transition to flowering. 330-bp clock-responsive promoter has been
FLC, a floral repressor, negatively defined for the Arabidopsis RCA gene,
regulates AGL-20. FLC and CO are whose expression oscillates in phase
proposed to have antagonistic effects with the LHC genes. A clock-responsive
on the expression of AGL-20 and FT. element sufficient to confer a low-ampli-
tude circadian oscillation lies within 317 bp
of the transcription start, but other ele-
䊏 MECHANISMS THAT CONFER ments necessary for high-amplitude circa-
CIRCADIAN REGULATION dian oscillation lie upstream of -317 and
downstream of -970. A CCA1 binding site
Both transcriptional regulation and post- is found in both LHCA and RCA function-
translational modifications have been ally defined minimal promoters, although
implicated in the control of the plant circa- the functional relevance of this motif to the
dian oscillator. circadian transcription of LHCA or RCA
has not yet been established.
In addition to promoters that confer
Promoter Elements
morning-specific transcription such as
Minimal nuclear promoters able to confer LHCB and RCA promoters, dusk-specific
circadian transcription have been identified promoters have also been studied. To
for several LHCB genes in different species, confer circadian transcription with a dusk-
tomato LHCA genes, the wheat psbD gene specific phase, 490 bp of the Arabidopsis
and the Arabidopsis RCA, GER3, CAT3, GER3 promoter are enough. Clock-
and TOC1 genes. The circadian clock asso- response elements contributing to high-
ciated gene CCA1, previously implicated in amplitude ATGER3 oscillations largely
phytochrome regulation, binds to a consen- reside between -967 and -299.
sus motif (AAa/cAATCT) widely con- On the other hand, 265 bp of the Ara-
served among CAB genes. bidopsis CCR2 promoter confer a robust
A promoter deletion analysis of four high-amplitude rhythm with dusk-specific
LHC tomato genes in transgenic tobacco phase, and as few as 56 bp are sufficient to
plants led to the identification of a short confer a low amplitude oscillation. The
sequence of 47 nucleotides that is necessary smallest CCR2 promoter fragment contains
for conferring circadian Lhc mRNA oscil- a CCA1 binding site, as does a minimal pro-
lations. A novel motif, CAANNNNATC, is moter (230 bp) of the CAT3 catalase gene
conserved in 5¢ upstream regions of clock- that is sufficient to confer dusk-specific
controlled Lhc genes and overlaps with a transcription. The functional significance of
sequence for relevant phytochrome medi- the CCA1 binding site in the dusk-specific
ated gene expression. promoters has not been directly tested, but
In vivo analysis of progressively trun- the existence of CCA1 binding sites in pro-
cated LHCB 1*1 (CAB2) promoter frag- moters that are transcribed nearly 180°C
ments fused to luciferase reporter gene out of phase suggest that the mechanism by
defined a 36-bp region sufficient to confer which the phase of transcription is deter-
circadian transcription. In vitro analysis of mined will be very complex and due not
DNA binding by EMSA and DNA foot- only to phase-specific transcriptional acti-
printing identified binding sites for multiple vators. A model in which CCA1 and LHY
complexes in this promoter fragment from simultaneously regulate genes that are
-111 to -74. Minimal promoter fragments phased to different times of the day has
RHY8 2/6/04 3:57 PM Page 206

206 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

been proposed. MYB factors act as nega- other circadian clock-controlled genes and
tive elements that repress TOC1 expression causes early flowering in both LD and SD
and probably other genes having evening conditions, suggesting a role for protein
elements in their promoters, which include, phosphorylation mediated by CK2 in the
for example, GIGANTEA and CCR2. function of the arabidopsis circadian clock.
Furthermore, given the overall similarity In plants overexpressing CKB3, CCA1 may
between CCA1 and LHY, it is likely that be phosphorylated more rapidly, causing a
LHY protein, as CCA1, is also a positive shorter period length due to an increased
regulator of CAB genes. rate of degradation of the protein. This
might involve F-box proteins and the ubiq-
uitination pathways.
Phosphorylation
Some studies have revealed an involve-
In addition to transcriptional regulation, ment of CK2a in plant phototransduction
posttranslational modifications such as pathways. In Arabidopsis, CK2a antisense
phosphorylation and regulation of protein RNA affects the expression of some light-
stability have also emerged as pivotal regulated genes, although it has little effect
control steps in the plant circadian oscilla- on the inhibition of hypocotyl elongation
tor. Phosphorylation may regulate multiple by red light.
properties of clock proteins, including sta- In rice, heading date is determined
bility and intracellular localization. Inter- mainly by two factors: duration of the basic
estingly, CCA1 was found to associate with vegetative growth and photoperiod sensi-
CKB3, a regulatory b subunit of the Ara- tivity. Hd6, a QTL involved in circadian
bidopsis casein kinase 2 (CK2) in a yeast photoperiod sensitivity, encodes the a
two-hybrid screen. CK2 is a serine/threo- subunit of the protein kinase CK2. The
nine kinase that is present in all eukaryotic Kasalath allele of CK2a increases days to
cells examined to date. It has a heterote- heading. This effect is produced by a single
trameric structure a2-b2, consisting of two nucleotide substitution, which changes the
catalytic (a) and two regulatory (b) sub- premature stop codon (TAG) in a japonica
units. CK2 can phosphorylate several variety (Nipponbare) to a lysine codon
transcription factors that bind to the pro- (AAG) in an indica variety (Kasalath).
moter regions of light-regulated genes, These findings indicate again that the same
including CCA1, LHY, and G-box factors genes appear to be involved in photoperiod
(GBFs) in vitro. CK2 was also proposed to response in both SD and LD plants.
be involved in cyclic phosphorylation of an A well-defined system of post-
endosperm-specific transcription factor, transcriptional regulation is provided by
Opaque 2 (O2), whose phosphorylation analysis of PEP carboxylase. In CAM
status changes diurnally. O2 activity is plants, PEP carboxylase activity cycles as a
downregulated at night by both a reduction result of a circadian rhythm in the phos-
in O2 transcript and hyperphosphorylation phorylation state of the enzyme. Sucrose
of residual O2 protein, suggesting that reg- phosphate synthase activity in tomato is
ulatory gene activity during endosperm regulated in a circadian manner by a protein
development may be acutely sensitive to phosphatase. The rhythm in nitrate reduc-
a diurnal signal directed to the developing tase mRNA abundance in Arabidopsis also
seeds. reflects posttranscriptional control. Cyclic
Overexpression of CKB3 increases CK2 nitrate reductase activity is affected by cir-
activity in transgenic Arabidopsis plants cadian regulation of its expression, in large
and shortens the period of the rhythmic part through phosphorylation dependent
expression of CCA1, of LHY, and of four mechanisms that affect the level of mRNA.
RHY8 2/6/04 3:57 PM Page 207

MULTIPLE AUTONOMOUS CLOCKS IN PLANTS 207

䊏 MULTIPLE AUTONOMOUS CLOCKS dynamics of metabolic pools and limita-


IN PLANTS tions of CO2 diffusion between tightly
packed cells.
Considerable evidence supports the exis- Animal species support multiple periph-
tence of multiple oscillators in multicellular eral clocks in addition to the neural
plants. Most of the evidence takes the form pacemakers, such as the suprachiasmatic
of multiple rhythms running with different nucleus. Peripheral animal tissues maintain
periods (internal desynchronization), which circadian rhythms when isolated in culture,
was demonstrated in Phaseolus coccineus. indicating that these tissues contain
In Phaseolus vulgaris, rhythms in CO2 circadian clocks. Likewise, isolated plant
assimilation and stomatal aperture exhibit organs and tissues support circadian
a different period from the rhythm in leaf rhythms, as do the specialized cells that
movement. In Arabidopsis, the freerunning drive rhythmic leaf and stomatal move-
periods in leaf movements and CAB ments. In plants it is unclear whether
expression are different, although both are peripheral circadian clocks are mutually
shortened by the toc1-1 mutation. Similarly, coupled and whether there is a centralized
the gi-2 mutation shortens the period in leaf circadian pacemaker. The levels of several
movement but lengthens the period in gene plant hormones exhibit circadian rhythms,
expression. In the dark, the Lhcb transcrip- but it is unknown whether any such coor-
tion period lengthens to 30 hours, whereas dinated, rhythmic signals can synchronize
the oscillations in CCR1, CCR2, and CAT3 cellular clocks.
mRNA abundance and transcription retain In vivo reporter gene imaging has been
a 24-hour period, suggesting that they are used to demonstrate that the circadian
driven by distinct oscillators. systems of plant organs and localized areas
Transcription of the Arabidopsis and of tissues are functionally independent. The
wheat Cab genes in single tobacco trans- rhythmic expression of a single gene can be
genic seedlings exhibit independent set to three phases in different anatomic
rhythms in a developmentally regulated locations of a single plant, by applying
fashion. Indeed, a red-light-insensitive light–dark treatments to restricted tissue
oscillator and a phytochrome-coupled cir- areas. The rhythms of one organ are unaf-
cadian clock coregulate the oscillating fected by entrainment of the rest of the
expression of individual CAB genes at the plant, indicating that phase resetting signals
level of transcription at an early develop- are also autonomous. Thus, higher plants
mental stage. Free calcium is resonsible for appear to contain a spatial array of
driving the rhythm of Lhcb expression. autonomous circadian clocks that regulate
Although the rhythms of cytosolic calcium gene expression without a localized central
and Lhcb::luc peak at the same time, they pacemaker. The circadian rhythms of
freerun with different periods in continu- cyanobacteria and of unicellular eukary-
ous light, suggesting that different circadian otes, such as Chlamydomonas, also demon-
pacemakers control these rhythms. strate that a circadian clock (or clocks) can
The well-known circadian rhythm of the exist within a single cell and several mam-
CAM mode of photosynthesis is expressed malian cell types retain a functional circa-
as independent dynamic patterns of photo- dian oscillator in culture. Thus, it is possible
synthetic efficiency over a single leaf. The that circadian oscillators are distributed
biological clock controlling this rhythm is a throughout the intact plant and function
spatiotemporal product of many weakly independent of any central pacemaker and
coupled individual oscillators that operate possibly of each other. Simultaneous but
independently as a consequence of the independent entrainment of these clocks in
RHY8 2/6/04 3:57 PM Page 208

208 PHYSIOLOGICAL AND MOLECULAR CHARACTERISTICS OF PLANT CIRCADIAN CLOCKS

different anatomic parts may be responsible associated in an intricate manner. The


for synchronizing their circadian rhythms. distinction between light input and oscil-
lator mechanism is becoming increasingly
blurred, supporting the suggestion that the
䊏 ADAPTIVE SIGNIFICANCE OF circadian clock may have arisen from pho-
CIRCADIAN CLOCKS totransduction pathways. Similar regula-
tory mechanisms are employed in plant and
As circadian rhythms are ubiquitous in animal circadian clocks. But so far, the
nature, it appears that an accurate tempo- putative components of the Arabidopsis
ral control likely confers some adap- oscillator have no orthologs in the circadian
tive advantage. This adaptive fitness would systems of cyanobacteria, fungi, or animals.
be enhanced by the synchronization of an One common evolutionary characteris-
organism’s internal clock with the light– tic may be the presence of domains related
dark cycle imposed by the environment, to two-component signaling systems. The
and the advantages in phasing sunlight- PAS domains present in clock-associated
sensitive cellular events to the night has genes of animals and fungi show similarities
been suggested. More recent studies have to sensor domains and are found in several
addressed the adaptive fitness of dawn response genes to environmental, develop-
anticipation in cyanobacteria and green mental, or cellular signals. In Arabidopsis
algae. Chlamydomonas exhibits circadian several putative components of the circa-
rhythms in cell division and in sensitivity dian clock (phytochromes, PIF proteins,
to UV radiation. At the end of the day and ZTL and ZTL-like proteins) contain
maximal UV sensitivity occurs, and this is PAS-like domains. Phytochromes contain
coincident with the DNA division period. two PAS repeats at their C-termini, and a
histidine kinase–related domain similar to
that of sensor kinases of two component
䊏 CONCLUSIONS AND PERSPECTIVES signaling systems. However, phytochromes
seem to function as serine kinases.
Study of the plant circadian system has wit- Phytochrome-mediated phosphorylation
nessed a great deal of progress with identi- would be a mode of action for very rapidly
fication of many molecular components. induced phytochrome responses. PIF
The next and more difficult step will be the proteins containing a PAS domain are
assembly of these components in a coher- necessary for normal photoinduced signal
ent molecular model. There are no reliable transduction, and they bind to G-box
criteria with which to unambiguously assign sequence motifs found in various light-
molecules to roles as input, output, or oscil- and circadian-regulated promoters. ZTL/
lator components. The data available so far ADO1 and the related LKP2/ADO2 and
suggest that transcriptional regulation is FKF1/ADO3 have a degenerate form of a
very important to the function of the plant PAS domain more similar to that of the
circadian clock, as it is to other circadian phototropins PHOT1 and PHOT2 and to
systems. It is becoming increasingly clear neurospora WC1 and VIVID proteins. Such
that posttranscriptional regulation also LOV motifs are found in proteins that bind
plays a central role in the function of the flavins (FMN in the phototropin case and
circadian oscillator. The multiple levels of FAD for WC1) and are involved in redox
feedback on light input pathways indicate reactions. An argument for the common
that phototransduction pathways and the ancestry of clocks perhaps is the existence
circadian oscillator of Arabidopsis are of PAS-containing proteins in the circadian
RHY8 2/6/04 3:57 PM Page 209

FURTHER READING 209

clock from distantly related organisms. Fur- Carre, I (2001): Day-length perception and the
thermore, the G-box element, found in the photoperiodic regulation of flowering in
promoters of many light- and circadian- Arabidopsis. J Biol Rhythms 16: 415–423.
regulated plant genes, is the same sequence Devlin, P, Kay, S (2001): Circadian photopercep-
tion. Annu Rev Physiol 63: 677–694.
as the E-box promoter element that plays
Devlin, PF (2002): Signs of the time: Environ-
an essential role in the circadian regulation
mental input to the circadian clock. J Exp Bot
of animal PER genes. An alternative point 53: 1535–1550.
of view is that different clocks may have Harmer, SL, Panda, S, Kay, SA (2001): Molecu-
evolved convergently from related lar bases of circadian rhythms. Annu Rev Cell
molecules whose original function was to Dev Biol 17: 215–253.
mediate light perception. This is consistent McClung, C (2001): Circadian rhythms in plants.
with the idea that circadian clocks evolved Annu Rev Plant Physiol Plant Mol Biol 52:
under selective pressure to set some 139–162.
cellular events to the light or dark period of Millar, A (1999): Biological clocks in Arabidop-
the day as we mentioned earlier. The sis thaliana. New Phytol 141: 175–197.
Piechulla, B (1999): Circadian expression of the
elucidation of the circadian clock mecha-
light-harvesting complex protein genes in
nisms of plants will shed more light on the
plants. Chronobiol Int 16: 115–128.
evolutionary origins of the circadian Roden, LC, Carre, IA (2001): The molecular
rhythms. genetics of circadian rhythms in Arabidopsis.
Semin Cell Dev Biol 12: 305–315.
Somers, D (1999): The physiology and molecu-
䊏 ACKNOWLEDGMENTS lar bases of the plant circadian clock. Plant
Physiol 121: 9–19.
Work in the laboratory of ARC was sup- Staiger, D, Heintzen, C (1999): The circadian
ported by grants from NIH and USDA. system of Arabidopsis thaliana: Forward and
reverse genetic approaches. Chronobiol Int
16: 1–16.
FURTHER READING Yanovsky, MJ, Kay, SA (2001): Signaling net-
works in the plant circadian system. Curr
Barak, S, Tobin, EM, Andronis, C, Sugano, S, Opin Plant Biol 4: 429–435.
Green, RM (2000): All in good time: The Ara- Young, MW, Kay, SA (2001): Time zones: A com-
bidopsis circadian clock. Trends Plant Sci 5: parative genetics of circadian clocks. Nat Rev
517–522. Genet 2: 702–715.
RHY9 2/6/04 3:53 PM Page 211

Part IV
CIRCADIAN ORGANIZATION IN
COMPLEX ORGANISMS
RHY9 2/6/04 3:53 PM Page 213

9
MULTIPLE OSCILLATORS
Jadwiga M. Giebultowicz

䊏 INTRODUCTION oscillators in the organization of the timing


system vary substantially among different
In the previous chapters we learned that groups of animals.
molecular mechanisms underlying the cir-
cadian clock are governed by similar prin-
ciples in all organisms from the simplest 䊏 PHYSIOLOGICAL EVIDENCE FOR
(bacteria) to the most complicated MULTIOSCILLATORY SYSTEMS
(mammals). We also found that, no matter
how complex the organism, circadian oscil- Individual organisms display a multitude of
lations are generated at the level of a single behavioral, physiological, and molecular
cell. In this chapter, we discuss how cellular rhythms in a highly specific temporal order.
oscillators are organized into coherent cir- Various rhythms reach their peaks at dif-
cadian systems. Several lines of evidence ferent phases of the daily light–dark cycle,
demonstrate that animals contain multiple and show a specific waveform. Rhythmic
circadian oscillators. Circadian systems in changes in several physiological parameters
mammals seem to be organized hierarchi- monitored in human subjects provide a
cally with a central, or master, clock, which good example of this general principle (Fig.
coordinates many rhythmic phenomena 9.1). Body temperature is elevated through-
in the body. It is increasingly evident, out the day. Plasma levels of growth
however, that circadian systems also hormone rise to a brief and sharp peak in
include many peripheral oscillators coordi- the early night, while levels of cortisol
nating tissue-specific functions. The relative display a broad peak during the late night
contributions of central and peripheral and morning. The rate of urinary potassium

Molecular Biology of Circadian Rhythms. Edited by Amita Sehgal


ISBN 0-471-41824-2 Copyright © 2004 John Wiley & Sons, Inc.

213
RHY9 2/6/04 3:53 PM Page 214

214 MULTIPLE OSCILLATORS

Body temperature provide an excellent model system in which


to explore circadian organization, because
they possess a rich repertoire of overt
rhythmic activities such as ecdysis, foraging,
wandering, mating, and oviposition. How
can one test whether one or more clocks
Growth hormone
drive the different rhythms? One way is
to monitor several rhythms in the same
species in constant darkness and tempera-
ture. In these conditions, devoid of zeitge-
bers, each rhythm should reveal its
Cortisol endogenous, freerunning period. Experi-
ments of this kind performed in the fleshfly
demonstrated that various rhythms had dif-
ferent freerunning periods. For example,
the rhythm of larval wandering displayed a
Potassium periodicity of about 20 hours, while the
rhythm of pupal eclosion had a period close
to 24 hours. It was concluded that function-
ally separable oscillators are likely to gen-
erate rhythms with different endogenous
periods. However, it is conceivable that the
same oscillator may change its endogenous
17 23 5 11
periodicity in subsequent developmental
Time of day (hours)
stages.
Figure 9.1. Approximate profiles of several
rhythmic physiological parameters in a
human subject monitored in light–dark
cycles. Each rhythm has a characteristic phase Internal Desynchronization between
relationship and waveform. [Modified from Different Rhythms in an Organism
Moore-Ede et al. (1982).]
To test the relationships between different
rhythms co-existing in the body, one can
excretion is high during the day and low at monitor their adjustments following a
night. Are different rhythms under the phase shift of a light–dark cycle. It may be
command of a single central oscillator, or expected that rhythms driven by the same
are they governed by many separate ones? oscillator would shift in a more coordinated
In 1960, Dr. Colin Pittendrigh predicted fashion than would those driven by sepa-
that “A multicellular system is literally a rate oscillators. When such experiments
population of oscillators” based on very were performed in humans, some rhythms,
limited experimental data available at the such as rest-activity cycles reentrained
time. Since then, much experimental evi- within a few days, while other rhythms, such
dence has accumulated in support of this as fluctuations in body temperature or
statement. cation excretion, took longer to reentrain.
Similar results were obtained in the squir-
rel monkey. After an 8-hour phase delay of
Freerunning Period Variation
the light–dark cycle the activity rhythm
Different rhythms in one species may have resynchronized in one day, the body tem-
divergent freerunning periods. Insects perature rhythm took 3 days, and the
RHY9 2/6/04 3:53 PM Page 215

MOLECULAR EVIDENCE FOR MULTIOSCILLATORY SYSTEMS 215

rhythm of urinary potassium excretion took testes–vas deferens complexes cultured in


7 days. Different rates of reentrainment vitro.
result in a transient internal desynchro- Similar local and tissue-specific oscilla-
nization, during which coexisting rhythms tors exist in cockroaches. A daily rhythm
display abnormal phase relations with each has been reported in the formation of the
other. outer body cover, the cuticle, by underlying
Another interesting observation con- epidermal cells. When small pieces of cock-
cerning the organization of circadian roach epidermis were cultured in vitro,
rhythms was made on humans living in iso- they maintained the rhythmicity of cuticle
lation without any time cues. In conditions secretion.
deprived of Zeitgebers, circadian rhythms The persistence of rhythmic activities in
in some subjects begun to freerun with isolated tissues is not limited to inverte-
independent periods, a phenomenon brates. A classic example of an oscillator
termed spontaneous internal desynchro- that continues to operate in vitro is the
nization. For example, in one human pineal gland of many nonmammalian ver-
subject, the rhythm of rest–activity and the tebrates. This gland is responsible for the
rhythm of urinary calcium excretion spon- rhythmic synthesis and release of mela-
taneously began to oscillate with a period tonin (Chapter 10). It is well documented
of 32.6 hours, while the rhythm of body that the rhythmic production of melatonin
temperature continued to oscillate with a persists in isolated pineal glands and even
period of 24.7 hours. Similar phenomena in dispersed pinealocytes of birds. Similarly,
have been observed in other mammals retinas of several vertebrates rhythmically
kept in constant conditions. Both transient produce melatonin, and these rhythms
and spontaneous internal de-synchroniza- persist in cultured eyes. The major timing
tion suggests that separate oscillators center of mammals, the suprachiasmatic
may generate different rhythms in one nucleus (SCN) of the brain, displays strong
organism. rhythms in electrical activity, metabolic
rate, and neuropeptide levels. Daily fluctu-
ations in spontaneous neural activity con-
tinue in the SCN maintained in vitro and,
Circadian Rhythms in Isolated Organs
impressively, even in a few cultured
A strong argument for the existence of mul- neurons derived from the SCN. Nonneural
tiple circadian oscillators in animals is the tissues of mammals also display physiolog-
persistence of circadian rhythms in differ- ical rhythms when isolated in vitro. More
ent organs isolated from the body and recently, molecular rhythms have been
maintained in culture. Work on mollusks reported in such tissues (see below).
and insects suggests that oscillators in
invertebrates reside in various tissues.
Marine mollusks, Aplysia and Bulla, display 䊏 MOLECULAR EVIDENCE FOR
rhythms of electrical activity in a group of MULTIOSCILLATORY SYSTEMS
neurons at the base of the eye. Those basal
retinal neurons maintain robust rhythms Several clock genes have been identified
when cultured in isolation from the rest of and characterized in the fruitfly (Chapter 3)
the body. In insects, some physiological and mouse (Chapter 4), and their homologs
rhythms are maintained in peripheral have been cloned in other invertebrates
tissues that lack innervations. For example, and vertebrates. Studies of the spatial pat-
the rhythm of sperm release from the testes terns of clock gene expression provide com-
to the vas deferens in moths persists in the pelling support for the earlier physiological
RHY9 2/6/04 3:53 PM Page 216

216 MULTIPLE OSCILLATORS

data that predicted the existence of mul- eral organs and are consistent with their
tioscillatory circadian systems in animals. function as clock proteins (see Chapter 3).
Rhythmic activities of clock genes and their PER and TIM dimerize in the cytoplasm
products are found in a surprisingly broad during the early night, and later accumulate
range of tissues in all examined animals, in cell nuclei. In both the brain and periph-
from insects, through lower vertebrates to eral tissues, TIM is degraded in response to
mammals. the morning light, while PER remains in the
nuclei through midday. A temporal profile
of both proteins in the excretory organ of
Drosophila, the Malpighian tubules, is
Clock Genes in Drosophila
shown in Figure 9.2.
Various techniques used to study the To study the expression of per and tim
expression of clock genes in the fruitfly genes in greater detail, several lines of
consistently revealed wide distribution of transgenic flies were created which report
per mRNA and protein in many tissues. the activity of clock genes. Particularly
Cycling PER expression was detected in useful are flies that carry sequences coding
certain areas of the CNS, including several for firefly luciferase fused to the promoter
subsets of brain neurons, glial cells, pho- region of either period (per-luc) or timeless
toreceptors, and chemosensory hairs. The (tim-luc) genes. Tissues expressing clock
PER protein was also found in many genes emit measurable light (when
peripheral tissues outside of the central luciferase acts on its substrate, luciferin) in
nervous system, such as the digestive, excre- proportion to the activity of the respective
tory, and reproductive systems (Table 9.1). transgene. This allows for the observation
Tissues that express PER also express of the activities of per and tim genes in real
another clock protein, TIMELESS (TIM). time. The sensitivity of this technique is so
Patterns of PER and TIM expression are high that per or tim expression can be con-
similar in the cells of the CNS and periph- tinually monitored in individual organs

䊏TABLE 9.1. Expression of Clock Genes in Insects and Vertebrates


Insectsa Vertebratesb
Cycling Cycling
Tissues and Organs Present In vivo In vitro Present In vivo In vitro
Brain neurons + + ? + + +
Brain glial cells + + ? ?
Eye + + ? + + +
Chemosensory organs + + + +
Skeletal muscles - + + +
Heart ? + + +
Respiratory system - + + +
Digestive system + + ? ?
Liver (fat body)c + + ? + + +
Renal organs + + + + + +
Male reproductive system + + + + ? ?
a
Data based mostly on Drosophila.
b
Based on mammals and zebrafish.
c
Fat body of insects is a functional equivalent of vertebrate liver.
RHY9 2/6/04 3:53 PM Page 217

ORGANIZATION OF MULTIOSCILLATORY SYSTEMS 217

(a) 4 8 12 20 24
rhythms. This indeed turned out to be the
16
case, but, unexpectedly, high expression of
PER clock genes was also detected in other parts
of the brain, and in various peripheral
tissues. Clock genes that show rhythmic
TIM activity in the SCN are also rhythmic in
other tissues (Table 9.1). As in flies, trans-
genic technology helped to demonstrate
(b)
that the rhythmic expression of clock genes
TIM
persisted in isolated organs. Rhythmic bio-
PER
luminescence was recorded in cultured
Nuclear stain

livers, lungs, and skeletal muscles taken


from transgenic rats expressing luciferase
under the control of the Per1 promoter. The
circadian mechanism appears to exist not
only in internal organs freshly explanted
from the body but is also preserved in rat
fibroblasts that existed for many years only
4 8 12 16 20 24 4
as cell lines. These fibroblasts can initiate
Time (hours) rhythmic expression of clock genes and
Figure 9.2. Expression of clock proteins PER some clock-controlled genes when high
and TIM in Malpighian (excretory) tubules of concentrations of serum are added to their
D. melanogaster: (a) fragments of tubules culture medium.
showing nuclear location of both proteins; Besides mammalian tissues, oscillations
(b) quantification of the intensity of PER and in the expression of clock genes were
TIM signal [modified from Ivanczenko et al.
(2001)]. observed in the cultured organs of
zebrafish, such as the heart and kidney.
These examples of rhythmicity in phyloge-
over several days. An example of the record netically distant organisms, from insects,
of per and tim activity in individually cul- through fish, to mammals, suggest that self-
tured Malpighian tubules and rectal tissues sustained peripheral oscillations may be a
is shown in Figure 9.3. Results of such conserved feature in all complex animals.
experiments confirmed that various fly This phenomenon may be explained from
organs rhythmically express clock genes an evolutionary perspective. Circadian
and demonstrated that these rhythms are mechanisms are very ancient; they have
self-sustained. been identified in bacteria and unicellular
eukaryotes. As these simple organisms
evolved into tissues of higher animals, the
Clock Genes in Vertebrates
descendent tissues apparently retained
Clock genes are rhythmic in many verte- their rhythm-generating capacities.
brate organs. Several genes involved in the
circadian mechanism are known in
mammals, including Clock (Clk) and three 䊏 ORGANIZATION OF
orthologs of the period gene of Drosophila. MULTIOSCILLATORY SYSTEMS
Where in the body are those genes
expressed? Many expected to find Clk and The relative independence of different
per mRNA in the SCN, which plays a rhythms in a single organism and the per-
central role in generating many mammalian sistence of clock functions in various
RHY9 2/6/04 3:53 PM Page 218

218 MULTIPLE OSCILLATORS

4000
per-luc MT
3500 per-luc rectum

3000

2500

2000

1500

1000
Bioluminiscence

8500

tim-luc MT
6500 tim-luc rectum

4500

2500

500

Time (hours)

Figure 9.3. Real-time expression of per-luc and tim-luc reporter genes in individual body
tissues. Bioluminescence emanating from individual Malpighian tubules (MT) or rectal tissues
(rectum) was recorded hourly. The upper panel shows the average plot of per-luc expressing
tissues, the lower panel shows the average plot of tim-luc expressing tissues. Black and white
bars indicate 12-hour periods when the lights were on and off, respectively; shaded bars indi-
cate dark periods, when the lights would be on in LD. An increase in the amplitude of cycling
on reexposure of tissues to LD (arrow) indicates that the two peripheral oscillators are directly
light-responsive. [Reprinted from Current Biology 10, Giebultowicz et al., Transplanted
Drosophila excretory tubules maintain circadian clock cycling out of phase with the host, pp
107–110, [copyright (2000) with permission from Elsevier Science.]

isolated organs demonstrated that animal master, or central, oscillator, which is


circadian systems consist of multiple oscilla- entrainable by light and regulates the light-
tors. How do these oscillators work together insensitive peripheral oscillator via neural
to ensure internal temporal order? There or humoral signals. In the following para-
are sparse experimental data available from graphs, we will review available data con-
different taxonomic groups that show a cerning the organization of the circadian
broad spectrum of relationships between system in different groups of animals.
oscillators. At one end of this spectrum are
insects, whose circadian systems include
Autonomous Oscillators in Insects
autonomous, independently entrained oscil-
lators. At the other end are mammals with a Among the many behavioral rhythms dis-
hierarchical circadian system containing a played by insects, the best studied are the
RHY9 2/6/04 3:53 PM Page 219

ORGANIZATION OF MULTIOSCILLATORY SYSTEMS 219

rhythm of rest–activity and the rhythmic nae were separately cultured in vitro for
emergence of adults from pupal cases, a several days. Bioluminescence emitted
process known as eclosion. From early on, from these appendages was rhythmic.
scientists were compelled to search for Moreover, the phase of these rhythms
pacemaking centers controlling those shifted in response to a shift in the
rhythms. In a set of classic experiments per- light–dark cycle applied in vitro. Further
formed on moths, it was demonstrated that experiments attributed oscillatory proper-
the oscillator located in the central part of ties to chemosensory organs, which are dis-
the brain controls the eclosion rhythm. tributed on most fly appendages. In another
Cross-transplantation of brains between set of experiments, various internal organs,
two species of moths showing eclosion such as the Malpighian tubules, rectum, and
peaks at different times of the day, caused testes, taken from per-luc and tim-luc flies
the recipient moths to eclose at the time were cultured in vitro. Despite being sepa-
characteristic for the donor species! Oscil- rated from each other, different tissues
lators, which control rest–activity rhythms displayed synchronous oscillations that
in cockroaches, crickets, and flies, are also persisted in constant darkness and in-
located in the brain. The most extensive creased in amplitude upon re-introduction
studies on the localization of the circadian of light–dark cycles (Fig. 9.3). Collectively,
oscillator controlling behavior have been these experiments demonstrate that fly
performed in Drosophila melanogaster. The peripheral organs contain self-sustaining
task was difficult because the fly brain con- circadian oscillators, which can be directly
tains many groups of neurons and glial cells entrained by light–dark cycles.
that express clock genes. As described in The fact that the fly circadian system is
Chapter 3 , several genetic and molecular composed of the central oscillator in the
approaches were used to locate the circa- brain, and an array of peripheral oscillators,
dian pacemaker controlling behavioral prompted us to examine relations between
rhythms in a cluster of ventrally located those components of the timing system.
lateral neurons (LNv) at the border Several lines of evidence suggest that oscil-
between the central brain and the optic lators in peripheral organs may be inde-
lobes. Genetic removal of all LNvs renders pendent of the clock in LNvs or any other
flies behaviorally arrhythmic, demonstrat- part of the brain. First, clock molecules
ing that these cells are essential for gener- cycle with similar phases in the brain and
ating the rest–activity rhythm. LNvs peripheral tissues. For example, rhythms of
are often referred to as the central clock in per, tim, and their respective proteins do
flies, because of their master role in the not show any significant phase lag in renal
control of behavior and their location in the tubules and rectal tissues relative to the
brain. brain clocks, when examined in vitro (Fig.
The clock in the LNvs is one of many 9.3) or in decapitated flies (Fig. 9.4a). The
components of the fly circadian system. second argument against the dominant role
Clock genes are widely expressed in D. of the brain comes from monitoring of the
melanogaster in a pattern consistent with resetting of peripheral clocks in the
the existence of other oscillators. Use of Malpighian tubules. For this experiment, a
transgenic flies made it possible to demon- group of flies was decapitated and sub-
strate that such oscillators are indeed jected, along with intact flies, to a reversal
located in various organs and that they of the LD cycle. The phase shift in the oscil-
display a remarkable degree of autonomy. lation of the per gene in response to
In one experiment, parts of per-luc flies reversed LD occurred with a very similar
such as the wings, legs, proboscis, and anten- time course in intact and decapitated flies
RHY9 2/6/04 3:53 PM Page 220

220 MULTIPLE OSCILLATORS

Intact Decapitated
(a)

Relative per levels


(b)

0 12 24 36 48 60
Time (hours)

Figure 9.4. The temporal expression of a PER reporter in the Malpighian tubules of D.
melanogaster. The onset of the first experimental dark period is termed 0 hour. Headless flies
(black lines) were decapitated a few hours prior to time 0 hour and later treated as control
intact flies (dotted lines). Malpighian tubules were assayed every 6 hours. (a) Flies maintained
in LD conditions exhibit similar PER oscillations regardless of whether the head oscillators are
present. (b) Intact and headless flies subjected to a phase shift of LD cycle, with a 12 hour
extension of the first dark period, reset with a similar time course. [Adapted from Hege et
al. (1997).]

(Fig. 9.4b). These data suggest that the the vas deferens was described in moths, it
central clock does not mediate resetting of was first assumed that the brain would also
the peripheral clock, but rather the control it. Experiments, however, proved
Malpighian tubule clocks seem to be otherwise. The rhythm of sperm release
directly reentrained by environmental persisted in brainless moths with a phase
cycles. In a third experimental approach, similar to that in intact moths. Furthermore,
tubules were transplanted into host flies light applied to isolated testes–vas deferens
entrained to an opposite light–dark cycle complexes shifted the phase of sperm
(relative to donor flies) and kept in con- release rhythm. These experiments clearly
stant darkness after the surgery. Under demonstrated that the reproductive system
those conditions, the clock in the donor of the male moth contains an autonomous
tubules cycled out of phase relative to host oscillator that is directly photoresponsive
tubules, even though both sets of tubules and able to operate independently of the
shared the same hormonal milieu. Appar- brain.
ently, the rhythms of the transplanted organ Despite the great autonomy of some
were unaffected by the host fly. Thus, at peripheral clocks, specific insect oscillators
least some peripheral oscillators in may be subject to modulating signals from
Drosophila seem to operate as independent other oscillators, as was demonstrated in
units, achieving synchrony due to their sen- the hemipteran bug, Rhodnius prolixus.
sitivity to a common Zeitgeber in the form These insects display pronounced daily
of external light–dark signals (Fig. 9.6a). fluctuations in the levels of circulating
Similar brain independence of periph- molting hormone, ecdysone. Glands pro-
eral oscillators is observed in other insects. ducing this hormone contain their own pho-
It has been known for many decades that tosensitive circadian oscillator, which is
the circadian clock located in the brain con- capable of timing the release of ecdysone in
trols behavioral rhythms in moths. When vitro. In intact animals, however, this oscil-
the rhythm of sperm release from testes to lator is entrained by the rhythmic release of
RHY9 2/6/04 3:53 PM Page 221

ORGANIZATION OF MULTIOSCILLATORY SYSTEMS 221

tropic hormone that is controlled by perature but not the rhythm of rest–activ-
another oscillator located in the brain. ity, or the rhythm of retinal sensitivity to
Similar relationships may exist between the light. Circadian organization has been also
brain and the endocrine glands in other studied in several species of birds.The avian
insects. The experiments performed in circadian system is composed of several
Rhodnius show that some oscillators with interacting sites, each containing photosen-
direct sensitivity to light may be neverthe- sitive circadian oscillators. These sites
less modulated by internal messengers include the pineal gland, the suprachias-
derived from other clocks in the body. matic nucleus of the brain, and in some
Therefore, the conclusions about the inde- species, the eyes. The pineal gland seems to
pendent status of a given oscillator should be the locus of a major circadian pace-
be made only after phase of the oscillations maker, which may control the period and
was determined both in vivo and in vitro. the phase of other circadian oscillators. In
the absence of this gland, some birds, such
as sparrows, are unable to maintain a coher-
Multioscillatory Circadian Organization
ent circadian organization. However, signi-
in Lower Vertebrates
ficant variations occur among birds in the
The results discussed above suggest that relative importance of this organ. For
peripheral pacemakers in flies have a high example, removal of the pineal gland does
degree of autonomy. The same situation not affect body temperature or activity
seems to prevail in lower vertebrates as rhythms in the Japanese quail.
well. In the zebrafish, the expression of the
Clk gene is rhythmic in the kidney, spleen,
Hierarchical Organization of
and heart, and the oscillations have similar
Circadian System in Mammals
phases in vivo and in vitro. Subsequent
studies have shown that Clk rhythms in iso- Mammals provide compelling evidence
lated zebrafish organs are entrainable for a hierarchically organized circadian
directly by light–dark cycles. The resetting system. Virtually all mammals display daily
of the Clk mRNA rhythms in vitro suggests rest–activity cycles. These rhythms have
that the brain does not mediate this process. been studied particularly closely in rodents
Thus the organization of the circadian such as the hamster and rat, where long-
system in the zebrafish appears to be term behavioral records can be easily
similar to that of Drosophila in that many obtained. The realization that these
internal organs harbor self-sustained pho- animals are able to maintain an activity
toreceptive oscillators. The degree of cycle of about 24 hours without any envi-
autonomy these oscillators have in vivo and ronmental cues led to a search for the loca-
their possible coordination by any humoral tion of the “master clock.” Selective
messengers have not been investigated in destruction of a part of the hypothalamus,
the zebrafish. the suprachiasmatic nucleus (SCN),
Multioscillatory circadian organization resulted in the abolishment of all behav-
also extends to other vertebrates. Experi- ioral and endocrine rhythms (see also
mental and surgical manipulations in a Chapter 4). Removal of other parts of the
reptile, the green iguana, have shown that brain, endocrine glands, or peripheral
their timing system is composed of multiple tissues did not cause such global arrhythmia
circadian oscillators that reside in different as the removal of the SCN. The central
tissues, and have specific and different roles. status of the SCN was confirmed using
For example, the removal of the pineal mutant hamsters with a significantly shorter
gland abolishes the rhythm of body tem- circadian period. When the SCNs of such
RHY9 2/6/04 3:53 PM Page 222

222 MULTIPLE OSCILLATORS

mutants were transplanted into the hypo- hours relative to the SCN. Moreover, in
thalamus of the SCN-lesioned wild-type response to advances or delays in the envi-
hamsters, the host animals developed ronmental light cycle, the circadian rhythm
rhythms with a shorter period characteris- in the SCN shifted more rapidly than the
tic of the SCN donors. rhythms in peripheral tissues. These studies
How does the SCN control multiple confirm the special status of the SCN oscil-
rhythms in the body? Some rhythms, lator, and suggest that the light-entrainable
such as behavioral activity, can be restored SCN may coordinate phases of peripheral
by a transplanted SCN, which is experi- oscillators, which are not entrainable by
mentally prevented from establishing light input, as depicted in Figure 9.6b.
neural connections with the host nervous While the SCN acts as a master clock in
system. This suggests that the SCN conveying light–dark information to the
pacemaker produces humoral outputs rest of the body, there may be other entrain-
affecting target tissues. Other rhythms, such ing factors that affect rhythmic behavior
as cortisol or melatonin secretion, are not irrespective of the SCN. It has been known
restored by transplants and may require for a number of years that cycles of food
formation of precise connections between availability exert powerful entraining
the SCN graft and the target neurons. effects on the rhythm of locomotor activity
Thus, the SCN may transmit clock signals to in both intact and SCN-lesioned rodents.
various tissues by both humoral and One study compared the effects of
neural pathways. Taken together, these restricted feeding and light–dark cycles on
experiments left little doubt that the the rhythms of clock genes in the SCN and
mammalian circadian system is hierarchi- peripheral oscillators. Restricted feeding
cally organized, and that the SCN is at the rapidly entrained the liver, shifting its
top of hierarchy. rhythm by 10 hours within 2 days, while the
While the SCN is the central pacemaker, oscillations in the SCN remained phase-
the mammalian timing system appears to locked to the light-dark cycle. It is not yet
also have other components: (1) some phys- known which kind of signals associated
iological rhythms persist in mammals with with feeding affect the phase of the liver
lesioned SCN, and even in isolated periph- oscillator. Nevertheless, these experiments
eral organs in vitro; and (2) many organs demonstrate that individual peripheral
outside of CNS rhythmically express clock clocks may be sensitive to timing signals
genes (see previous section). Thus, periph- that are relevant to physiological functions
eral oscillators exist in mammals. The status of a given organ (Fig. 9.6b). Thus, the hier-
of those oscillators relative to the SCN was archy of the mammalian circadian system is
studied in a series of interesting experi- not rigid but flexible; direct sensitivity of
ments. To monitor oscillations in peripheral peripheral oscillators to external resetting
tissues, researchers used transgenic rats car- factors may aid in the adaptation of the
rying the promoter of the Per1 gene linked organism to its surroundings.
to a luciferase reporter. Cultured SCNs
taken from transgenic rats showed robust
Interspecies Variation in
oscillations, which persisted for several
Multioscillatory Communication
months. Other organs such as liver, lung,
and skeletal muscles also expressed circa- The degree of communication between dif-
dian rhythms in vitro, but those rhythms ferent oscillators within one circadian
tend to dampen sooner than in the SCN system depends on the phylogenetic posi-
(Fig. 9.5). The phase of the oscillations in tion of the organism. Evolution seems to
peripheral organs was delayed by several have proceeded from autonomous oscilla-
RHY9 2/6/04 3:53 PM Page 223

ORGANIZATION OF MULTIOSCILLATORY SYSTEMS 223

13,000

Bioluminescence (counts per min)


12,000

11,000

10,000

9000

8000

7000
0 1 2 3 4 5 6 2 9 30 31 32
Day

SCN Skeletal muscle


16,000
10,000
Bioluminescence (counts per min)

8000 12,000

6000 8000
0 1 2 3 4 0 1 2 3 4
Liver 90,000 Lung
200,000

75,000

100,000
60,000

0 1 2 3 4 0 1 2 3 4
Day

Figure 9.5. The circadian rhythm of bioluminescence from tissues of Per1-luc transgenic rats.
In the SCN the near-24-hour rhythm was observed, which peaked in the middle of the sub-
jective day and persisted for more than 32 days in vitro (upper panel). Circadian rhythms were
expressed in vitro in different tissues from the same animal (lower panel). Note that oscilla-
tions in peripheral tissues peaked several hours later than in the SCN. In contrast to SCN,
rhythms in peripheral tissues dampened after only a few cycles in vitro. [Reprinted with per-
mission from Yamazaki et al. (2000). Copyright (2000) American Association for the Advance-
ment of Science.]

tors that are independent of each other, and to restricted tissue areas. The initial
synchronized by external light–dark cycles, phases were stably maintained after the
to a less autonomous and more coupled entraining treatments ended, indicating that
system of oscillators. An extreme autonomy circadian oscillators in intact plants are
has been documented in plants. The rhyth- autonomous and do not communicate with
mic expression of a single gene could be set each other to coordinate the phases of their
at three phases in different anatomical oscillations.
locations of a single Arabidopsis plant, A similar lack of communication
by applying different light–dark treatments between oscillating organs has been docu-
RHY9 2/6/04 3:53 PM Page 224

224 MULTIPLE OSCILLATORS

(a) (b)

Nonphotic
zeitgebers

Figure 9.6. Relationship between multiple oscillators in circadian systems. (a) Multiple oscil-
lators are independently responsive to light–dark cycles; however, the output of the brain
oscillator may modulate the phase of another oscillator. Such relationships are observed
in insects and may also occur between different oscillators in lower vertebrates. (b) In
mammals, the SCN oscillator assumes the central role. This oscillator receives light input and
coordinates the phases of light-insensitive peripheral oscillators via its rhythmic neuronal and
humoral outputs. Peripheral oscillators may be independently entrained by external factors
other than light.

mented in insects, and may also occur in Humoral factors may be involved in the
lower vertebrates. The circadian system in communication between different oscilla-
these animals appears to function as decen- tors in vertebrate circadian systems (see
tralized collection of oscillators. The whole also Chapter 10). Blood levels of many
system achieves coordination by the sensi- vertebrate hormones display circadian
tivity of its components to common rhythms (see Fig. 9.1). While these hor-
light–dark cycles. However, insects have a monal fluctuations must have profound
well-developed neuroendocrine system, effects on the physiology of the target
and at least some insect hormones show tissues, it is not clear to what degree they
daily fluctuations in their concentrations. affect the phases of peripheral oscillators. A
Rhythmic humoral factors may play a role good candidate for a humoral circadian
in the internal coupling of some of the messenger in vertebrates, especially in
peripheral oscillators. However, they do not birds, is melatonin. This hormone, which is
seem to affect the phase of clock gene rhythmically produced by pineal glands and
cycling in the Malpighian tubules of retinas, shows robust circadian fluctuations
Drosophila (see previous section). in the blood of all examined vertebrates.
RHY9 2/6/04 3:53 PM Page 225

ORGANIZATION OF MULTIOSCILLATORY SYSTEMS 225

Exogenous administration of melatonin the adrenal gland, can transiently reset the
can cause either arrhythmicity or changes phase of oscillations in other organs.
in the period of freerunning rhythms. Con- In addition to hormonal signals, oscilla-
versely, periodic infusion of melatonin tors in specific tissues may be regulated by
restores rhythmicity to birds with a lesioned rhythmically fluctuating metabolites pro-
pineal gland. Many regions of the brain, as duced within or outside these tissues. This
well as cells of peripheral organs, have idea gained support from the demonstrated
melatonin receptors, and therefore, could molecular link between circadian oscilla-
be affected by this hormone’s changing tions and energy homeostasis. It appears
levels. The mechanism of melatonin action that cellular redox states are under circa-
as a transducer of biological time is not yet dian control and, via a feedback mecha-
well understood. nism, may affect the activities of the
The information channels within the molecular components of the circadian
multioscillatory system seem to be most clock. It is possible that other channels of
complex in mammals. What sets the mam- communication within the circadian system
malian circadian system apart is that will be revealed as we learn more about
peripheral oscillators do not appear to be functions and outputs of peripheral oscilla-
entrained by light–dark cycles. Instead they tors in animals.
seem to rely on humoral and neural timing
signals distributed by the light-entrainable
Variation in Molecular Clock
master oscillator in the SCN. Experiments
Components in Tissues of the
comparing immortalized cells derived from
Same Organism
the SCN and a fibroblast line demonstrated
the importance of the SCN-specific outputs While the core clock mechanism is shared
for circadian coordination (Fig. 9.7). SCN in cells forming various tissues, some clock
cells endogenously generate circadian components and light-entrainment path-
rhythms in Per gene expression and in ways appear to have tissue-specific roles. In
metabolic activity, and impose those Drosophila melanogaster, disruption of the
rhythms on cocultured fibroblasts via a night by even short light exposures results
diffusible signal. In contrast, cultured in degradation of the clock protein TIME-
fibroblasts, when induced by a serum LESS (TIM) leading to shifts in the fly
shock, generate a circadian rhythm in Per molecular and behavioral rhythms
gene expression but not in metabolic (Chapter 3). The blue-light photoreceptor
activity. Serum-shocked fibroblasts could cryptochrome (CRY) is involved in TIM-
not confer rhythmic functions on other mediated entrainment of the oscillators in
fibroblasts. the LNs and in the Malpighian tubules.
Although the special pacemaking prop- Experiments on flies with a mutated cryp-
erties of the SCN are very important in tochrome gene demonstrated that freerun-
circadian coordination, other channels of ning clock oscillations can continue without
communication also exist in the mam- CRY in LNs but not in Malpighian tubules
malian circadian systems. Peripheral oscil- or antennae. Thus, besides its role as a pho-
lators were shown to be entrained by toreceptor, cryptochrome is also an indis-
nonphotic external factors, which may pensable component of the endogenous
cause their uncoupling from the master clock mechanism in the fly excretory
clock. Internally, peripheral oscillators may system, but not in the brain. The lack of
communicate with each other via their CRY may be compensated for by as yet
rhythmic outputs. For example, corticos- unrevealed LN-specific clock components,
teroids, which are rhythmically produced by or by input from other structures in the
RHY9 2/6/04 3:53 PM Page 226

226 MULTIPLE OSCILLATORS

central nervous system. LNs are part of a oscillators present in the forebrain and in
complicated neural network, undoubtedly the smooth muscle of the vasculature. In
interacting with other cells via chemical and these tissues MOP4 (NPAS2), a transcrip-
electrical signaling. In contrast, Malpighian tion factor highly related to Clock, can
tubules consist of noninnervated epithe- replace the latter in forming heterodimers
lium in which clock functions do not seem with BMAL1. Despite the commonality
to be affected by the fly internal milieu. of the molecular circadian mechanism,
Thus, there may be more redundant mech- various oscillators play different roles in the
anisms keeping freerunning clocks in gear organism. This seems to be linked to the
in the brain than in the peripheral organs. nature of the output rhythms produced by
The molecular components of the clock oscillating cells. For example, a cluster of
mechanism may also differ in various oscil- neurons in the fly brain or in the mam-
lators of mammals. In the central SCN malian SCN may affect many target tissues,
pacemaker, two transcription factors, Clock via neural and humoral outputs, producing
and BMAL1, form heterodimers essential a response at the level of the whole organ-
for function of the oscillating feedback ism. On the other hand, an oscillator in the
loop. It has been found that another tran- excretory epithelial cells will likely exert
scription factor constitutes a component of only local effects, controlling levels of

(a)

SCN / fibroblast
cocultures

(b)
Serum Shock (SS)

Fibroblast (SS) / fibroblast


cocultures

Figure 9.7. Experiment demonstrating that SCN cells are distinguished by special pacemak-
ing properties. (a) Immortalized cells derived from the rat SCN (SCN2.2 cell line) generate
metabolic (solid line) and molecular (dashed line) rhythms and are able to confer these
rhythms onto cocultured fibroblasts (NIH/3T3 cell). (b) Serum-shocked fibroblasts generate
only molecular oscillations and cannot drive rhythmicity in cocultures of untreated fibrob-
lasts. (Diagram courtesy of David J. Earnest.)
RHY9 2/6/04 3:53 PM Page 227

ROLES OF PERIPHERAL CLOCKS IN PHYSIOLOGY 227

enzymes, ion channels, and other compo- H+ATPase. Rhythms associated with sperm
nents in those cells. release are driven by the per-based circa-
dian mechanism. per mRNA and PER
protein are rhythmically expressed in all
䊏 ROLES OF PERIPHERAL epithelial cells of the moth vas deferens.
CLOCKS IN PHYSIOLOGY Thus, this circadian system appears to
provide a temporal framework for coordi-
Rhythmic activities of clock genes and their nated maturation of sperm.
products are found in a surprisingly broad Another example of a peripheral oscilla-
range of organs. Clock molecules cycle in tor with a known function is found in
many loci within the CNS, in tissues Drosophila. A defined physiological output
involved in food intake, metabolism, excre- has been assigned to oscillators located in
tion, and reproduction. This strongly the chemosensory hairs on the fly’s anten-
suggests that peripheral circadian oscilla- nae. These organs display a rhythm in elec-
tors may be involved in coordinating trophysiological responses to two different
many physiological processes in a tissue- classes of olfactory stimuli. Olfactory
autonomous fashion. However, many rhythms are driven by clock genes
peripheral oscillators were identified on the expressed locally in the antenna, and they
basis of cyclic expression of clock genes, persist when the fly’s central clock in the
and their relevance to the organism’s phys- LNvs is genetically removed.
iology is not yet clear. The links between clocks and their phys-
There are a few cases in insects where iological outputs include several intermedi-
the physiological role of peripheral oscilla- ate steps leading from clock genes via other
tors is relatively well understood. The transcription factors to the effector genes
importance of the circadian coordination of that are involved in cellular physiology.
physiological processes is manifested dra- Some steps are known in both central and
matically in moths, in which constant light peripheral oscillators; a handful of rhythmic
(a known disrupter of insect rhythms) leads transcription factors and effector genes
to male sterility. This intriguing discovery have been identified, but in no case do we
was followed by the realization that the understand the whole clock-to-physiology
testes–vas deferens complex displays many cascade. Given this paucity of information
coordinated rhythms associated with a it is not clear whether circadian clocks
daily cycle of sperm release and matura- control diverse cellular processes in differ-
tion. Clones of differentiated spermatozoa ent tissues, or whether many cell types
(sperm bundles) are released from the share specific rhythmic aspects of cellular
testis to the vas deferens by penetrating the physiology. Convergent clock-controlled
epithelial barrier separating the two organs output pathways seem to occur in neurons
during the circadian gate at the end of the and epithelial cells. For example, one of the
day. After nighttime retention in the vas mammalian transcription factors, DBP,
deferens lumen, sperm is moved from this shows a circadian rhythm in both central
compartment because of the morning (SCN) and peripheral (liver) oscillators. In
increase in the intensity of contraction of D. melanogaster, clock-controlled oscilla-
the vas deferens muscles. The peak of tory expression of the gene takeout, which
sperm accumulation in the vas deferens is implicated in the control of feeding,
lumen is correlated with periodic acidifica- occurs in the brain but also in segments of
tion of this compartment (Fig. 9.8). Acidifi- the alimentary tract. We are probably
cation appears to be caused by a rhythm in seeing only the tip of the iceberg when it
the levels of the proton pump, vacuolar comes to clock-controlled genes. Microchip
RHY9 2/6/04 3:53 PM Page 228

228 MULTIPLE OSCILLATORS

(a) (b)
1200
7.2
1000
7
A
800
600 6.8

400 6.6
Mean number of sperm bundles in the UVD

200 6.4
0
6.2

1200 7.2
B

pH in the UVD lumen


1000
7
800
6.8
600
400 6.6

200 6.4
0
6.2

1200 7. 2
1000
C
7
800
6.8
600
400 6.6

200 6.4
0 6.2
4 8 1 2 16 20 24 4 8 12 16 20 24
Time (hours) Time (hours)

Figure 9.8. Pattern of sperm release (a) and pH changes (b) in the vas deferens (UVD) of S.
littoralis. In LD and DD, sperm bundles accumulate in the UVD at night, and this rhythm cor-
relates with maximum acidification of the UVD lumen in the middle of the dark phase. The
rhythms in both sperm release and pH are abolished in constant light. Horizontal bars rep-
resent day (white), night (black), and former day (hatched) portions of the photoregime.
(Composite figure from the author’s result.)

technology is revealing hundreds of rhyth- fusible signals from immortalized suprachias-


mically expressed genes in Arabidopsis, matic nucleus cells regulate circadian rhyth-
Drosophila, and mammals. Further studies micity in cultured fibroblasts. J Neurosci 21:
of the clock-controlled genes should help 7937–7943.
Buijs, RM, Kalsbeek, A (2001): Hypothalamic
reveal how the timing system ensure opti-
integration of central and peripheral clocks.
mization of everyday functioning of the
Nature Rev Neurosci 2: 521–526.
body, as well as adaptability to external Giebultowicz, JM (2001): Peripheral clocks and
environment. their role in circadian timing: Insights from
insects. Philos Trans Roy Soc Lond B Biol Sci
356: 1791–1799.
FURTHER READING Glossop, NR, Hardin, PE (2002): Central and
peripheral circadian oscillator mechanisms in
Allen, G, Rappe, J, Earnest, DJ, Cassone, VM flies and mammals. J Cell Sci 115: 3369–
(2001): Oscillating on borrowed time: Dif- 3377.
RHY9 2/6/04 3:53 PM Page 229

FURTHER READING 229

Hege, DM, Stanewsky, R, Hall, JC, Giebultow- Cold Spr Harb Symp Quant Biol 25: 159–
icz, JM (1997): Rhythmic expression of a 182.
PER-reporter in the Malpighian tubules of Schibler, U, Sassone-Corsi, P (2002): A web of
decapitated Drosophila: Evidence for a circadian pacemakers. Cell 111: 919–922.
brain-independent circadian clock. J Biol Tosini, G, Menaker, M (1998): Multioscillatory
Rhythms 12: 300–308. circadian organization in a vertebrate, Iguana
Ivanchenko, M, Stanewsky, R, Giebultowicz, iguana. J Neurosci 18: 1106–1114.
JM (2001): Circadian photoreception in Underwood, HA, Wassmer, GT, and Page, TL
Drosophila: Functions of cryptochrome in (1997): Daily and seasonal rhythms. In Hand-
peripheral and central clocks. J Biol Rhythms book of Physiology, Dantzler. WH, ed,
16: 205–215. Oxford University Press, New York, pp
Krishnan, B, Levine, JD, Lynch, MK, Dowse, HB, 1653–1763.
Funes, P, Hall, JC, Hardin, PE, Dryer, SE Whitmore, D, Foulkes, NS, Sassone-Corsi, P
(2001): A new role for cryptochrome in a (2000): Light acts directly on organs and cells
Drosophila circadian oscillator. Nature 411: in culture to set the vertebrate circadian
313–317. clock. Nature 404: 87–91.
Moore-Ede, MC, Sulzman, FM, Fuller, CA Yamazaki, S, Numano, R, Abe, M, Hida, A,
(1982): The Clocks that Time Us. Cambridge, Takahashi, R, Ueda, M, Block, GD, Sakaki, Y,
MA: Harvard University Press. Menaker, M, Tei, H (2000): Resetting central
Pittendrigh, CS (1960): Circadian rhythms and and peripheral circadian oscillators in trans-
circadian organization of the living systems. genic rats. Science 288: 682–685.
RHY10 2/6/04 3:56 PM Page 231

10
HORMONAL RHYTHMS
Peter McNamara

䊏 INTRODUCTION basis in many animal species, including


humans. Underlying the behavioral adapta-
The ubiquity of circadian rhythms in all tions to the daily changes in the external
aspects of eukaryotic life emphasizes their environment are a multitude of metabolic,
evolutionary importance for the survival hormonal, and biochemical rhythms. For
of the species. The functional significance of example, changes in the digestive system
circadian rhythms may provide synchro- occur before the behavioral changes that
nization with the pronounced periodic fluc- lead to food intake, and the rise in body
tuations of the external environment and temperature occurs before an animal wakes
may also organize the internal milieu so up in anticipation of the increased meta-
that there is coordination and synchroniza- bolic demands. This intricate temporal
tion of internal processes. Many behaviors organization provides the endocrine system
such as feeding, drinking, sleep, exploration, with remarkable flexibility. Not only can
and reproductive activity that change on a specific physiological processes be turned
daily basis are correlated with daily changes on and off depending on the presence or
in the physical environment (e.g., tempera- absence of a particular hormone, but also
ture, illumination, humidity) as well as the precise pattern of hormonal release
changes in the biological environment (e.g., may provide specific signaling information.
food availability, presence of predators, There is virtually neither tissue nor func-
parasites, competitors, reproductive mates). tion within the human organism that does
Associated with these behavioral changes not manifest regular changes from day to
are changes in perception, sensation, learn- night. Apart from rhythms in the endocrine
ing, and performance that occur on a daily system, which is the focus of this chapter,

Molecular Biology of Circadian Rhythms. Edited by Amita Sehgal


ISBN 0-471-41824-2 Copyright © 2004 John Wiley & Sons, Inc.

231
RHY10 2/6/04 3:56 PM Page 232

232 HORMONAL RHYTHMS

circadian variations outside the endocrine diurnal and circadian rhythms, since almost
system include innumerable physiological all diurnal rhythms are expressed under
variables, such as body temperature, heart constant environmental conditions in the
rate, blood pressure, urinary volume, laboratory. In this chapter the use of the
propensity for rapid eye movement (REM) term circadian rhythm is extended to mean
sleep, intraocular pressure, and taste thresh- all diurnal variations recurring regularly at
old for salt; a wide variety of blood con- a time interval of approximately 24-hour,
stituents, such as white blood cells, amino and we will concentrate on hormonal rhy-
acids, and phosphorus; as well as behavioral thms in humans with in-depth descriptions
parameters such as time of birth, time of of some of the more well-characterized
death, mood, reaction time, performance on hormonal axes.
learning tasks, computation skills, pattern
recognition, and relative coordination.
There are also rhythms in responsive to 䊏 NIGHTTIME MELATONIN
various challenges such as drugs and stress. PRODUCTION: A KEY HORMONAL
Circadian rhythmicity is maintained when OUTPUT FROM THE CLOCK
subjects are sleep-deprived, when they are
starved, and when they receive equal In vertebrates melatonin is synthesized
amounts of food at short intervals over the only by the retina and pineal gland, which
day. The timing of single meals, however, is a small structure located in the center of
can have effects on the pattern of at least the skull between the two cerebral hemi-
some variables, including hormones, and spheres. In birds, reptiles, amphibia, and fish
the sleep–wake cycle can have phase- the pineal gland is directly light-sensitive
setting as well as phase-masking effects and possesses an independent circadian
on many rhythms, especially those of the clock. In contrast, in mammals, pinealocytes
endocrine system. neither are light-sensitive nor possess a
Many diurnal hormonal patterns are clock, which is instead located in the SCN.
dependent, to some degree, on the circa- The nocturnal rise starts early in the
dian clock. The relative contributions of evening, between 8 and 10 pm, and the
circadian rhythmicity versus homeostatic maximum occurs around the middle of
control in the temporal organization of the sleep period. The melatonin rhythm and
hormonal release differ from one the rhythm of body temperature undergo
endocrine axis to another. For most pitu- equivalent phase shifts following a regimen
itary hormones, the 24-hour profiles reflect of scheduled exposures to bright light and
the superposition of circadian signals on an darkness, supporting the concept that both
ultradian, or pulsatile, release and result rhythms are controlled by the central cir-
from interaction of the circadian clock with cadian pacemaker. Exposure to light of
sleep–wake homeostasis (ultradian rhythm sufficient intensity results in an acute dose-
is used to designate rhythmicities with dependent inhibition of nocturnal mela-
periods ranging from fraction of hours to tonin secretion, with a rapid return to high
several hours). Several rhythmic and non- nighttime levels when darkness resumes.
rhythmic facets, such as periodic food The melatonin rhythm appears to be
intake, postural changes and levels of phys- largely insensitive to stimuli other
ical activity may also exert modulatory than light, such as sleep and food intake.
effects on diurnal hormonal secretary Among the primary roles of melatonin in
patterns. mammals are the regulation of seasonal
However, for practical purposes, there is changes in reproductive activity in response
little reason to make a distinction between to changes in daylength and in retinal
RHY10 2/6/04 3:56 PM Page 233

NIGHTTIME MELATONIN PRODUCTION: A KEY HORMONAL OUTPUT FROM THE CLOCK 233

regulation of photopigment disk shedding, pineal gland there is a strong circadian


phagocytosis, and the inhibition of rhythm of melatonin synthesis. Whereas
retinal dopamine where melatonin is also serotonin levels are much lower at night
synthesized. than during the day, melatonin concentra-
tions display a reverse rhythm with highest
concentration at night associated with ele-
Synthesis of Melatonin
vated circulating levels of melatonin. The
The synthesis of melatonin begins with the link between these two reciprocal rhythms
N-acetylation of serotonin followed by the is the rate limiting enzyme for melatonin
addition of a methyl group at the 5-hydroxy synthesis, serotonin N-acetyltransferase
position via the enzyme hydroxyindole- (AANAT) (Fig. 10.1). This displays a
O-methyltransferase (HIOMT) (Fig. 10.1). diurnal rhythm of activity with levels at the
Melatonin mediates its biological effects by nighttime peaking up to 100 times higher
binding to high-affinity and low-affinity than in the daytime.The AANAT and mela-
receptors ML1 and ML2 belonging to the tonin rhythms derive from activation at
GPCR superfamily. The pars tuberalis (PT) night of the pineal’s sympathetic innerva-
of the pituitary shows the highest density tion in mammals. Norepinephrine binds
of melatonin receptors and apart from the to b-adrenoreceptors and thus stimulates
SCN is the only structure to be found con- adenylate cyclase activity. The resulting
sistently labeled in all mammalian studies increase in cAMP levels has been shown to
so far. Thus, these are considered the two stimulate AANAT transcription and trans-
major sites of melatonin action. In the lation and also maintains the enzyme in an

CH2CH(NH 2)COOH
Tryptophan
N
H
Tryptophan hydroxylase

5-Hydroxytryptophan
Aromatic amino acid
decarboxylase
Serotonin
N-Acetyltransferase
(AANAT)

N-Acetylserotonin
Hydroxyindole-O-methyltransferase
(HIOMT)
Melatonin
CH3O CH2 CH2 NHCOCH 3

N
H

Figure 10.1. The melatonin synthesis pathway.


RHY10 2/6/04 3:56 PM Page 234

234 HORMONAL RHYTHMS

active form. Alpha-1 adrenergic receptors part of the night, just preceding the decline
also participate in AANAT stimulation, of melatonin synthesis. Via binding to the
apparently by activating the phosphoinosi- CRE in the AANAT promoter and exert-
tide (PI) cycle and protein kinase A and ing a dampening effect, ICER modulates
C (PKA and PKC), which potentiates the rate and magnitude of melatonin induc-
b-receptor-induced cAMP production. tion in response to adrenergic signals. The
Targets of cAMP include a group of balance between the proportion of phos-
transcription factors that modulate the phorylated CREB and ICER protein levels
expression of cAMP responsive genes. determines the transcriptional activity of
These bZip factors constitute a family of the AANAT promoter. Thus the promoter
both activators and repressors that bind as cycles between activated and repressed
homo- and heterodimers to cAMP- states as a function of time. In this way,
responsive elements (CREs) and include AANAT mRNAs oscillates between high
CREB, CREM, and the CREM product nighttime and low basal daytime levels and
ICER. Nighttime adrenergic signals, determines the characteristic day–night
originating from the SCN, activate PKA oscillation of AANAT activity. This ensures
and thus phosphorylate CREB. Phosphory- rhythmic melatonin synthesis (Fig. 10.2).
lated CREB activates the P2 promoter of
the CREM gene and thus induces the
Functional Significance of the
expression of ICER. ICER downregulates
Melatonin Rhythm
its own expression constituting the CREM
feedback loop. Like AANAT, ICER This has been well established for a wide
mRNA expression displays diurnal rhyth- variety of seasonal vertebrates. Environ-
micity in the pineal gland. The peak of mental lighting, acting through the eyes in
ICER mRNA occurs during the second mammals and in part directly on the pineal

SCN
clock

CREB
PKA Activated
+
+
AANAT Promoter
Activated cycles between
activated a nd
repressed states
CREM
feedback
loop
- Repressed

Figure 10.2. The role of the CREM feedback loop in transducing a rhythmic clock-directed
signal into rhythmic hormone synthesis. Cartoon representation of the mechanism by which
melatonin rhythms are induced. Activated PKA phosphorylates CREB, which in turn activates
CREM and induces expression of ICER. ICER represses its own expression. The level of AANAT
mRNA is under positive and negative control by CREB and ICER, respectively.
RHY10 2/6/04 3:56 PM Page 235

CONTROL OF HORMONAL RHYTHMS BY THE HYPOTHALAMUS 235

in some birds, lizards, and fish, has a pro- regions (anterior, dorsalis, intermedia, and
found effect on the rhythm of melatonin posterior) or into three longitudinal zones
synthesis. The duration of melatonin secre- (periventricular zone, medial zone, and
tion is positively correlated with the length lateral zone). The hypothalamic nuclei con-
of the night period. It is through these stitute that part of the corticodiencephalic
changes in the duration of melatonin syn- mechanism that activates, controls, and
thesis that the brain is able to integrate integrates the peripheral autonomic
photoperiodic information. This relation- mechanisms, endocrine activity, and many
ship explains the present use of melatonin somatic functions, such as a general regula-
in farming to control seasonal function tion of water balance, body temperature,
(e.g., growth, reproduction, milk produc- sleep, and food intake, and the develop-
tion). The exact mechanism of action is ment of secondary sex characteristics.
unclear. The duration of nocturnal mela- The hypothalamus secretes vasopressin and
tonin production is the key signal, but the oxytocin, which are stored in the pituitary,
existence, within this signal, of a melatonin- as well as many releasing factors
driven circadian rhythm of sensitivity to (hypophysiotropic hormones), including
melatonin has been proposed to explain the thyrotropin releasing hormone (TRHs),
photoperiodic response. The melatonin gonadotropin releasing hormone (GnRH),
receptors involved most probably are of growth hormone releasing hormone
the mt1 subtype as the gene of the other (GHRH), Ghrelin, corticotropin releasing
subtype in mammals, mt2, is nonfunctional hormone (CRH), somatostatin, and
in two highly photoperiodic species, the dopamine. All these agents are released
Siberian and Syrian hamsters. The target into the blood and travel immediately to
sites mediating the control by melatonin of the anterior lobe of the pituitary, where
photoperiod-dependent seasonal functions they exert their effects (Fig. 10.3). All of
and, in particular, the annual sexual cycle them are released in an ultradian rhythm in
have not yet been clearly determined, pulsatile spurts. In fact, replacement
although the par tuberalis is suspected of hormone therapy with these hormones
being the target for melatonin seasonal does not work unless the replacements are
effects. There is growing evidence to indi- also given in a similar fashion.
cate that while the rhythm of melatonin
secretion is driven by the circadian pace-
Pituitary Gland
maker, this rhythm also feeds back on the
clock. Indeed, to a minor extent exogenous The pituitary gland is often portrayed as the
administration of melatonin has been “master gland” of the body. Such praise is
shown to resynchronize certain overt justified in the sense that the anterior and
rhythms, and the sleep–wake cycle in a posterior pituitary secrete a battery of
variety of conditions, including jet lag, and hormones that collectively influence all
freerunning rhythms in blind subjects. cells and affect virtually all physiologic
processes. The pituitary gland, also known
as the hypophysis, is a roundish organ that
䊏 CONTROL OF HORMONAL lies immediately beneath the hypothala-
RHYTHMS BY THE mus, resting in a depression of the base of
HYPOTHALAMUS the skull called the sella turcica (“Turkish
saddle”). In an adult human or sheep, the
The hypothalamus is located in the middle pituitary is roughly the size and shape of a
of the base of the brain, and encapsulates large garbanzo bean. Careful examination
the ventral portion of the third ventricle. of the pituitary gland reveals that it is
The hypothalamus may be divided into four composed of two distinctive parts: (1) the
RHY10 2/6/04 3:56 PM Page 236

236 HORMONAL RHYTHMS

Oxytocin Ghrelin Vasopressin


Hypothalamus
Somatostatin
Dopamine Inhibiting hormones

Releasing hormones
TRH
GnRH Anterior
GHRH pituitary
CRH

Systemic target
organs
Figure 10.3. Control of hormonal rhythms by the hypothalamus. Specific interactions
between the hypothalamus, pituitary, and target endocrine glands form a series of closed-
loop regulatory units that are the core of the endocrine system. The hypothalamic hormones
involved in this type of regulation are small, labile peptides that are delivered to the ante-
rior pituitary through a special vascular portal system and stimulate the release of anterior
pituitary hormones, which are delivered to target glands by the systemic circulation.

anterior pituitary (adenohypophysis), which systems must be in place to prevent over-


is composed predominantly of cells that or undersecretion of hypothalamic and
secrete protein hormones; and the posterior anterior pituitary hormones. A prominent
pituitary (neurohypophysis), which is not mechanism for control of the releasing and
really an organ but an extension of the inhibiting hormones is negative feedback.
hypothalamus, composed largely of the A key to understanding the endocrine
axons of hypothalamic neurons that extend relationship between hypothalamus and
downward as a large bundle behind the the anterior pituitary is to appreciate the
anterior pituitary. It also forms the so-called vascular connections between these organs.
pituitary stalk, which appears to suspend Secretion of hormones from the anterior
the anterior gland from the hypothalamus. pituitary is under strict control by hypo-
The pituitary gland may be king, but thalamic hormones. These hypothalamic
the power behind the throne is clearly hormones reach the anterior pituitary
the hypothalamus. Some of the neurons through the following route. A branch of
within the hypothalamus—neurosecretory the hypophyseal artery extends into a
neurons—secrete hormones that strictly capillary bed in the lower hypothalamus,
control secretion of hormones from the and hypothalamic hormones destined for
anterior pituitary. The hypothalamic hor- the anterior pituitary are secreted into that
mones are referred to as releasing hor- capillary blood. Blood from those capillar-
mones and inhibiting hormones, reflecting ies drains into hypothalamic–hypophyseal
their influence on anterior pituitary hor- portal veins. Portal veins are defined as
mones. Specific hypothalamic hormones veins between two capillary beds; the hypo-
bind to receptors on specific anterior pitu- thalamic–hypophyseal portal veins branch
itary cells, modulating the release of the again into another series of capillaries
hormone they produce. The anterior pitu- within the anterior pituitary. Capillaries
itary hormones enter the systemic circula- within the anterior pituitary, which carry
tion and bind to their receptors on other hormones secreted by that gland, coalesce
target organs. Clearly, robust control into veins that drain into the systemic
RHY10 2/6/04 3:56 PM Page 237

CONTROL OF HORMONAL RHYTHMS BY THE HYPOTHALAMUS 237

venous blood. Those veins also collect area is called the zona glomerulosa and is
capillary blood from the posterior pituitary associated with the production of miner-
gland. The utility of this unconventional alocorticoids. Next is the zona fasciculata,
vascular system is that minute quantities which, with the zona reticularis, produces
of hypothalamic hormones are carried in a glucocorticoids and androgens. The adrenal
concentrated form directly to their target steroid hormones are synthesized from
cells in the anterior pituitary, and are not cholesterol that is derived mostly from the
diluted out in the systemic circulation. plasma, but a small portion is synthesized in
situ from acetyl-CoA via mevalonate and
squalene.
Adrenal Glands
The adrenal glands secrete epinephrine,
Pituitary–Adrenal Secretion
norepinephrine, and dozens of different
steroid molecules. The two adrenal glands, Among all endocrine circadian rhythms
lie very near the kidneys. In mammals each known to occur in normal humans, the 24-
adrenal is actually a double gland, com- hour periodicity of pituitary–adrenal secre-
posed of an inner corelike medulla and tion has been studied most extensively. It
an outer barklike cortex. The medulla is is endogenous in nature, largely unaffected
responsible for secreting epinephrine and by short-term manipulations of the sleep–
norepinephrine, and the cortex synthesizes wake cycle, and it persists during complete
different steroid molecules, but only a few fast or continuous feeding. To cause a
of these have biological activity. These sort rapid shift in the rhythm, crucially timed
into three classes of hormones: glucocorti- exposure to bright light is necessary.
coids, mineralocorticoids, and androgens. Stimulation of the adrenal by adrenocorti-
These hormones initiate their action by cotropic hormone (ACTH) (Fig. 10.4)
activating members of the nuclear receptor or cAMP, activates an esterase that con-
superfamily. The glucocorticoids are 21- verts stored esterified cholesterol to preg-
carbon steroids with many actions, the most nenolone and starts the series of reactions
important of which is to promote gluco- that occur in either the mitochondria or
neogenesis in addition to being an essential endoplasmic reticulum.There is little, if any,
component of adaptation to severe stress. storage of steroid hormones within the
Cortisol is the predominent glucocorticoid adrenal cell, since these hormones are
in humans. Corticosterone is less abundant released into the plasma when they are
in humans but is the dominent glucocorti- made.
coid in rodents. The adult cortex has three Cortisol release occurs with a periodicity
distinct layers or zones. The subcapsular that is regulated by the diurnal rhythm of

Ser Tyr Ser Met Glu His Phe Arg Trp Gly Lys

Conserved region; required for full biologic activity Pro


Val
Ala Pro Tyr Val Lys Val Pro Arg Arg Lys Lys Gly
Gly Variable region; not required for biologic activity
Glu
Asp Asp Glu Ala Ser Glu Ala Phe Pro Leu Glu Phe

Figure 10.4. Structure of human ACTH.


RHY10 2/6/04 3:56 PM Page 238

238 HORMONAL RHYTHMS

ACTH release. ACTH has little control early morning maximum, declining levels
over secretion of aldosterone, the other throughout daytime, a quiescent period of
major steroid hormone from the adrenal minimal secretory activity centered around
cortex. Cortisol circulates in plasma in midnight, and an abrupt elevation during
protein-bound and free forms. The main late sleep. This circadian profile is produced
plasma binding protein is an a-globulin by modulation of the height of successive
called transcortin. ACTH is secreted from secretory pulses. The 24-hour rhythm of
the anterior pituitary in response to corti- adrenal secretion is primarily dependent on
cotropin releasing hormone (CRH) from the circadian pattern of ACTH release. The
the hypothalamus. CRH is a peptide of 41 rhythm in ACTH release results, in turn,
amino acids and is secreted in response to from periodic changes in the level of stimu-
many types of stress, which makes sense in lation by CRH. A circadian variation paral-
view of the “stress management” functions lel to that of cortisol has been demonstrated
of glucocorticoids. Corticotropin releasing for the plasma levels of adrenal steroids
hormone itself is inhibited by glucocorti- (Fig. 10.6). Pulses of plasma concentration
coids, making it part of a classical negative of adrenal steroids occur in remarkable syn-
feedback loop. Within the pituitary gland, chrony with bursts of cortisol secretion. A
ACTH is produced in a process that also distinct rhythm of serum cortisol levels
generates several other hormones. A large emerges at approximately 6 months of age.
precursor protein named proopiome- Once this periodicity is established, it per-
lanocortin (POMC) is synthesized and sists throughout adulthood. In humans the
proteolytically chopped into several frag- 24-hour profile of cortisol appears to be
ments as depicted (Fig. 10.5). Not all of the similar for both sexes. In females, oral con-
cleavages occur in all species and some traceptive therapy results in a large increase
occur only in the intermediate lobe of of the mean cortisol level and the amplitude
the pituitary. of the rhythm, resulting from estrogen-
Figure 10.6 shows 24-hour profiles of induced elevation of binding capacity of the
ACTH and cortisol typical of young adults. cortisol binding protein: transcortin.
Changes in plasma cortisol occur in parallel Disease states in which alterations of the
with those in ACTH.Their pattern shows an cortisol rhythm have been observed include

Proopiomelanocortin (POMC)

Amino-terminal fragment ACTH B-Lipotropin

Y-MSH A-MSH CLIP Y-Lipotropin B-Endorphin

B-MSH Met-enkephalin

Figure 10.5. POMC is synthesized and proteolytically chopped into several fragments.
RHY10 2/6/04 3:56 PM Page 239

CONTROL OF HORMONAL RHYTHMS BY THE HYPOTHALAMUS 239

ACTH Cortisol

08 12 16 20 00 04 08 08 12 16 20 00 04 08
Time (24 hours)

Figure 10.6. Mean 24-hour profiles of plasma ACTH and cortisol.

Growth Liver
Hormone

IGF-1

Fat

Indirect
Direct effect
effect

Figure 10.7. A 24-hour profile of plasma growth hormone (GH) and cartoon of GH effects.
In normal adult subjects, the 24-hour profile of GH levels consists of stable low levels abruptly
interrupted by bursts of secretion. The most reproducible pulse occurs shortly after sleep
onset, in association with the first phase of SW sleep.

primarily (1) disorders involving abnormal- itary. In normal adult subjects, the 24-hour
ities in binding and/or metabolism of corti- profile of plasma GH levels consists of
sol, (2) the various forms of Cushing’s stable low levels abruptly interrupted by
syndrome, and (3) affective illness (i.e., bursts of secretion (Fig. 10.7). The most
depression). reproducible pulse occurs shortly after
sleep onset, in association with the first
phase of slow-wave (SW) sleep. Other
Growth Hormone Release
pulses may occur in later sleep and during
Growth hormone release is affected by wakefulness, in the absence of any identifi-
sleep and circadian influences. GH is a able stimulus. In men, the sleep-onset GH
protein hormone of about 190 amino acids pulse is generally the largest and often the
that is synthesized and secreted by cells only pulse observed over the 24-hour span.
called somatotrophs in the anterior pitu- In women, daytime GH pulses are more
RHY10 2/6/04 3:56 PM Page 240

240 HORMONAL RHYTHMS

frequent, and the sleep-associated pulse, of hormones that serves to maintain blood
while still present in most cases, does not glucose within a normal range. GH is often
generally account for the majority of the said to have antiinsulin activity, because it
24-hour GH release. Circulating estradiol suppresses the abilities of insulin to stimu-
levels play an important role in determin- late uptake of glucose in peripheral tissues
ing overall levels of spontaneous GH secre- and enhance glucose synthesis in the liver.
tion. Sleep onset will elicit a pulse of GH Somewhat paradoxically, administration
secretion regardless of whether sleep is of GH stimulates insulin secretion, leading
advanced, delayed, interrupted, or frag- to hyperinsulinemia. GH has important
mented. While SW sleep is clearly a major effects on protein, lipid, and carbohydrate
determinant of the 24-hour profile of GH metabolism. In general, GH stimulates
secretion in humans, there is also evidence protein anabolism in many tissues. This
of a circadian modulation. Observation of effect reflects increased amino acid uptake,
the nocturnal profile of the GH pulse increased protein synthesis and decreased
reveals that it occurs within 1 hour of the oxidation of proteins. Indirect effects are
usual bedtime even in normal subjects sub- mediated primarily by insulinlike growth
jected to sleep delay. The total amount and factor 1 (IGF1), a hormone that is secreted
the temporal distribution of GH release is from the liver and other tissues in response
strongly dependent on age. Spontaneous to GH (Fig. 10.7). A majority of the growth
GH secretion is detectable in term infants, promoting effects of GH are actually due to
who appear to have a high level of tonic IGF1 acting on its target cells. Growth is a
secretion. As the infant matures, GH pulse very complex process, and requires the
frequency and pulse amplitude decrease, coordinated action of several hormones.
and tonic secretion diminishes. A pulsatile The major role of GH in stimulating body
pattern of GH release, with increased pulse growth is to stimulate the liver and other
amplitude during sleep, is present in prepu- tissues to secrete IGF1. IGF1 stimulates
bertal boys and girls. During puberty, the proliferation of chondrocytes (cartilage
amplitude of the pulses, but not the fre- cells), resulting in bone growth, and also
quency, is increased, particularly at night. appears to be the key player in muscle
Maximal overall GH concentrations are growth (Kato et al. 2001).
reached in early puberty in girls and in late
puberty in boys. Age-related decreases in
GHRH and Somatostatin
GH secretion have been well documented
in both men and women. This decline in The primary controllers of GH secretion
overall GH secretion appears to be are two hypothalamic hormones: growth
achieved by a decrease in amplitude rather hormone releasing hormone (GHRH)
than frequency of GH pulses. and somatostatin. Production of growth
hormone is modulated by many factors,
including stress, exercise, nutrition, sleep,
Growth Hormone Activity
and growth hormone itself. GHRH is a
GH activity has two distinct types of effects. mixture of two peptides, one containing 40
Direct effects are the result of GH binding amino acids; the other, 44. As the term indi-
its receptor on target cells. Fat cells cates, GHRH stimulates cells in the ante-
(adipocytes), for example, have GH recep- rior lobe of the pituitary to secrete growth
tors, and GH stimulates them to break hormone (GH). Somatostatin is produced
down triglyceride and suppresses their by several tissues in the body, including the
ability to take up and accumulate circulat- hypothalamus and is a mixture of two pep-
ing lipids (Fig. 10.7). GH is one of a battery tides, one of 14 amino acids, the other of 28.
RHY10 2/6/04 3:56 PM Page 241

CONTROL OF HORMONAL RHYTHMS BY THE HYPOTHALAMUS 241

It acts on the anterior lobe of the pituitary usually resulting from a tumor of
to inhibit the release of growth hormone somatotropes.
(GH) and of thyroid stimulating hor- 2. Acromegaly, which results from
mone (TSH) in response to GHRH and to excessive secretion of growth
other stimulatory factors such as low hormone in adults with a pulsatile
blood glucose concentration. Growth pattern superimposed over elevated
hormone secretion is also part of a basal levels. The onset of this disorder
negative-feedback loop involving IGF1. is typically insideous. Clinically, an
High blood levels of IGF1 lead to overgrowth of bone and connective
decreased secretion of growth hormone not tissue leads to a change in appearance
only by directly suppressing the lactotroph that might be described as having
but also by stimulating release of somato- “coarse features.”
statin from the hypothalamus. Growth
hormone also feeds back to inhibit GHRH The excessive growth hormone and IGF1
secretion and probably has a direct also lead to metabolic derangements,
(autocrine) inhibitory effect on secretion including glucose intolerance. Diurnal and
from the lactotroph. nocturnal episodes of GH secretion are
Abnormalities in the 24-hour profile of more frequent and of higher amplitude in
plasma GH have been reported in a variety adult subjects with hyperthyroidism, who
of metabolic, endocrine, neurological, and have an overall daily GH production rate
psychiatric conditions. There is an inverse fourfold above normal.
relationship between adiposity and GH
release, which results in a marked suppres-
Prolactin
sion of GH levels (reduction in both pulse
frequency and GH half-life) throughout the Prolactin, a single-chain protein hormone
24-hour span in obese subjects. States of closely related to GH, is secreted by the
both growth hormone deficiency and excess lactotrophs in the anterior pituitary. In
provide very visible testaments to the role addition to the lactotrophs, prolactin is also
of this hormone in normal physiology. Such synthesized and secreted by a broad range
disorders can reflect lesions in the hypo- of other cells in the body, most prominently
thalamus, the pituitary or in target cells. A various immune cells, the brain and the
deficiency state can result not only from a decidua of the pregnant uterus. The con-
deficiency in production of the hormone ventional view of prolactin is that its major
but also in the target cell’s response to the target organ is the mammary gland, and
hormone. Clinically, deficiency in growth stimulating mammary gland development
hormone or its receptor is manifested as and milk production pretty well defines its
growth retardation or dwarfism. The sever- functions. Such a picture is true as far as
ity depends on the age of onset of the dis- it goes, but it fails to convey an accurate
order and can result from either heritable depiction of this multifunctional hormone.
or acquired disease. The effect of excessive It is difficult to point to a tissue that
secretion of growth hormone is also very does not express prolactin receptors,
dependent on the age of onset and is seen and although the anterior pituitary is the
as two distinctive disorders: major source of prolactin, the hormone is
synthesized and secreted in many other
1. Giantism, which is the result of exces- tissues.
sive growth hormone secretion that Under normal conditions, the 24-hour
begins in young children or adoles- profile of prolactin follows a bimodal
cents. It is a very rare disorder, pattern, with minimal concentrations
RHY10 2/6/04 3:56 PM Page 242

242 HORMONAL RHYTHMS

around noon; an afternoon phase of slightly after giving birth. Prolactin, along with cor-
augmented secretion; and a major noctur- tisol and insulin, stimulates transcription of
nal elevation starting shortly after sleep the genes that encode milk proteins. The
onset and culminating around midsleep. critical role of prolactin in lactation has
In men there is an average increase of been confirmed in mice with targeted dele-
more than 250% above the minimum noon tions in the prolactin gene. Female mice
level. Pulses occur during daytime as well that are heterozygous for the deleted pro-
as during the night. Their number appears lactin gene (and produce roughly half the
to be a reproducible individual characteris- normal amount of prolactin) show failure
tic, ranging between 7 and 22 per 24-hour to lactate after their first pregnancy. Pro-
span. lactin also appears important in several
Studies of prolactin levels during nonlactational aspects of reproduction.
daytime naps or after shifts of the normal In some species (rodents, dogs, skunks),
sleep period have consistently demon- prolactin is necessary for maintenance
strated increased prolactin secretion associ- of corpora lutea (ovarian structures that
ated with sleep onset. Pharmacological secrete progesterone, the “hormone of
studies have tended to implicate dopamin- pregnancy”). Mice that are homozygous for
ergic as well as serotoninergic mechanisms an inactivated prolactin gene and thus
in this sleep-related elevation of secretion. incapable of secreting prolactin are infertile
While early reports described the 24-hour because of defects in ovulation, fertiliza-
rhythm of plasma prolactin as “entirely tion, preimplantation development, and
dependent on sleep,” the existence of a cir- implantation.
cadian component in the secretary pattern Finally, prolactin appears to have stimu-
of prolactin is now well recognized. Abrupt latory effects in some species on reproduc-
shifts of the sleep–wake cycle during real or tive or maternal behaviors such as nest
stimulated jet lag indeed reveal the exis- building and retrieval of scattered young.
tence of a sleep-independent secretory rise. The prolactin receptor is widely expressed
Under normal conditions, this circadian rise by immune cells, and some types of lym-
is synchronized with the early part of sleep, phocytes synthesize and secrete prolactin.
and both circadian and sleep effects are These observations suggest that prolactin
superimposed. Current data indicate that may act as an autocrine or paracrine mod-
the day–night difference in prolactin levels ulator of immune activity. Interestingly,
is present in newborns and persists into the mice with homozygous deletions of the
ninth decade. During pregnancy, serum pro- prolactin gene fail to show significant
lactin levels rise, but the 24-hour pattern of abnormalities in immune responses. A
secretion is maintained, albeit at a higher considerable amount of research is in
level. The nocturnal rise is also maintained progress to delineate the role of prolactin
throughout the postpartum period. in normal and pathologic immune
responses. At the current time (2003),
however, the significance of these potential
Mammary Gland Development, Milk
functions remains poorly understood.
Production, and Reproduction
Excessive secretion of prolactin—hyper-
Overall, several hundred different actions prolactinemia—is a relatively common
have been reported for prolactin in various disorder in humans. This condition has
species. Some of its major effects are sum- numerous causes, including prolactin
marized here. Prolactin induces lobuloalve- secreting tumors and therapy with certain
olar growth of the mammary gland and drugs. Common manifestations of hyper-
stimulates lactogenesis or milk production prolactinemia in women include amenor-
RHY10 2/6/04 3:56 PM Page 243

CONTROL OF HORMONAL RHYTHMS BY THE HYPOTHALAMUS 243

rhea (lack of menstrural cycles) and galac- logic effects of the gonadotrophins are
torrhea (excessive or spontaneous secre- known only in the ovaries and testes.
tion of milk). Men with hyperprolactinemia Together, they regulate many aspects of
typically show hypogonadism, with gonadal function in both males and females
decreased sex drive, decreased sperm (Fig. 10.8).
production, and impotence. In both sexes, LH stimulates secretion of
sex steroids from the gonads. In the testes,
LH binds to receptors on Leydig cells, stim-
Luteinizing and Follicle
ulating synthesis and secretion of testos-
Stimulating Hormones
terone. Theca cells in the ovary respond to
Luteinizing hormone (LH) and follicle- LH stimulation by secretion of estrogens.
stimulating hormone (FSH)—the gonad- In females, ovulation of mature follicles in
otropins—although not necessary for life, the ovary is induced by a large burst of
are essential for reproduction. These two LH secretion known as the preovulatory
hormones are secreted from cells in the LH surge. Residual cells within ovulated
anterior pituitary called gonadotrophs. follicles proliferate to form corpora lutea,
Most gonadotrophs secrete only LH or which secrete the steroid hormones prog-
FSH, but some appear to secrete both hor- esterone and estradiol. Progesterone is
mones. They are large glycoproteins com- necessary for maintenance of pregnancy,
posed of alpha and beta subunits. The alpha and, in most mammals, LH is required for
subunit is identical in both of these anterior continued development and function of
pituitary hormones, while the beta subunit corpora lutea. The name luteinizing
is unique and endows each hormone with hormone derives from this effect of induc-
the ability to bind its own receptor. Physio- ing luteinization of ovarian follicles.

Hypothalamus
GnRH

Pituitary
LH and FSH
- +
-
Testosterone Estrogen and
progesterone

Testes Ovaries

Figure 10.8. Physiologic effects of the gonadotrophins are known only in the ovaries and
testes. These hormones are responsible for gametogenesis and steroidogenesis in the gonads
and are all synthesized as prohormones, which are subject to posttranslational processing
within the cell to yield glycosylated proteins. Testosterone, estrogen, and progesterone
provide feedback control at the hypothalamus and pituitary through inhibition of GnRH, LH,
and FSH release and/or production.
RHY10 2/6/04 3:56 PM Page 244

244 HORMONAL RHYTHMS

As its name implies, FSH stimulates the become undetectable, although a number
maturation of ovarian follicles. Administra- of studies point to modest elevation of
tion of FSH to humans and animals induces nocturnal LH and FSH. A marked diurnal
“superovulation,” or development of more rhythm in circulating testosterone levels in
than the usual number of mature follicles young adults, with minimal levels in the late
and hence an increased number of mature evening and maximal levels in the early
gametes. FSH is also critical for sperm pro- morning, has been well demonstrated. With
duction. It supports the function of Sertoli a 15-minute sampling interval, 17–18 testos-
cells, which in turn support many aspects of terone pulses per 24-hour span can be
sperm cell maturation. detected. Since the 24-hour pattern of
peripheral LH concentration is relatively
inconsistent and of modest magnitude,
The Gonadotropic Axis
the robust circadian rhythm of plasma
Rhythms in the gonadotropic axis cover a testosterone may be partially controlled by
wide range of frequencies, from ultrafast other factors such as sleep-associated
oscillations of LH levels recurring at inter- variations in testicular blood flow, diurnal
vals of a few minutes, to episodic release in changes in Leydig cell response to
the hourly range, to diurnal rhythmicity, LH, and/or circadian fluctations in other
and finally, to monthly and seasonal cycles. hormones.
These various rhythms interact to provide In adult women, the 24-hour variation in
a coordinated temporal program governing plasma LH is markedly modulated by the
the development of the reproductive axis menstrual cycle. In the early follicular
and its operation at every stage of matura- phase, LH pulses are large and infrequent,
tion. Both LH and FSH are secreted in a and a slowing of the frequency of secretory
pulsatile pattern, and an augmentation of pulses occurs during sleep. In the midfollic-
this pulsatile activity is associated with ular phase, pulse amplitude is decreased,
sleep onset in a majority of boys and girls. pulse frequency is increased, and the fre-
In pubertal children the magnitude of the quency modulation of LH pulsatility by
nocturnal pulses of LH and FSH is consis- sleep is less apparent. Pulse amplitude
tently increased during sleep. As the pubes- increases again by the late follicular phase.
cent child enters adulthood, the daytime No modulation by sleep is apparent until
pulse amplitude increases as well, eliminat- the early luteal phase, when nocturnal
ing or diminishing the diurnal rhythm. slowing of pulsatility is again evident.
When the sleep–wake cycle is reversed in During the luteal–follicular transition,
pubertal boys, LH augmentation occurs there is four- to fivefold increase in LH
during the daytime sleep period, although pulse frequency, which accompanies the
there is also an elevation in LH concentra- selective FSH rise necessary for normal fol-
tions at the time when sleep occurred under liculogenesis. An interaction between the
basal conditions, indicating that although menstrual cycle and circadian rhythmicity is
the rhythm is sleep-dependent, there is an involved in the timing of the preovulatory
underlying inherent circadian component. LH surge. In normal women, onset of the
In pubertal boys, the nocturnal rise in LH surge occurs most often in late sleep or
testosterone coincides with the elevation of early morning. Moreover, a seasonal varia-
gonadotropins (Mitamura et al. 1999). tion of this circadian timing has been
Patterns of LH release in adult men reported, consisting of the appearance of a
exhibit large interindividual variability. The biphasic pattern of occurrence in spring
diurnal rhythm is dampened and may only. Toward menopause, gonadotropin
RHY10 2/6/04 3:56 PM Page 245

CONTROL OF HORMONAL RHYTHMS BY THE HYPOTHALAMUS 245

levels are elevated but show no consistent vated levels of gonadotropins per se have
circadian pattern. no biological effect.

Gonadotropin Releasing Hormone Regulation of Thyrotropin Hormones


The principal regulator of LH and FSH Increased Oxidative Metabolism
secretion is gonadotropin releasing
The most characteristic effect of thy-
hormone or GnRH (also known as LH
rotropin hormones (TH) is stimulation of
releasing hormone). GnRH is a ten amino
oxidative metabolism of most tissues of the
acid peptide that is synthesized and
body. The secretion of thyroid hormones
secreted from hypothalamic neurons and
(T3, T4 and calcitonin) is controlled by
binds to receptors on gonadotrophs (Fig.
thyrotropin stimulating hormone (TSH)
10.8). GnRH stimulates secretion of LH,
and thyrotropin releasing hormone
which in turn stimulates gonadal secretion
(TRH). TSH, also known as thyroid secret-
of the sex steroids testosterone, estrogen,
ing hormone is secreted from cells in
and progesterone. In a classical negative-
the anterior pituitary called the thy-
feedback loop, sex steroids inhibit secretion
rotrophs, finds its receptors on epithelial
of GnRH and also appear to have direct
cells in the thyroid gland, and stimulates
negative effects on gonadotrophs. This reg-
that gland to synthesize and release thy-
ulatory loop leads to a pulsatile secretion of
rotropic (thyroid) hormones (Fig. 10.9).
LH and, to a much lesser extent, FSH. A
TSH is a glycoprotein hormone composed
hyposecretion of GnRH may result from
of two subunits, which are noncovalently
intense physical training and anorexia
bound to one another. The alpha subunit of
nervosa.
TSH is also present in two other pituitary
Numerous hormones influence GnRH
glycoprotein hormones, follicle stimulating
secretion, and positive and negative control
hormone and luteinizing hormone, and in
over GnRH and gonadotropin secretion is
primates, in the placental hormone chori-
actually considerably more complex than
onic gonadotropin. Each of these hormones
depicted in Figure 10.8. For example, the
gonads secrete at least two additional hor-
mones—inhibin and activin—which selec- Hypothalamus
tively inhibit and activate FSH secretion GnRH
from the pituitary. Diminished secretion of
LH or FSH can result in failure of gonadal +
function (hypogonadism). This condition is -
typically manifest in males as failure in pro- Pituitary
duction of normal numbers of sperm. In TSH -
females, cessation of reproductive cycles is
commonly observed. Elevated blood levels +
of gonadotropins usually reflect lack of Thyroid hormones
steroid negative feedback. Removal of the
gonads from either males or females, as is
commonly done to animals, leads to per- Thyroid Gland
sistent elevation in LH and FSH. In
humans, excessive secretion of FSH and/or Figure 10.9. The thyrotropin hormone axis.
LH is most commonly the result of gonadal Feedback mechanisms that function within
failure or pituitary tumors. In general, ele- this axis are indicated.
RHY10 2/6/04 3:56 PM Page 246

246 HORMONAL RHYTHMS

and begin to increase in the late afternoon


b or early evening. Maximal levels occur
a b shortly before sleep. During sleep, TSH
a b
a levels generally decline slowly. A sharp
Thyroid- decrease occurs in the early morning hours.
Luteinizing Follicle-
stimulating Studies involving sleep deprivation and
hormone stimulating
hormone shifts of the sleep–wake cycle have consis-
hormone tently indicated that an inhibitory influence
Figure 10.10. Common structure of pitu- is exerted on TSH during sleep. The timing
itary glycoprotein hormones TSH, LH, and of the evening rise seems to be controlled
FSH. by circadian rhythmicity. The temporal
pattern of TSH secretion seems to reflect
both tonic and pulsatile release, with both
also has a unique beta subunit, which pro- the frequency and amplitude of the pulses
vides receptor specificity (Fig. 10.10). increasing during the nighttime. Pulses of
The most important controller of TSH secretion persist under both somato-
TSH secretion is TRH. TRH is secreted statin and dopamine treatment, suggesting
by hypothalamic neurons into hypothala- that the control of pulsatility is largely
mic–hypophyseal portal blood, finds its TRH-dependent. When the depth of sleep
receptors on thyrotrophs in the anterior is increased by prior sleep deprivation, the
pituitary, and stimulates secretion of TSH nocturnal TSH rise is markedly decreased,
and prolactin. One interesting aspect of suggesting that slow-wave sleep is probably
TRH is that it is only three amino acids the primary determinant of the sleep-
long. Its basic sequence is glutamic associated fall.
acid–histidine–proline, although both ends Studies measuring free T3 or the T3/T4
of the peptide are modified. Secretion of ratio have shown that changes in thyroid
TRH and hence TSH is inhibited by high hormone secretion occur in parallel with
blood levels of thyroid hormones in a clas- those of plasma TSH, but the amplitude
sical negative-feedback loop. Most verte- of this rhythm is very modest. However,
brates have two thyroid glands, located in under conditions of sleep deprivation, the
the neck; in humans the two have fused to increased amplitude of the TSH rhythm
form a single gland. Thyroxin (T4) is an results in an increased amplitude of the T3
amino acid containing four atoms of iodine. rhythm. The diurnal rhythmicity of plasma
Triiodothyronine (T3) contains only three TSH levels is not apparent until the second
atoms of iodine but is 3–5 times more active month of life but then seems to remain
than thyroxin, but is secreted in smaller stable throughout childhood, without
amounts. Calcitonin (another thyroid notable changes at the time of puberty.
hormone) is functionally unrelated to T3, is The nocturnal TSH surge is diminished
also secreted by the thyroid. Its chief role is or absent in hyperthyroidism, central
to prevent excessive concentration of hypothyroidism, and conditions of hyper-
calcium in the blood. cortisolism. In poorly controlled diabetic
states the surge also disappears.
The thyroid gland releases inactive thy-
Robust Circadian Rhythms
roxine (T4) into blood circulation, which is
All aspects of thyrotropic (thyroid) subsequently dionized to the active T3 form
hormone signaling display robust circadian by the liver enzyme deiodinase 1. Circadi-
rhythms. In normal adult men and women, anly regulated synthesis and release of T4
TSH levels are low throughout the daytime from the thyroid has long been considered
RHY10 2/6/04 3:56 PM Page 247

CONTROL OF HORMONAL RHYTHMS BY THE HYPOTHALAMUS 247

the underlying mechanism in maintaining a coined. In healthy humans, vasopressin has


daily rhythm in serum T3. In addition, we minimal pressor effects. The most impor-
have observed a rhythm in deiodinase 1 tant variable regulating vasopressin secre-
that further explains the observed rhythm tion is plasma osmolarity, or the
in serum T3. In addition, studies and our concentration of solutes in blood. Osmolar-
own observations have demonstrated that ity is sensed in the hypothalamus by
the thyroid hormone receptor, a member of neurons known as osmoreceptors, and those
the nuclear receptor superfamily and the neurons, in turn, simulate secretion from
intracellular receptor for T3 and T4, under- the neurons that produce vasopressin.
goes marked circadian variation at the gene Secretion of vasopressin is also simulated
expression level. by decreases in blood pressure and volume,
conditions sensed by stretch receptors in
the heart and large arteries. Changes in
Vasopressin
blood pressure and volume are not nearly
Vasopressin, which functions to conserve as sensitive a stimulator as increased osmo-
body water by reducing the output of urine, larity, but are nonetheless potent in severe
is a nine-amino-acid peptide secreted from conditions. For example, loss of 15 or 20%
the posterior pituitary (Fig. 10.11). of blood volume by hemorrhage results in
Within hypothalamic neurons, the hormone massive secretion of antidiuretic hormone.
is packaged in secretory vesicles with a Another potent stimulus of antidiuretic
carrier protein called neurophysin, and hormone is nausea and vomiting, both of
both are released on hormone secretion. which are controlled by regions in the brain
Vasopressin binds to receptors in the distal with links to the hypothalamus. The most
or collecting tubules of the kidney and pro- common vasopressin-related disease in
motes reabsorbtion of water back into the humans and animals is diabetes insipidus.
circulation. In its absence, the kidney This condition can arise from either of two
tubules are virtually impermeable to water, situations: (1) hypothalamic (“central”)
and it flows out as urine. In many species, diabetes insipidus, resulting from a defi-
high concentrations of vasopressin cause ciency in secretion of vasopressin from the
widespread constriction of arterioles, which posterior pituitary, caused by head trauma,
leads to increased arterial pressure. It was infections or tumors involving the hypo-
for this effect that the term vasopressin was thalamus, and other disorders; and (2)

Asn Cys Pro


Leu Gly
Gln s
Ile
s
Tyr Cys Oxytocin

Asn Cys Pro


Arg Gly
Gln s
Phe
s
Tyr Cys
Vasopressin
Figure 10.11. Amino acid sequence of oxytocin and vasopressin.
RHY10 2/6/04 3:56 PM Page 248

248 HORMONAL RHYTHMS

nephrogenic diabetes insipidus, which secrete oxytocin—what about its role in


occurs when the kidney is unable to males? Males synthesize oxytocin in the
respond to antidiuretic hormone. Most same regions of the hypothalamus as in
commonly, this results from some type of females, and also within the testes and
renal disease, but mutations in the arginine perhaps other reproductive tissues. Pulses
vasopressin (AVP) receptor gene or in the of oxytocin can be detected during ejacula-
gene encoding aquaporin-2 have also been tion. Current evidence suggests that oxy-
demonstrated in affected humans. tocin is involved in facilitating sperm
transport within the male reproductive
system and perhaps also in the female,
Oxytocin
because of its presence in seminal fluid. It
Oxytocin, a nine-amino-acid peptide syn- may also have effects on some aspects of
thesized in hypothalamic neurons, has male sexual behavior.
profound physiological effects. Oxytocin is
transported down axons of the posterior
Oxytocin and Vasopressin Secretions
pituitary for secretion into blood. It is also
during Sleep
secreted within the brain and from a few
other tissues, including the ovaries and The neurohypophysial hormones oxytocin
testes. Oxytocin differs from vasopressin in and vasopressin show daily rhythms of
two of the nine amino acids (Fig. 10.11). In secretion with elevated hormone release
years past, oxytocin had the reputation of during the hours of sleep in both rats and
being an “uncomplicated” hormone, with humans. The changes in plasma hormone
only a few well-defined activities related to concentrations in the rat are reflected by
birth and lactation. As has been the case changes in the posterior pituitary and hypo-
with so many hormones, further research thalamic hormone content. The amount in
has demonstrated many subtle but pro- the pituitary falls as the plasma concentra-
found influences of this little peptide. tion increases, while the hypothalamic
Nevertheless, it has been best studied in content is in phase with the plasma con-
females, where it clearly mediates three centration. This pattern can be modulated
major effects: stimulation of milk ejection, by ovarian steroids and alters with age. The
stimulation of uterine smooth-muscle con- pattern appears to be due in part to the noc-
traction at birth, and establishment of turnal increases in melatonin synthesis,
maternal behavior. While there is no doubt which stimulate hormone release in
that oxytocin stimulates all the effects humans but is inhibitory in rats. In women,
described above, doubt has been cast on plasma vasopressin concentrations and the
its necessity in parturition and maternal nocturnal peak are highest in the follicular
behavior. Mice that are unable to secrete phase of the menstrual cycle and attenu-
oxytocin because of targeted disruptions of ated in women on oral contraceptives.
the oxytocin gene will mate, deliver their
pups without apparent difficulty, and
display normal maternal behavior. 䊏 OTHER HORMONAL OR
However, they do show deficits in milk ejec-
HORMONE-DEPENDENT RHYTHMS
tion and have subtle derangements in social
behavior. It may be best to view oxytocin as
The Parathyroid Hormone
a major facilitator of parturition and mater-
nal behavior rather than a necessary com- The parathyroid hormone (PTH) helps to
ponent of these processes. Both sexes regulate the calcium-phosphate balance
RHY10 2/6/04 3:56 PM Page 249

OTHER HORMONAL OR HORMONE-DEPENDENT RHYTHMS 249

between the blood and the other tissues. and pulsatile PTH secretion are largely
The parathyroid glands in humans are small speculative. PTH is stored in cytoplasmic
pealike organs, usually four in number, granules before release. The most impor-
located on the surface of the thyroid. PTH tant physiological regulator of instanta-
increases the concentration of calcium in neous PTH secretion is ionized Ca2+. A
the blood (thus functioning as a calcitonin specific cell membrane receptor for Ca2+
antagonist) and decreases the concentra- has been identified on parathyroid
tion of phosphate, by acting on at least two cells. Activation of this receptor by acute
organs—the kidneys and the intestines— hypercalcemia instantaneously suppresses
and it stimulates the release of calcium PTH secretion, whereas hypocalcemia
into the blood from bone. The parathyroid results in an immediate increase of PTH
gland quadruplet was among the most release. Besides ionized calcium, active
recent endocrine organs for which a pul- vitamin D (calcitrol, 1a,25-dihyroxy-
satile secretion mode was demonstrated in vitamin D3) is an important regulator of
both animals and humans. It is now clear PTH secretion. Calcitrol reduces PTH
that the plasma concentrations of PTH fluc- availability by suppressing PTH transcrip-
tuate episodically. The pulses are of small tion. These effects are mediated both
amplitude and have rapid decay rates, directly via a specific vitamin D receptor
explaining why they escaped some early and indirectly by stimulation of intestinal
studies. PTH, which normally increases Ca2+ resorption.
during sleep, also has an endogenous circa-
dian component that persists in the absence
of sleep. The primary peak of the endoge-
Rhythms in Glucose Regulation
nous component occurs at approximately 8
pm, with a minimum around 10 am; the Robust variations in glucose regulation
amplitude and shape of the rhythm are dif- across the 24-hour cycle are now well
ferent under CR conditions than under demonstrated in both normal conditions
ambulatory conditions. and states of impaired glucose tolerance. A
The PTH receptor is coupled to both large number of studies have documented
adenylate cyclase and phospholipase C, reproducible changes in daytime and/or
thus stimulating both in parallel. PTH is an nighttime glucose utilization (both periph-
84-amino-acid peptide, and full biological eral and central), insulin secretion, and
activity resides in the amino terminal third insulin sensitivity in healthy subjects.
of the molecule. In humans PTH is appar- Plasma glucose responses to oral
ently secreted by two separate mechanisms. glucose, intravenous glucose, or meals are
Under normocalcemic conditions about markedly higher in the evening than in the
30% of total PTH secretion is released in morning. Diminished insulin sensitivity and
an episodic or pulsatile fashion with a mean decreased insulin secretion in relation to
frequency of 6–7 pulses per hour and a elevated glucose levels are both involved in
burst half-duration of approximately 2.5 causing reduced glucose tolerance later in
minutes. The average maximal secretion the day. It has been shown that glucose tol-
rate during spontaneous bursts is approxi- erance further deteriorates as the evening
mately 25% of the prevailing mean plasma progresses, reaching a minimum around the
level. The remaining 70% of total PTH middle of the night. This diurnal variation
secretion is attributable to tonic (time- is not caused by changes in activity level,
invariant) hormone release. At present the since it persists during continuous bedrest,
physiological mechanisms underlying tonic and is thus sleep-independent. Indeed sleep
RHY10 2/6/04 3:56 PM Page 250

250 HORMONAL RHYTHMS

deprivation studies indicate the existence Asynchronous Circadian Rhythms


of an intrinsic circadian modulation of
glucose regulation. Examination of correla- Asynchronous circadian rhythms within the
tions with the variations of the counterreg- vascular system may contribute to under-
ulatory hormones cortisol and GH indicate lying cardiovascular disease. Although
that the diurnal variation in insulin secre- day–night variations in heart rate and blood
tion is inversely related to the cortisol pressure have been described since the sev-
rhythm. Sleep-associated rises in glucose enteenth century and are the best-known
correlated with the amount of concomitant periodic functions in the cardiovascular
GH secreted. These studies show that system, other parameters have been shown
glucose regulation is markedly influenced to exhibit circadian variation as well,
by circadian rhythmicity and sleep and including stroke volume; cardiac output;
suggest that these effects could be partially blood volume; peripheral resistance;
mediated by cortisol and GH. Rhythms in parameters of ECG recordings; plasma
insulin secretion also have been found concentrations of pressor hormones such
outside the circadian range, demonstrating as noradrenaline, renin, angiotensin, and
that human insulin secretion is a complex aldosterone, atrial natriuretic hormone;
oscillatory process including rapid pulses plasma cAMP concentrations; blood vis-
recurring every 10–15 minutes superim- cosity; aggregability; and fibrinolytic activ-
posed on slower, ultradian oscillations with ity. Although molecular evidence of a
period in the range of 90–120 minutes. vascular clock was not extant, there is
These ultradian oscillations are tightly circumstantial evidence that it may exist.
coupled to glucose. For example, blood pressure undergoes
Alteration of the normal daily profiles of a marked circadian variability, which is
these parameters of glucose tolerance have increased in patients with hypertension and
been identified in normal aging, obesity, coincides with a temporal variability in the
and non-insulin- and insulin-dependent incidence of acute vascular events, such
diabetes. Observations have indicated that as myocardial infarction, sudden cardiac
the morning versus evening difference in death, and stroke.
glucose tolerance observed in normal sub- In normotensive people as well as in
jects is abolished in obese subjects and primary hypertension, there is, in general, a
reversed in diabetic subjects who have nightly drop in blood pressure, whereas in
instead higher glucose responses in secondary hypertension due to conditions
the morning than in the evening. Both the such as renal disease, gestation, and
rapid and ultradian oscillations of insulin Cushing’s disease, the rhythm in blood
secretion are perturbed in non-insulin- pressure is in ~70% of cases abolished or
dependent diabetes. While the ultimate even reversed, with highest values at night.
cause of these diurnal variations obviously This is of particular interest since the loss in
resides in the alteration of wake and sleep nocturnal blood pressure fall correlates
states and in the intrinsic effects of circa- with increased end organ damage in
dian rhythmicity, the primary factors cardiac, cerebral, vascular, and renal tissues.
responsible for transmitting these modula- Pathophysiological events within the car-
tory effects of CNS status are still poorly diovascular system also display a circadian
understood. variability. The onset of nonfatal or fatal
myocardial infarction predominates
around 6 am–12 noon. A similar circadian
time pattern has been shown for sudden
cardiac death, stroke, ventricular arrhyth-
RHY10 2/6/04 3:56 PM Page 251

OTHER HORMONAL OR HORMONE-DEPENDENT RHYTHMS 251

mias, and arterial embolism. These early- 3–9 hours relative to the oscillation in the
morning cardiovascular events coincide SCN. Clock gene oscillations are lost in
with the rapid rise in blood pressure, a rapid SCN-lesioned animals, suggesting that the
increase in sympathetic tone and in the con- peripheral oscillations may be driven or
centration of pressor hormones, and the synchronized by the SCN. It has been sug-
highest values in peripheral resistance.Thus gested that the SCN clock may synchronize
it appears that the early-morning hours are peripheral clocks via both neural and
the hours of highest cardiovascular risk. hormonal signals. In addition, more recent
Using radiotelemetry it is now possible observations suggest that feeding cycles
to study cardiovascular parameters in can entrain peripheral clocks independent
freely moving and undisturbed animals of light entrainment.
under freerun conditions. In these experi- Observations by our group and others
ments the rhythms in blood pressure, heart suggest that steroid hormones and vitamins
rate, and motor activity were studied in nor- may serve as candidate regulators of
motensive rats and transgenic hypertensive peripheral clocks. Circulating concentra-
rats during alternating light–dark cycles tions of some steroids undergo circadian
(L : D 12 : 12), during constant darkness variation, and examples of phase shifting
(DD) or during constant light (LL). In LD of circadian genes in peripheral organs
all parameters in both strains exhibited by glucocorticoids has been observed. In
significant and dominant circadian rhythms, addition, we reported a mechanism
which were maintained under freerunning whereby peripheral circadian oscillators
conditions (DD). During LL circadian may be regulated by humoral factors. This
rhythmicity was almost abolished in all involves a novel, ligand-dependent interac-
parameters and in both strains, while ultra- tion between the retinoid receptors, RXRa
dian components became more dominent. and RARa, and the bHLH-PAS circadian
Electrocoagulation of the SCN in normo- transcription factors, CLOCK and NPAS2,
and hypertensive abolished circadian which directly affects vascular clock func-
rhythms in blood pressure and heart rate. tion and is of potential importance in both
Interestingly, the hypertensive blood pres- vascular physiology and clinical vascular
sure values were not reduced by SCN events.
lesion, clearly indicating that the SCN is The mammalian circadian system is
involved in rhythm generation but not in organized such that self-sustained oscilla-
hypertension. These findings clearly indi- tors in the SCN entrain peripheral oscilla-
cate that the rhythms in blood pressure and tors by releasing a continuous stream of
heart rate must be under the central control rhythmic signals. The phase and amplitude
of the SCN. of peripheral clocks vary between different
tissues and organs, in addition to differing
from the phase and amplitude in the SCN.
Humoral Signals
The circadian orchestration of relevant
Humoral signals can phase-shift and reset targets in these tissues would synchronize
peripheral clocks. Molecular clocks similar the systems-level behavior and physiology
to those operating in SCN neurons have in accordance with daily changes in the
been uncovered in peripheral tissues and in environment. While in the unicellular algae
immortalized rat 1 fibroblast cell lines. In clock regulation of a large number of genes
peripheral tissues, such as the liver, kidney, within the same cell offers an adaptive
and heart, circadian rhythms in RNA abun- advantage to the organism, in higher organ-
dance are apparent for each mPer gene, isms, the clock has evolved to regulate sep-
although the phase of oscillation is delayed arate sets of genes in different tissue types
RHY10 2/6/04 3:56 PM Page 252

252 HORMONAL RHYTHMS

in a tissue-specific manner. This highlights FURTHER READING


the importance of the SCN as a master
oscillator, which functions to coordinate all Fisher, DA (1996): Physiological variations in
peripheral clocks. One function of a periph- thyroid hormones: Physiological and patho-
eral clock might be to regenerate a weak physiological considerations. Clin Chem
or dampened SCN signal, thus amplifying 42(1): 135–139.
the oscillation of the signal in that Forsling, ML (1993): Neurohypophysial hor-
peripheral tissue. A vascular clock may mones and circadian rhythm. Ann NY Acad
Sci 22(689): 382–395.
generate an amplified and synchronized
Foulkes, NS, Cermakian, N, Whitmore, D,
vascular rhythm in response to uncoordi- Sassone-Corsi, P (2000): Rhytmic transcrip-
nated bloodborne signals from central tion: The molecular basis of oscillatory mela-
and peripheral clocks. These signals may tonin synthesis. Novertis Found Symp 227:
include hemodynamic stress, the diurnal 5–18.
release of catecholamines and steroids or Goffin,V, Binart, N,Touraine, P, Kelly, PA (2002):
the periodic bioavailability of signals, such Prolactin: the new biology of an old hormone.
as vitamins, associated with feeding and Annu Rev Physiol 64: 47–67.
metabolic cycling. Granner, DK (1996): Pituitary and hypothalamic
The existence of multiple oscillators is a hormones Murray. In Harpers Biochemistry,
common characteristic of all circadian 24th ed., Granner, DK, Mayes, PA, Rodwell,
VW, eds, Appleton & Lange, Stamford, CT.
systems described thus far in multicellular
Kato, Y, Murakami, Y, Sohmiya, M, Nishiki, M
organisms. The clock regulates the initia- (2001): Regulation of human growth hor-
tion of locomotor activity in the SCN (and mone secretion and its disorders. Intern Med
concomitant feeding behaviors elsewhere 41(1): 7–13.
in the hypothalamus) while it is gearing up Lemmer, B (1999): Chronopharmacology and its
for nutrient metabolism in the liver. There impact on antihypertensive treatment. Acta
is a core set of circadian oscillators in the Physiol Pharmacol Bulg 24(3): 71–80.
SCN, liver, and aorta (and presumably all McNamara, P, Seo, SP, Rudic, RD, Sehgal, A,
tissue types); some of these are clock com- Chakravarti, D, FitzGerald, GA (2001): Reg-
ponents such as bmal, clock, npas2, the per ulation of CLOCK and MOP4 by nuclear
genes, and cry. Somehow, this core oscilla- hormone receptors in the vasculature: A
humoral mechanism to reset a peripheral
tor conscripts the tissue-specific transcrip-
clock. Cell 105(7): 877–889.
tional machinery to direct circadian Mitamura, R, Yano, K, Suzuki, N, Ito, Y, Makita,
oscillation of a subset of those genes—some Y, Okuno, A (1999): Diurnal rhythms of
of these are transcription factors that in luteinizing hormone, follicle-stimulating
turn regulate their targets in a tissue- hormone, and testosterone secretion before
specific fashion. Peripheral oscillators the onset of male puberty. J Clin Endocrinol
permit tuning of biological rhythms without Metab 84(1): 29–37.
difficulty to small, gradual changes in the Panda, S, Antoch, MP, Miller, BH, Su, AI,
phase of the input signal. Distorted envi- Schook, AB, Straume, M, Schultz, PG, Kay,
ronmental cues such as transatlantic air SA, Takahashi, JS, Hogenesch, JB (2002):
travel, the changeover from a day to a Coordinated transcription of key pathways in
the mouse by the circadian clock. Cell 109(3):
night work schedule, or a large hormonal
307–320.
release such as sympathoadrenal activation Reppert, SM, Weaver, DR (2002): Coordination
under conditions of severe stress, may of circadian timing in mammals. Nature
abolish the phase relationships between 418(6901): 935–941.
SCN and peripheral clocks, resulting Sassone-Corsi, P (1995): Transcription facroes
temporarily in a severely disorganized cir- responsive to camp. Annu Rev Cell Dev Biol
cadian system. 11: 355–377.
RHY10 2/6/04 3:56 PM Page 253

FURTHER READING 253

Schaefer, F (2000): Pulsatile parathyroid Witte, K, Lemmer, B (1996): Signal trans-


hormone secretion in health and disease. duction in animal models of normotension
Novartis Found Symp 227: 225–239. and hypertension. Ann NY Acad Sci 783:
Van Cauter, E (1990): Diurnal and ultradian 71–83.
rhythms in human endocrine function: A
minireview. Horm Res 34: 45–53.
RHY11 2/6/04 3:53 PM Page 255

11
HUMAN CIRCADIAN RHYTHMS
Hans P. A. Van Dongen, Gerard A. Kerkhof, and David F. Dinges

While the molecular biology of the circa- continues to provide clues that may apply
dian clock has been revealed in depth for to humans, there is a need for more
various animal species, research on humans research focusing on humans as well.
in this area is still in the early stages. Func- In this chapter, we discuss some successful
tional genetic sequences appear to be well approaches for laboratory-based research
preserved across species, suggesting that on human circadian rhythms, and the most
the molecular biology of human circadian profound human behavior interfering with
rhythms may be elucidated in the near this kind of research: voluntary, self-moti-
future. Humans have a full complement of vated control of sleep and waking activities.
anatomic (e.g., suprachiasmatic nuclei), We also look at morning-type and evening-
neuronal (e.g., retinohypothalamic tract), type individuals as phenotypes of circadian
and neuroendocrine (e.g., melatonin) variability in humans.
systems involved in circadian rhythm gen-
eration and entrainment, strongly suggest-
ing that the circadian biology in humans is 䊏 CIRCADIAN REGULATION AND
similar to that in animals. However, humans
OVERT CIRCADIAN RHYTHMS IN
express many behaviors for which no clear
equivalents are known in the rest of the
HUMANS
animal kingdom (e.g., use of clocks and
A Broad Spectrum of Circadian Rhythms
watches; application of artificial light; vol-
in Humans
untary night work; self-administration of
chronobiotic drugs), which may pose a chal- Human circadian rhythms have been
lenge to generalizing animal phenotypes to observed in a wide range of variables,
humans. Thus, while research on animals including aspects of behavior, physiology,

Molecular Biology of Circadian Rhythms. Edited by Amita Sehgal


ISBN 0-471-41824-2 Copyright © 2004 John Wiley & Sons, Inc.

255
RHY11 2/6/04 3:53 PM Page 256

256 HUMAN CIRCADIAN RHYTHMS

endocrinology, neurology, and metabolism. were largely preserved throughout evolu-


Circadian rhythms in humans can be tion, the information on other organisms
observed in both normal everyday life and given elsewhere in this book is likely to be
highly controlled laboratory experiments; relevant to humans.
and they are not only present in healthy
humans but also implicated in the symp-
Light: The Dominant Zeitgeber
toms of various diseases. Other chapters
for Humans
in this book are filled with examples of
circadian rhythms in a variety of animal There is overwhelming evidence that the
species—by and large, the same rhythms most important environmental zeitgeber
have been documented for humans as well. (i.e., circadian entrainment agent) for
Documentation of circadian rhythmicity in humans is the light–dark cycle. Until rela-
humans is often, for practical and ethical tively recently it was believed that only
reasons, limited to overt circadian rhythms bright light could entrain human circadian
that can be readily observed. The implica- rhythms, but it is now known that even ordi-
tions of this limitation are discussed below. nary indoor light can serve as a zeitgeber.
Table 11.1 gives an overview of the broad A phase-response curve and a dose-
spectrum of circadian rhythms documented response curve for humans have been
in humans thus far; this table is not established (see elsewhere in this book for
intended to be all-inclusive but serves to get in-depth discussions of response curves). In
a sense of the pervasiveness of circadian this age of artificial light, however, ambient
rhythmicity in humans. light of sufficient strength for entrainment
is available all day and beyond the period
of natural daylight. The available phase-
The Human Biological Clock Contained
and dose-response curves were assessed
in the SCN
using relatively brief exposures to light, and
It is generally believed that, in healthy for this reason cannot be applied to under-
humans, at least, the multitude of circadian stand entrainment in normal life. Because
rhythms is tightly coordinated to create human civilization tends towards a society
optimal relationships among the various active at all 24 hours of the day, animal
organs and physiological systems and the “wild type” behavior is not entirely repre-
environment at all times of the day. The sentative for human “wild type” behavior,
master biological clock responsible for and animal models are not sufficient to fully
this coordination is in the suprachiasmatic understand entrainment in humans. Much
nuclei (SCN) of the hypothalamus in the more work is needed to explain how
brain. The SCN send out signals to the rest entrainment by light, and possibly by other
of the brain and to peripheral oscillators zeitgebers, occurs from day to day in
and tissues in order to pass on or coordi- humans living under normal (i.e., nonlabo-
nate the body’s “internal” time of day. ratory) circumstances.
It is largely unknown what the mechanisms
for communication between the SCN and
Freerunning Rhythms in
the rest of the brain are; they may involve
Absence of Zeitgebers
neuronal firing, neurotransmitter release,
and/or humoral cues. Communication with Early cave experiments and later isolated
the rest of the body probably involves relay laboratory (“bunker”) studies have shown
nuclei elsewhere in the brain, but this, that humans, just like other animals, have
too, is mostly undocumented for humans. freerunning rhythms in the absence of
Nevertheless, because these mechanisms zeitgebers. The average period of circadian
RHY11 2/6/04 3:53 PM Page 257

CIRCADIAN REGULATION AND OVERT CIRCADIAN RHYTHMS IN HUMANS 257

䊏TABLE 11.1. Circadian Rhythms Observed in


Humansa
Category Example Variables
Cardiovascular system Heart rate
Circulating blood volume
Blood pressure
Respiration Rate of breathing
Airway resistance
Severity of asthma
Endocrine system Growth hormone
Cortisol
Norepinephrine
Gastrointestinal system Gastric acid secretion
Hunger
Excretion
Thermoregulation Core body temperature
Rate of sweating
Vasoconstrictor response
Immune system Antigen production
Production of cytokines
Efficacy of immunosuppressive drugs
Visual perception Retinal sensitivity
Pupillary reflex
Saccadic velocity
Brain function Cognitive performance
Melatonin secretion
Sensitivity to magnetic stimulation
Physical behavior Sleep–wake cycle
Athletic performance
Muscle strength
Disease Cancer progression
Occurrence of ischemic stroke
Efficacy of drug treatment
a
This table presents an overview of overt circadian rhythms documented for
humans, organized by arbitrary categories. Three variables are given as
examples for each category.This table is not intended to be complete; rather,
it is intended to give a sense of how pervasive circadian rhythms are in
humans. All variables in the right-hand column change markedly with a
(near-)24-hour periodicity. However, the circumstances under which these
periodic changes have been observed (e.g., laboratory- or field-based, with
or without sleep deprivation) vary greatly. The implications thereof are dis-
cussed in this chapter.

rhythmicity in the bunker experiments was Thus, feedback on the biological clock from
found to be 25 hours. A note of caution is in light sources at the disposal of the research
order for the interpretation of this value, subjects probably biased the estimate
however, as these experiments were not of the freerunning period in humans. Later
conducted under constant-dark conditions. laboratory experiments employing a differ-
RHY11 2/6/04 3:53 PM Page 258

258 HUMAN CIRCADIAN RHYTHMS

ent experimental paradigm, discussed in waking activity is discussed extensively in


some detail below, have revealed that this chapter.
the intrinsic period of the human biological
clock is much closer to 24 hours than to
25 hours. 䊏 ENDOGENOUS AND EXOGENOUS
DRIVES FOR CIRCADIAN RHYTHMS
Entrainment by Nonphotic Zeitgebers
Exogenous Drives of Circadian Rhythms
The cave and bunker experiments done
to induce freerunning circadian rhythms It is of interest to know the origin of the
raised the question as to what other stimuli, various circadian rhythms demonstrated to
besides light, would be capable of entrain- exist in humans. It is often implicitly
ing human circadian rhythms. In these assumed that all overt circadian rhythms
studies, steps were taken to remove a wide originate, through direct or indirect path-
range of potential zeitgebers, including 24- ways, in the biological clock located in the
hour cycles in ambient temperature, noise, SCN. Although this assumption is an over-
electromagnetism, availability of food, and simplification, it has lead to the study of
social interactions. There are some indica- “masking,” that is, the effect of exogenous
tions that meals may serve to entrain the influences that induce, enhance, reduce, or
biological clock, and social interactions otherwise alter the appearance of circadian
have long been suspected to serve as zeit- rhythmicity independently of the biological
gebers as well. Conclusive evidence for the clock. A useful working definition of
role of social zeitgebers will be difficult to masking in the context of chronobiology is
obtain, though, as social stimuli are difficult the appearance or modification of a circa-
to standardize for a well-controlled study. dian rhythm solely because of the presence
Social interaction is also one of the areas of an external influence. A good example is
where generalization from animal models gastrointestinal function. As food is nor-
is difficult, as animal social behavior may mally taken during the day and not during
differ substantially from human social the night, certain gastrointestinal organs
behavior. In addition, it may well be that (notably the stomach) are much more
the relative role of nonphotic zeitgebers in active during the day than during the night,
humans depends on the availability or creating the appearance of a circadian
absence of light for entrainment. rhythm. In the absence of food, or if food is
distributed evenly across a 24-hour period,
however, much of this rhythmicity disap-
Circadian Control of Sleep–Wake Cycle pears. Thus, the secretion of gastric acid, for
Perhaps the most profound circadian example, is driven primarily by exogenous
rhythm seen in humans—as in other animal factors (i.e., food intake) and not so much
species—is the (spontaneous) sleep–wake endogenously.
cycle. As we shall see later, this rhythm is
also quite difficult to deal with in humans,
Light as a Zeitgeber and
because of the unique voluntary control
Masking Factor
over sleep and waking activities that
humans have. Sleep in particular may or Many other circadian rhythms do not
may not influence circadian rhythmicity depend so heavily on external stimuli, and
directly, but it clearly limits exposure to persist in the absence of these exogenous
other zeitgebers. The role of sleep and influences. The circadian rhythm in mela-
RHY11 2/6/04 3:53 PM Page 259

ENDOGENOUS AND EXOGENOUS DRIVES FOR CIRCADIAN RHYTHMS 259

tonin secretion by the pineal gland exhibits zones, etc.), and the various neurophysio-
a regular (near-)24-hour pattern no matter logical systems cannot always adapt. Even
what happens in the environment. Even if though animal models are excellent tools
the biological clock is not entrained by any for understanding endogenous circadian
zeitgebers, this rhythm persists; thus, the cir- rhythms in humans, it is more difficult to
cadian rhythm in melatonin secretion is not use animal models to study exogenous cir-
a result of masking. It is known, though, cadian rhythms and rhythm disruptions
that bright-light exposure during the mela- that are the result of the interaction with
tonin secretory phase suppresses melatonin the habitat that humans have created for
secretion; therefore, light can act as a themselves. Differentiating endogenous
masking factor and alter or even eliminate from exogenous circadian rhythms helps us
the melatonin rhythm if exposure is appro- understand the human problems of desyn-
priately timed. It should be noted that chrony encountered during jet lag, shift
doing so typically involves keeping a person work, or illness.
awake in order to allow for light exposure
during the nocturnal period, which is when
Constant Routine and Circadian
melatonin is normally secreted. During
Rhythms
the day, another masking effect of bright-
light exposure can be observed. Bright-light The gold standard to distinguish endoge-
exposure causes an acute, short-lasting nous from exogenous drives of circadian
enhancement of alertness in humans (and rhythmicity is the laboratory experimental
other diurnal species), masking the under- protocol called constant routine. The objec-
lying circadian rhythm of alertness. Thus, tive of this paradigm is to eliminate or keep
light can function as a masking factor as constant all possible masking factors to
well as a zeitgeber. expose the underlying endogenous circa-
dian rhythmicity in variables of interest
(such as body temperature, heart rate, or
Importance of Endogenous and
cortisol secretion). In a constant-routine
Exogenous Factors in Circadian Function
paradigm, research subjects are studied in
It has been suggested that distinguishing an isolated laboratory for at least 24 hours
endogenous from exogenous sources of cir- to allow for the sampling of at least one full
cadian rhythms is artificial. After all, circadian cycle. Ambient light and temper-
humans interact with their environment all ature are kept constant, noise is eliminated,
the time, and the environment is an integral and social interactions are minimized. Sub-
component of human neurophysiology. jects’ posture is held constant, typically
Furthermore, the effects of masking factors near-supine. Food and drink is distributed
can be of the same order of magnitude as evenly over the experiment, usually as
the endogenous circadian rhythmicity. hourly isocaloric snacks (and not including
Along the same lines, therefore, it has been any caffeine or alcohol). In addition,
suggested that the effects of masking research subjects in a constant routine are
factors are just as important as the effects kept awake for at least 24 hours, to elimi-
of zeitgebers for the overall optimization of nate the effects of sleep on the variables
behavior with respect to the environment. measured. Figure 11.1 shows the endoge-
On the other hand, humans have an unpar- nous circadian rhythms of body tempera-
alleled capability to alter their environment ture, urinary cortisol, and heart rate
(artificial light, temperature control, high- recorded under constant routine (averages
speed transportation to different time for six individuals). The figure also shows
RHY11 2/6/04 3:53 PM Page 260

260 HUMAN CIRCADIAN RHYTHMS

(a) (b)

(c) (d)

Figure 11.1. Endogenous circadian rhythms. The figure shows endogenous temporal profiles
of core body temperature (upper left-hand panel), cortisol excretion (lower left-hand panel),
heart rate (upper right-hand panel), and mean arterial blood pressure (lower right-hand
panel), recorded during constant routine (averaged over n = 6 healthy subjects). Body tem-
perature (CBT) was measured every 2 minutes by means of a rectal probe [hourly averages
are plotted in panel (a)]; cortisol excretion (CORT) was derived from urine samples taken every
3 hours; heart rate (HR) and blood pressure (BP) were measured hourly with an oscillometric
monitor; and mean arterial pressure (MAP) was calculated as diastolic BP plus one-third of
the difference between diastolic and systolic BP. Endogenous circadian rhythmicity is clear in
CBT, CORT, and HR. There are fluctuations over time also in BP, which may or may not reflect
ultradian rhythms, but there is no endogenous circadian rhythmicity in BP.

the 24-hour profile recorded for blood pres- human physiology, endocrinology, neurol-
sure, which is essentially flat—using the ogy, immunology, and behavior.
constant routine paradigm, it has been
demonstrated that blood pressure does not
Voluntary Sleep–Wake Cycle Control
exhibit any endogenous circadian rhyth-
micity. It is important to recognize, though, The constant routine involves keeping indi-
that under ambulatory conditions, blood viduals awake, but the sleep–wake cycle is
pressure is not normally found to be con- also under circadian control. Thus, this
stant. There are other mechanisms that experimental paradigm exposes endoge-
affect blood pressure, which would be con- nous circadian rhythms while eliminating
sidered masking from a circadian perspec- another rhythm (i.e., that of sleep and wake-
tive, but that are no less important for fulness). In animal research, the sleep–wake
physiological function and health in cycle is often considered an integral part of
general. Thus, while circadian rhythms can circadian regulation and is even used as a
be studied in isolation to some extent, marker of circadian rhythmicity. In humans,
they must ultimately be considered in the however, this presents a problem due to the
context of the many other aspects of unique ability humans have to control their
RHY11 2/6/04 3:53 PM Page 261

THE CIRCADIAN AND HOMEOSTATIC REGULATION OF SLEEP 261

sleep times by self-motivation only. Humans waking activities, but this is actually an out-
can voluntarily stay awake for long periods standing question). The circadian control of
of time, during which time they can under- sleep, on the other hand, serves to place
take all kinds of activities, and they can wakefulness during the day and sleep
employ numerous environmental (e.g., tel- during the night. It is generally believed
evision) and pharmacological (e.g., coffee) that before the appearance of artificial
stimuli to help them do so. They can also light, climate control, and 24-hour avail-
plan to terminate their sleep at any time by ability of food, the circadian control of
using an alarm or by asking another person sleep ensured that waking activities and
to wake them up. The problem lies herein sleep took place at the times optimally
that sleep, or the absence of sleep, affects suited for survival (i.e., day and night,
many of the variables that show circadian respectively). Modern technology is often
rhythmicity. Thus, if sleep is displaced, then presumed to have eliminated the need for
the shape of a circadian rhythm can be circadian control of sleep, but the evolution
altered, or masked. This type of masking is of human physiology has not (yet?) been
not due to exogenous influences per se, and able to catch up with these new develop-
is therefore referred to as internal masking. ments. Thus, humans desire to stay awake at
The effect of internal masking is particu- times when their biological clock tells them
larly easy to show for body temperature, as they should sleep, and attempt to sleep
illustrated in Figure 11.2. The recognition of when the circadian drive for wakefulness is
internal masking has led to the practice of at a maximum. The simultaneous homeo-
not considering sleep an intrinsic compo- static control of wakefulness and sleep
nent of endogenous circadian rhythmicity further complicates this picture; for
in humans, even though by itself it is not an instance, staying awake at night involves
exogenous factor.To further understand the sleep loss, which results in an increased
special role of sleep, it is necessary to con- homeostatic drive for sleep during the day
sider the homeostatic mechanism that con- at the same time that the circadian drive for
trols sleep in addition to its circadian wakefulness is high. It is no wonder, then,
rhythm. that many nightshift workers experience
problems staying awake on the shift and/or
sleeping during the day, and some have
䊏 THE CIRCADIAN AND serious health problems involving gastro-
HOMEOSTATIC REGULATION intestinal, cardiovascular, and other patho-
logical conditions.
OF SLEEP

Homeostatic Drive for Sleep Exposing Circadian and Homeostatic


Regulation
Sleep is under the simultaneous control of
a circadian process (originating in the SCN) The complex interplay between the home-
and a homeostatic process (the neurophy- ostatic and circadian regulation of sleep
siological basis of which is as yet to be un- and wakefulness is well understood, and
raveled). The homeostatic control of sleep even captured in mathematical models
serves to balance sleep and wakefulness: (notably the two-process model of sleep
Longer periods of wakefulness are fol- regulation). Two experimental paradigms in
lowed by longer periods of sleep, and particular contributed substantially to our
shorter periods of wakefulness require less understanding of these processes: sleep
sleep for “recovery” (it is often implicitly deprivation and forced desynchrony. Sleep
assumed that sleep serves to recover from deprivation experiments typically involve
RHY11 2/6/04 3:53 PM Page 262

262 HUMAN CIRCADIAN RHYTHMS

CBT (°C)
CBT (°C)
CBT (°C)

Time of day

Figure 11.2. Masking of endogenous circadian rhythms by sleep. The figure shows typical
circadian profiles of core body temperature (CBT) in humans studied in a laboratory: normal
sleep–wake cycle (sleep from 22:00 until 06:00; top panel), displaced sleep–wake cycle (sleep
from 11:30 until 19:30; middle panel), and constant routine (no sleep; bottom panel). Sleep
periods are indicated with a black bar. The data illustrate that sleep–wake behavior, although
under circadian control, can mask endogenous circadian rhythmicity in other variables such
as CBT. Therefore, sleep is seldom considered an intrinsic component of endogenous circa-
dian rhythmicity in humans, but is treated as a separate neurophysiological system. The same
applies for the expression of (physical) activity during wakefulness. From studies of body tem-
perature profiles obtained under different experimental circumstances, mathematical “purifi-
cation” procedures have been developed to estimate the endogenous circadian profile of
body temperature in the presence of environmental and internal masking factors. Using
knowledge of sleep–wake times, activity patterns or light exposure, reasonably accurate esti-
mates of endogenous circadian rhythm parameters can be obtained with such purification
methods without the need for laboratory-based constant routine experiments. Note also that
even in the constant routine data of individual subjects (bottom panel), recorded under highly
controlled laboratory circumstances, there are fluctuations over time that cannot be
accounted for by circadian rhythmicity. These fluctuations probably reflect biological or meas-
urement noise, as they are largely averaged out when data for multiple subjects are pooled,
leading to a stereotypical sinusoidal profile of CBT circadian rhythmicity.
RHY11 2/6/04 3:53 PM Page 263

THE CIRCADIAN AND HOMEOSTATIC REGULATION OF SLEEP 263

keeping people awake for extended periods formance cannot be measured during sleep,
(usually one or two nights and days) and so sleep itself can just as well be a masking
subsequently giving them (unrestricted) factor. Two experimental paradigms have
recovery sleep. Forced desynchrony tackled this problem: the forced desyn-
involves scheduling wakefulness and sleep chrony paradigm and the ultradian
(and light and darkness) in cycles deviating sleep–wake cycle paradigm.
considerably from the 24-hour day. The bio-
logical clock is not capable of synchroniz-
Forced Desynchrony and Intrinsic Period
ing to the imposed cycles and resorts to a
of Circadian Pacemaker
freerunning state, effectively uncoupling
the sleep–wake cycle and the endogenous In the forced desynchrony laboratory para-
circadian cycle. Both experimental para- digm, subjects are exposed to an artificial
digms expose the interaction of the home- day–night cycle to which the biological
ostatic and circadian processes regulat- clock is unable to synchronize. Typically a
ing sleep, wakefulness, and other human 20-hour or a 28-hour artificial daylength is
behaviors. used. The forced desynchrony paradigm
uncouples the circadian and homeostatic
regulatory systems because the period of
the imposed light–dark cycle is outside the
Preserved Circadian Rhythms during
entrainment range of the biological clock.
Prolonged Total Sleep Deprivation
Instead, the biological clock reverts to
The interaction of circadian and homeosta- its intrinsic, freerunning periodicity. The
tic regulation during sleep deprivation is freerunning period of circadian rhythmicity
primarily seen in waking neurobehavioral under forced desynchrony is found to be
performance and other variables that track about 24.2 hours on average, which is
waking behavior (e.g., electroencephalo- substantially different from the 25 hours
graphic spectral power). Figure 11.3 shows observed in the classical freerun experi-
psychomotor vigilance performance across ments in isolated bunkers (discussed earlier
88 hours (3.7 days) of sustained wakeful- in this chapter). The difference is attributed
ness. For comparison, the 88-hour profile of to the feedback of (self-selected) light
melatonin concentration in blood plasma, exposure on the biological clock in the
which reflects circadian rhythmicity but not latter experiments, while this is believed not
homeostatic regulation, is shown as well. to contribute overall to the value for the
The same rhythmic pattern seen in mela- circadian period in forced desynchrony
tonin is also observed in psychomotor per- experiments. Thus, the 24.2-hour value of
formance. In addition, however, there is a the free-running circadian period found in
trend toward poorer performance as sleep forced desynchrony studies is thought to be
loss progresses. The net result is that per- a better reflection of the intrinsic period of
formance capability is reduced by extended the biological clock. However, in his early
sleep deprivation, but performance during studies of circadian rhythms, Aschoff
the day is still better than performance already noted that any estimate of circadian
during the previous night even after 3 period reflects, at least partially, the exper-
full days of sleep deprivation. Clearly, cir- imental or environmental circumstances.
cadian rhythms are preserved under total Thus, it may not be possible to give a uni-
sleep deprivation, but the effects of sleep versal estimate of the intrinsic period of
loss also serve to mask the endogenous human circadian rhythmicity without the
profile of circadian rhythmicity. On the connotation of the experimental conditions
other hand, waking neurobehavioral per- yielding the estimate.
RHY11 2/6/04 3:53 PM Page 264

264 HUMAN CIRCADIAN RHYTHMS

Figure 11.3. Circadian rhythms during total sleep deprivation. The figure shows the tempo-
ral profiles of psychomotor vigilance performance lapses (top panel) and plasma melatonin
concentration (bottom panel) across 88 hours of total sleep deprivation (for n = 10 subjects).
Melatonin concentrations are under circadian control only, while performance capability
shows the interaction of circadian rhythmicity and the buildup of homeostatic pressure for
sleep. Thus, sleep deprivation experiments serve to demonstrate the interaction of circadian
and homeostatic regulation for variables affected by both regulatory systems. However,
disentangling their respective contributions to the overall temporal profile presents a
mathematical problem.

Interaction of Circadian and the freerunning circadian rhythm shifts


Homeostatic Processes with respect to the imposed sleep–wake
(i.e., light–dark) cycle. By collapsing the
Besides its utility for estimating the intrin- data of interest over the free-running circa-
sic circadian period, the forced desyn- dian period, therefore, the cyclicity of sleep
chrony paradigm has another important and wakefulness is averaged out, resulting
use for the study of circadian rhythms—it in an average profile with no net effects of
makes it possible to average out the sleep and/or wakefulness. Similarly, by
“masking” influences of sleep and wakeful- folding the data over the imposed
ness. With each circadian cycle, the phase of light–dark cycle, circadian changes can be
RHY11 2/6/04 3:53 PM Page 265

THE CIRCADIAN AND HOMEOSTATIC REGULATION OF SLEEP 265

averaged out. It turns out that the same nous circadian rhythms in variables that are
variables that display an interaction of cir- affected by sleep and sleep loss and that
cadian and homeostatic influences under cannot, therefore, be measured under con-
total sleep deprivation also show this inter- stant routine. The interaction between
action under forced desynchrony. However, circadian and homeostatic regulation
by folding the data obtained under forced profoundly affects the ultradian day para-
desynchrony, the circadian and homeostatic digm, however. One-third of each sleep–
processes can be separated easily and wake cycle is reserved for sleep, but depend-
without the mathematical problems ing on the time of day, not all this time is
encountered for total sleep deprivation. actually spent asleep. Of course, the circa-
Interestingly, this led to the discovery that dian drive for wakefulness is the primary
the interaction between the circadian and factor responsible for this variation.To what
homeostatic mechanisms is nonlinear. This extent this phenomenon reduces the useful-
means that the contribution of the homeo- ness of the ultradian day paradigm for the
static process depends to some extent on study of circadian rhythms in variables also
the state of the circadian process, and vice affected by sleep has not been determined.
versa. (It should be noted that the conse-
quence of this discovery is that accurately
Jet Lag
separating the circadian and homeostatic
systems is not so simple after all in the The knowledge gained in sleep deprivation,
forced desynchrony context, as data folding forced desynchrony, and ultradian day
and averaging yields only a first-order studies about the combined circadian and
approximation of the relative contributions homeostatic influences on sleep, wakeful-
of the two processes.) This underlines the ness, and neurobehavioral performance
inherent interrelationship between circa- helps explain the phenomenon of jet lag.
dian rhythmicity and homeostatic regula- This is the malaise typically experienced
tion—they influence each other and after transmeridian travel (i.e., rapid travel
manipulating one process will have conse- to a different time zone) and caused by
quences for the other. circadian misalignment and desynchrony. It
takes some time (up to a week, depending
on the direction of travel) for the biologi-
Ultradian Day and Sleep Propensity
cal clock to synchronize with the light–dark
Another human experimental paradigm cycle in the new time zone, which causes the
that recognizes the importance of sleep– transient circadian misalignment. Circadian
wake regulation for the study of circadian rhythms that are (partially) driven by
rhythms exists, namely, the “ultradian day.” exogenous factors (in the new time zone)
This refers to a research paradigm using arti- appear to adjust more quickly, resulting in
ficial sleep–wake cycles that are extremely transient internal desynchrony. In addition,
short in duration—typically 20–90 a shift of the sleep–wake cycle is needed for
minutes—with about one-third of each cycle realignment to the light–dark cycle in the
reserved for sleep. Such schedules permit new time zone, leading to a transient dis-
the study of sleep and wakefulness at many turbance of the homeostatic regulation of
different circadian phases while, at least sleep and wakefulness. Jet lag is often
conceptually, leaving the overall 24-hour thought of as a purely circadian phenome-
homeostatic balance between sleep and non, but clearly the homeostatic process is
wakefulness intact.This is another paradigm involved as well, and contributes to the
that permits the measurement of endoge- symptoms of jet lag.
RHY11 2/6/04 3:53 PM Page 266

266 HUMAN CIRCADIAN RHYTHMS

Shiftwork countermeasures (e.g., caffeine) are avail-


able for the same purpose. From a chrono-
During shiftwork, the same mechanisms are
biological research point of view, these
at work to cause problems focusing on the
constitute masking factors. But in real life,
job, sleeping at unconventional times, and a
these “masking” factors are an intrinsic part
number of other work- and health-related
of how humans deal with, and control, the
problems. The involvement of homeostatic
cycle of day and night (see Fig. 11.4).
regulation is more readily apparent in this
case, as shiftwork, and nightwork in partic-
ular, often involves a period of acute sleep 䊏 MORNINGNESS/EVENINGNESS
loss. Furthermore, the circadian misalign- AND CIRCADIAN PHENOTYPES
ment may not be as transient as for jet lag,
for often there are no zeitgebers to help Differences in Circadian Phase Position
align the circadian rhythm with the of Biological Clock
work–rest schedule. On the other hand, it is
worth noting that problems of circadian One noteworthy illustration of human cir-
misalignment can be partially overcome, cadian rhythmicity is the existence of
even if no realignment takes place. The morning-type and evening-type individuals
temporary alerting effect of bright light, for (“larks” and “owls”). Morning types prefer
instance, may be used to compensate for a to be awake and active early during the day,
lack of circadian drive for wakefulness at which time they experience peak alert-
during the night. Various pharmacological ness, and they retire early in the evening.

Figure 11.4. Humans have voluntary, self-motivated control over sleep and wakefulness. The
development of intelligence and the use of tools has allowed the human species to step
beyond the influence of daily light–dark and other environmental controls of behavior. Para-
doxically, the rising of societies operating 24 hours a day, 7 days a week (i.e., 24/7) has led to
a lifestyle that is cognitively impairing, because circadian and homeostatic processes regulat-
ing sleep also appear to be critical for waking cognitive ability. The response to this dilemma
has been the invention and social acceptance of countermeasures serving to sustain wake-
fulness, alertness, and cognitive capability. As a result, human “wild type” behavior nowa-
days tends to ignore the endogenous regulatory neurobiology for sleep and waking activities.
(Artwork by Erik Timmers.)
RHY11 2/6/04 3:53 PM Page 267

MORNINGNESS/EVENINGNESS AND CIRCADIAN PHENOTYPES 267

Evening types, on the other hand, favor the


afternoon and evening for activities, as they

CBT (°C)
are more alert then, and they prefer to sleep
in the early morning. For a long time, it was
believed that morningness reflected an
aspect of personality, similar to introversion
and neuroticism. However, it was since
demonstrated that morning and evening
types differ in the phase of their circadian
rhythmicity. In other words, the biological
clock of evening types is delayed with

CBT (°C)
respect to morning types, as illustrated
in Figure 11.5. Moreover, this difference is
at least partially endogenous in nature.
The endogenous circadian rhythmicity of
(extreme) evening types, measured with
core body temperature under constant
routine, runs behind by approximately 2
Time of day
hours when compared to (extreme)
morning types. Oscillator theory would Figure 11.5. Core temperature rhythms
predict that morning types, therefore, in a morning-type individual and an evening-
would also have a somewhat shorter circa- type individual. The figure shows the
dian period than would evening-types difference between a randomly selected
morning-type individual (top panel) and a
under freerunning conditions, and some randomly selected evening-type individual
evidence to this extent has been docu- (bottom panel), in the circadian rhythm of
mented in the literature. It may also be that core body temperature (CBT) recorded under
morning and evening types differ in the field conditions. The arrows indicate the
entrainment properties of the biological minimum of CBT, the traditional phase
marker for CBT data, which is reached 1.8
clock, such that they synchronize differen- hours earlier (i.e., relatively phase-advanced)
tially to the natural light–dark cycle regard- in the morning-type than in the evening-
less of the period of their free-running type individual.
circadian rhythms.

Trait Variability in Circadian Rhythms


gotic twins have more similar scores on a
Although morningness/eveningness is morningness/eveningness questionnaire
partly affected by developmental changes than do dizygotic twins. In one twin study,
(puberty in humans is accompanied the total heritability for morningness/
by a tendency toward eveningness, and eveningness was found to be approximately
advanced aging is accompanied by a 45%. Further supporting a genetic basis for
tendency toward morningness), overall it morningness/eveningness is some preli-
appears to be an enduring trait. For this minary evidence that a polymorphism
reason, it has been suggested that morning- located in the 3¢ flanking region of the
ness/eveningness may be genetically deter- human Clock gene corresponds to a ten-
mined, and that morning types and evening dency toward eveningness. This discovery
types may be considered phenotypes of led to the development of a Clock mutant
human circadian variability. In support mouse model of morningness/eveningness.
of this idea comes the finding that monozy- Another polymorphism identified in the
RHY11 2/6/04 3:53 PM Page 268

268 HUMAN CIRCADIAN RHYTHMS

genomic machinery underlying human cir- light of earlier, comparable findings for
cadian rhythms, the A2634G substitution in mutations in rodents—strengthens the
the Timeless gene, appears to have no influ- notion that animal models can inform the
ence on morningness/eveningness. investigation of circadian rhythms in
humans. Given the strict practical and
ethical limitations of research involving
Advanced and Delayed
human beings, it is clear that animal models
Sleep Phase Syndromes
will continue to play an important role in
The tendency toward morningness or the investigation of the molecular biology
eveningness can be so extreme in an indi- of human circadian rhythmicity. When
vidual that the misalignment with the rest interpreting animal data in the context of
of society is disabling, resulting in pheno- humans (or vice versa), however, care must
types referred to as advanced sleep phase be taken also to consider the differences
syndrome (ASPS) or delayed sleep phase that exist between humans and other
syndrome (DSPS), respectively. Autosomal animal species. From a circadian perspec-
semidominant mutations in rodents with tive, the most important difference may
short or long circadian periods have been well be the human’s unique, voluntary
shown to be associated with similarly control over sleep and waking activities.
advanced or delayed sleep–wake rhythms. Circadian rhythms and sleep–wake pat-
These animal models predicted the exis- terns are strongly related in humans as
tence of familial circadian rhythm disorders they are in animals—but human behavior
in humans, a variation of which has been clearly shows that they can be easily
documented: familial advanced sleep phase dissociated. Thus, sleep–wake patterns
syndrome. In three families, ASPS segre- should not be indiscriminately used as a
gated as an autosomal dominant trait, and surrogate measure of circadian rhythms in
the disorder appeared to be caused by a humans.
missense mutation in the hPer2 gene, which
shortens the circadian period. Indeed, a
shorter circadian period would be expected FURTHER READING
to result in an earlier phase position under
Carskadon, MA, Dement,WC (1977): Sleepiness
entrained circumstances, resulting in the
and sleep state on a 90-min schedule. Psy-
extreme morning-type behavior of familial chophysiology 14: 127–133.
ASPS patients. The Mendelian inheritance Czeisler, CA, Khalsa, SBS (2000): The human
pattern of familial ASPS makes it unlikely, circadian timing system and sleep–wake
though, that the exact same genes are regulation. In Principles and Practice of Sleep
involved in this syndrome as in the morn- Medicine,3rd ed,Kryger,MH,Roth,T,Dement,
ingness/eveningness trait. DSPS has also WC, eds, Saunders, Philadelphia, pp. 353–
been associated with a genetic abnormality. 375.
In this case, a structural polymorphism Jones, CR, Campbell, SS, Zone, SE, Cooper, F,
in the hPer3 gene has been implicated in DeSano, A, Murphy, PJ, Jones, B, Czajkowski,
the pathogenesis of the disorder. L, Ptácek, LJ (1999): Familial advanced sleep-
phase syndrome: A short-period circadian
rhythm variant in humans. Nature Medicine 5:
1062–1065.
Kerkhof, GA, Van Dongen, HPA (1996):
䊏 CONCLUSION Morning-type and evening-type individuals
differ in the phase position of their endoge-
The documentation of human familial nous circadian oscillator. Neuroscience Lett.
advanced sleep phase syndrome—in the 218: 153–156.
RHY11 2/6/04 3:53 PM Page 269

FURTHER READING 269

Mrosovsky, N (1999): Masking: History, defini- Dement, WC, eds, Saunders, Philadelphia, pp.
tions, and measurement. Chronobiol. Int. 16: 391–399.
415–429. Vink,JM,Groot,AS,Kerkhof,GA,Boomsma,DI
Van Dongen, HPA, Dinges, DF (2000): Circa- (2001): Genetic analysis of morningness and
dian rhythms in fatigue, alertness, and eveningness. Chronobiol. Int. 18: 809–822.
performance. In Principles and Practice of Wever, RA (1979): The Circadian System of
Sleep Medicine, 3rd ed, Kryger, MH, Roth, T, Man, Springer-Verlag, New York.
RHYIndex 2/6/04 3:50 PM Page 271

INDEX

Abscisic acid (ABA), 200 Assays, 18


Acromegaly, 241 cell culture, 23
Actograms, 4, 34, 35 for circadian regulation, 27–29
ADAGIO (ADO) gene family, 196–197, 203–204, 208 Asynchronous circadian rhythms, 250–251
Adaptation, circadian rhythms as, 14–16 ATGER3 cell wall promoter, 200–201
Adaptive fitness, of plant circadian clocks, 208 Atmosphere, origin of, 144
Adenylate cyclase, 249 Autonomous clocks, in plants, 207–208
Adipocytes, 240 Autonomous oscillators, in insects, 218–221
Adiposity, 241 Autoregulatory loops, LHY and CCA1 in,
ado1/ado2 mutants, 196, 208 193–194
Adrenal glands, 237 Avian clock, molecular components of, 78–80
Adrenocorticotropic hormone (ACTH), 127–128, avp gene, 131
237–238
Advanced sleep phase syndrome (ASPS), 268 Bacteria, 143
Agamous-like 20 (AGL20) gene, 198, 203, 205 Basic loop–helix–loop (bHLH) domains, 41, 43, 47,
AHR protein, 125 48
AHRR protein, 125 Behavior, human circadian rhythms in, 257
Aldosterone, 238 Behavioral rhythm, clock control of, 15
1-Aminocyclopropane-1-carboxylic acid (ACC) Berkeley Drosophila Genome Project, 61, 68
oxidase, 199 bHLH PAS domain proteins, 124–126
Amphibians, frontal organ in, 76 Bioinformatics, 26
Anabaena, 146, 149 Biological clock
ANDANTE (AND) quantitative trait locus, 197 circadian phase position of, 266–267
Androgens, 237 human, 256
Animals, diurnal versus nocturnal, 7–8 Bioluminescence
Antidiuretic hormone (ADH), 130–131, 247, 248 arrhythmia of, 153
Aplysia, 215 assays, 148–149
Apoprotein, 187 circadian rhythm of, 223
APRR1/TOC1 family, 193 Bioluminescence resonance energy transfer (BRET),
Arabidopsis thaliana, 165, 186, 223 155
circadian clock in, 185 Birds. See also Avian clock
phytochromes of, 187–188 circadian system in, 76–82
TIM protein of, 198 multioscillatory system in, 76
Archaea, 166, 169 pineal gland in, 76–77
arg-13 mutation, 177 retina in, 78
Arginine vasopressin (AVP), 130–131, 248 suprachiasmatic nucleus in, 77
armadillo protein, 57 Blood pressure
ARNT protein, 125 asynchronous circadian rhythms in, 250–251
ARNT2 protein, 125 vasopressin and, 247
Arrhythmia Blue-green algae, 143. See also Cyanobacteria
of bioluminescence, 153 Blue light, mediation of Arabidopsis responses to,
mutations producing, 20 188–189
Asexual spores, production of, 171–172 Bmal1/Bmal2 genes, 78–80, 86, 87

271
RHYIndex 2/6/04 3:50 PM Page 272

272 INDEX

BMAL1/BMAL2 proteins, in feedback loops, Chloramphenicol acetyl transferase (CAT), 41


105–106 Chlorophyll A/B binding protein 2 (CAB2) gene,
BMAL1 knockout mice, 110 190
BMAL1 protein, 119, 125, 136, 191 Chloroplasts, 144
in mouse circadian rhythms, 110 chol-1 gene, 177–178
mammalian, 108–110 Chorionic gonadotropin, 245
Bmal genes, zebrafish clock and, 84–86 Chronobiology, terminology of, 13. See also
BMAL proteins, zebrafish, 86 Molecular chronobiology
Body temperature, circadian rhythms in, 213, cHsp90a protein, 82
214–215 Circadian amplifier, sasA gene as, 157–159
Brain Circadian clock-associated 1 (CCA1) gene, 201
circadian rhythms and, 215, 257 in autoregulatory loops, 193–194
in clock control, 10 Circadian-clock-controlled outputs, regulation by
Brain areas, SCN connection to, 127 oscillator mechanisms, 65–72
Bread mold, 171. See also Neurospora Circadian clocks. See also Multiple autonomous
Bulla, 215 clocks; Multiple oscillators; Oscillators;
bZip factors, 234 Peripheral clocks; Plant circadian clocks
in cyanobacteria, 143
CAB genes, 190, 192–193, 200, 201 endogenous, 4–5
Caenorhabditis elegans, 93 entrainment to input components, 7–9
Calcitonin, 246 evolution of, 169
Calcitrol, 249 freerunning, 6
Calcium/calmodulin-dependent kinase II (CaMKII), input/output of, 7
123 mechanisms of, 11–14
Calcium levels, circadian rhythms in, 199 multiple, 10–11
Calcium-phosphate balance, 248–249 phase shifting by photic pathways, 80–82
cAMP-responsive elements (CREs), 234 photic input pathways to, 97
Capillaries, pituitary and, 236–237 physiologic aspects of, 95–97
Carbohydrate storage, circadian control of, 147–148 resetting of, 5, 181
Carbonic anhydrase 2 (CAH2) gene, 201 role of cE4bp4 in, 83
Cardiovascular system temperature and, 181
asynchronous circadian rhythms in, 250–251 Circadian function, endogenous and exogenous
human circadian rhythms in, 257 factors in, 259
Casein kinase I, 53 Circadian genes
Casein kinase Id (CKId), 120 genetic screens and, 18–21, 28
Casein kinase Ie (CKIe), 106, 118–119 methods of isolating, 26–29
Catalase (CAT) genes, 200–201 molecular analysis of, 21–26, 28
cBmal1/cBmal2 genes, 80, 82 Circadian input kinase A (CikA), 164–165, 193
cCry1/cCry2 genes, 78–80 Circadian input kinase A (cikA) gene, 163, 164–165
cCRY2 cryptochrome, 77 Circadian input pathway, phytochrome–
cE4bp4 transcription factor, 82, 83, 91 cryptochrome interaction in, 190–191
cel-1 gene, 177–178 Circadian mutant strains, 168
Cell culture assays, 23 in Drosophila, 70–71
Cell culture systems, 21–22 Circadian oscillations, of period and timeless gene
Cell division products, 39–47
circadian clock and, 166 Circadian oscillator proteins, regulation by
in Synechococcus elongatus, 166, 167 phosphorylation, 52–57
Cellular redox state, 136–137 Circadian oscillators, 20
Central clocks, 10 core, 103–104
of Drosophila melanogaster, 219 Circadian pacemaker, forced desynchrony and
Central oscillator intrinsic period of, 263–264
feedback loop in, 105–106 Circadian periods, short, 116–117
mammalian, 94–95, 103–104 Circadian phase modifer A (cpmA) gene, 165–166
Chemosensory hairs, of Drosophila, 227 Circadian phase position, of biological clock,
Chenopodium, 198 266–267
Chicken, cycling clock genes in, 79, 83 Circadian phenotypes, morningness/eveningness and,
Chlamydomonas, 169, 207, 208 266–268
RHYIndex 2/6/04 3:50 PM Page 273

INDEX 273

Circadian photoreception, mammalian CRYs and, identification of, 201–202


115–116 promoter elements in, 182–183
Circadian photoreceptors, cryptochromes as, Clock function, plant genes affecting, 195–198
60–62 Clock genes, ix, 12–13, 23, 215–216. See also Clock
Circadian processes, interaction with homeostatic (clk) genes
processes, 264–265 in Drosophila, 216–217
Circadian regulation homology to, 26
assays for, 27–29 in vertebrates, 216–217
bioluminescence assays for, 148–149 Clock (clk) genes, 47, 48. See also Clock genes
human, 255–258 in birds, 78–80
in plants, 205–206 in Drosophila, 26
sasA disruption and, 159–163 in feedback loops, 105–106
Circadian rhythms, ix–x, 232. See also Asynchronous in mammals, 103, 106–108
circadian rhythms; Hormonal rhythms; Human in vertebrates, 217
circadian rhythms in Xenopus, 88–91
across species, 95 in zebrafish, 84–86
adaptive significance of, 14–16 Clock mechanisms, 11–14
analytical approaches to, 17–29 gene expression and, 13–14
basic attributes of, 3–11 modeling, 11–12
in cyanobacteria, 143–170 molecular level of, 12–13
defined, 5 Clock mutant mice, generating, 107
in Drosophila, 36–39, 57–65 Clock mutations, 33, 109
endogenous clock control of, 4–5 single-gene, 36
examples of, 3–4 CLOCK (CLK) protein, 125, 191. See also Clock
human, 231–232 proteins
in isolated organs, 215 BMAL1 protein and, 108–109, 110, 191
in levels of plant calcium, 199 Clock proteins. See also CLOCK (CLK) protein
mammalian, 93–138 circadian gene interaction with, 26–27
in Neurospora, 171–184 Drosophila, 34–36, 43
in nonmammalian vertebrates, 75–91 Kai, 153–159
properties of, 5–7 Clocks. See Circadian clocks
robust, 246–247 Clock transcription factor, 22
trait variability in, 267–268 Cloning, of tau gene, 117–118. See also Molecular
in Xenopus, 87–91 cloning
Circadian sleep regulation, 261–266 cl transgene, 98
Circadian systems, 5, 7 CO2 assimilation, plant circadian rhythms in,
in birds, 76–82 198–199
in mammals, 94–97, 221–222 Cockroaches, circadian rhythms in, 215
multiple oscillators in, 224 Coimmunoprecipitation assays, 24
in vertebrates, 221 Cold-circadian rhythm-RNA binding 2 (CCR2)
in zebrafish, 82–87 proteins, 191, 200, 206
Circadian time (CT), 6 Communication. See Multioscillatory communication
Circadian transcription, promoter elements for, Cones, 98
205–206 Conidia, production of, 171–172
cis elements, in circadian control, 150–151 “Consensus sites,” 28
CK2a protein, 206 CONSTANS (CO) gene, 202–205
CKB3 gene, 206 CONSTANS-like (COL1/COL2) genes, 203
CKII Neurospora mutant, 175 Constant routine, human circadian rhythms and,
Clock-associated genes, in plants, 194–198 259–260
Clock cells, CRY mediation of photoreception in, constitutive photomorphogenic 1 (COP1) mutant,
64–65 191–192, 203, 204
Clock cloning, PAS-containing protein in, 107–108 Core circadian clock, mammalian CRYs in, 116
Clock component mutations, disorders and, Core circadian oscillator, intracellular rhythms of,
137–138 103–104
Clock components, 13 Corticosterone, 237
Clock-controlled genes (CCGs), 127 SCN and, 127–128
genomic methods for yielding, 68 Corticotroph releasing factor (CRF), 128
RHYIndex 2/6/04 3:50 PM Page 274

274 INDEX

Corticotropin releasing hormone (CRH), 235, 238 Cytoplasm, PER protein in, 53–54
Cortisol, 237, 250 Cytoplasmic localization domain (CLD), 47
circadian rhythms in levels of, 214
release of, 237–239 D2 receptors, 88
cPer1/cPer2 genes, 80, 82 Day–night cycle, 5. See also Sleep–wake cycle
Crassulacean acid metabolism (CAM) idling, 199 dbp gene, 131
Crassulacean acid metabolism (CAM) plants, 191 DBT/PER association, 55
Creb mutant, 71 DBT protein, 53, 54
CREB (cAMP response element binding) protein, regulation of PER-driven feedback by, 55–57
cyclic activity of, 68–69, 99–101 dbt protein kinase, 58
CREB proteins, 234 dClock (dClk) mutant, 47–52, 70
CREM feedback loop, 234 Dead zone, in phase response curves, 179–180
CREM proteins, 234 DEC1/DEC2 proteins, 119–120
cryb mutation, in Drosophila, 61–62 deetiolated 1 (DET1) mutant, 191
CRY proteins. See also Mammalian CRYs Deinococcus, 165
in Arabidopsis, 188–189 Deiodinase 1, 246–247
in Arabidopsis phytochrome–cryptochrome Delayed sleep phase syndrome (DSPS), 268
interaction, 190–191 d-element binding protein (DBP), 131, 133, 227
light-dependent degradation of, 62–64 Desynchrony, forced, 261–263, 263–264
mediation of photoreception by, 64–65 Dfrizzled2 receptor, 57
TIM proteins and degradation of, 62–64 Diabetes, 247–248, 250
cryptochrome (cry) genes, 70 Diazotrophs, cyanobacteria as, 143–146
in Drosophila, 61–62, 225 Differential display experiments, 27–28
in feedback loops, 105–106 Dimerization, 114–115
in mammals, 98–99, 115–116 disconnected (disco) mutation, 37, 38, 71
in vertebrates, 80 Disease, human circadian rhythms in, 257
in Xenopus, 90 Disorders, clock component mutation and, 137–138
in zebrafish, 86–87 Diurnal animals, 7–8
Cryptochromes, 186 Diurnal rhythms, 5
as circadian photoreceptors, 60–62 DNA clones, 21
mammalian, 75 Dominant negative clock, 108–109
mediation of Arabidopsis responses to blue light Dopamine, 235
by, 188–189 prolactin and, 242
in zebrafish, 86–87 Dorsal medial nucleus of hypothalamus (DMH), 127
Cryptochrome signaling, 189 doubletime (dbt) mutant, 70
Cyanobacteria products of, 52, 53–55, 56–57
circadian rhythms in, 3, 143–170 Downstream genes, clock control of, 13–14
clocks in, ix, 11, 15–16 dreg genes, 65–66
fitness in, 166–168 Drosophila, 93. See also Drosophila melanogaster
gene expression in, 150–151 chemosensory hairs of, 227
as photoautotrophs and diazotrophs, 143–146 as chronobiological tool, 33–73
Cyanobacterial circadian clock circadian gene isolation from, 28–29
adaptive significance of, 166–169 circadian mutants in, 70–71
central component of, 155 clock control in, 13–14
control of cell division by, 166 clock genes in, ix, 12, 26, 216–217
kai gene cluster in, 151–152 clock mechanism models for, 12
molecular basis of, 148–163 clock proteins in, 43
Cyanobacterial circadian system, input/output entrainment of circadian rhythm by light in, 57–65
pathways of, 163–166 interlocked feedback loops in, 50–52
Cyanothece, carbohydrate storage by, 147 genetic analysis of, 72–73
Cycle-like factor (CLIF), 126 importance of clocks in, 15
cycle (Cyc) mutant, 47–52, 70 molecular clock model for, 67
Cyclic activity, tau gene and, 118–119 mutagenesis in, 18, 20, 21
Cyclic AMP (cAMP) production, 234 peripheral oscillators in, 10–11
Cyclic phosphorylation, 13 Drosophila Genome Project, 61, 68
Cycling clock genes, 79, 83 Drosophila melanogaster
in zebrafish, 85 central clock of, 219, 225
RHYIndex 2/6/04 3:50 PM Page 275

INDEX 275

circadian rhythm in, 33–73 in clocks, 11, 12–13


clock proteins in, 34–36, 216, 217 PER, CRY, CLOCK, and BMAL1 proteins in, 119
eclosion and locomotor activity in, 35 Feeding, restricted, 135–136
locomotor activity rhythms in, 36–38 fkf1 mutant, 196
rest–activity cycles of, 3–4 Flavin adenine dinucleotide (FAD), 179
Drosophila pseudoobscura, circadian rhythms of, 5, 6 FLAVIN-BINDING KELCH REPEAT F-BOX 1
dunce (dnc) mutant, 71, 72 (FKF1) gene, 196–197
Dwarfism, 241 Flavin mononucleotide (FMN), 148
flowering locus C (FLC) gene, 197–198
E4BP4 transcription factor, 133 flowering locus M (FLM) gene, 197
Early flowering 3 (ELF3) genes, 195 flowering locus T (FT) gene, 202, 203, 205
Earth’s rotation, circadian rhythms and, 4–5 Flowering time, plant genes affecting, 195–198
E box, 47–50, 73 Follicle-stimulating hormone (FSH), 243–244,
Ecdysone, 220–221 245–246
Eclosion, 5 Forced desynchrony, 261–263, 263–264
in Drosophila, 33–34, 35 Forward mutagenesis, 18–19
Electrophoretic mobility shift assays (EMSAs), 179 fragile X syndrome (dfmr1) mutant, 71
Electroretinogram (ERG), 83 Freerunning periods, variation in, 214
Endocrine function, mammalian, 96–97. See also Freerunning rhythms, 6
Hormones in the absence of zeitgebers, 256–258
Endocrine system during total sleep deprivation, 263, 264
circadian rhythms outside, 231–232 frequency (frq) genes, 172–173, 173–174, 184. See also
human circadian rhythms in, 257 frq entries
Endogenous circadian rhythms, 259, 260 molecular oscillations of, 176
Endogenous clocks photic resetting of Neurospora clock by, 178–179
as adaptation, 14–16 wc-1/wc-2 genes as regulators of, 174–175
control of circadian rhythms by, 4–5 Frogs, measuring rhythms in, 87–88
Endothelial PAS domain protein 1 (EPAS1), 126 Frontal organ, 76
ENHANCER OF SPLIT protein, 120 frq light response, mediation by WC-1
Entrainment, 20 photoreceptor, 179
defined, 6 frq mutants, 173
by light, 5, 7–9, 99–101 frq-null mutant, 176
of mammal circadian systems, 94, 95 metabolic defects and, 177–178
noncell-autonomous, 65 FRQ protein
nonparametric versus parametric, 8 post-translational regulation and, 173–174, 176–177
by nonphotic zeitgebers, 258 temperature regulation by, 180–182
Entrainment pathways, 7 Fyn kinase, 123
redundant, 66
ENU (N-ethyl-N-nitrosurea), 106 GAL4 transcription factor, 64–65
Enzyme oscillation, in plants, 199–200 g-aminobutyric acid (GABA), 104
Epidermal growth factor (EGF), 129 Gas exchange, plant circadian rhythms in, 198–199
Epidermal growth factor receptor (EGFR), 129 Gastrin releasing peptide (GRP), 123
Epinephrine, 237 Gastrointestinal system
9-cis-Epoxycarotenoid dioxygenase (NCED), 200 human circadian rhythms in, 257
Escherichia coli, 148, 155 masking and, 258
photolyase of, 189 Gated responses, 7
Estradiol, 240, 243 G-box factors (GBFs), 206
Eukaryotes, circadian rhythms of, 3, 143, 231–232 Gene expression
Eveningness, circadian phenotypes and, 266–268 assaying in mutant backgrounds, 24–25
Exogenous circadian rhythms, 259 circadian control of, 200–201
Exogenous drives, of circadian rhythms, 258 in cyanobacteria, 150–151
Extraretinal photoreceptors, 80–81 rhythms of, 13–14
transgenic clock, 25–26
Familial advanced sleep phase syndrome (FASPS) Gene promoters, 13
gene, 137–138 Genes, peripheral cycling by, 132–133
Feedback loops. See also Interlocked feedback loops Gene sequencing, 21
in the central oscillator, 105–106 Genetic screens, 17, 18–21
RHYIndex 2/6/04 3:50 PM Page 276

276 INDEX

Genetically directed representational difference hPER gene, 112


analysis (GDRGA), 118 hPer2 gene, 268
Geniculohypothalamic tract (GHT), 103 Human biological clock, 256
Genomic methods, for yielding clock-controlled Human circadian rhythms, 231–232, 255–268
genes, 68 constant routine and, 259–260
Germline assays, 18 endogenous and exogenous drives for, 258–261
Ghrelin, 235 spectrum of, 255–256
gi mutants, 196 Humans, morning and evening types of, 266–268
Giantism, 241 Humoral factors, 224
Gibberellic acid (GA) levels, 199–200 Humoral signals, 251–252
GIGANTEA (GI) gene, 195–196, 198, 206 Hydroxyindole-O-methyltransferase (HIOMT), 233
Global circadian control, 150–151 Hyperinsulinemia, 240
Glucocorticoids, 237 Hyperprolactinemia, 242–243
circadian gene expression and, 135 Hypertension, asynchronous circadian rhythms in,
Glucose regulation, rhythms in, 249–250 250–251
Glutathione-S-transferase (GST), 26–27, 78 Hyperthyroidism, 246
Glycogen synthase kinase (GSK), 106 Hypocotyl elongation, 198
Gonadotrophins, 243 Hypogonadism, 245
Gonadotrophs, 243 Hypophysiotropic hormones, 235
Gonadotropic axis, 244–245 Hypophysis, 235. See also Pituitary gland
Gonadotropin levels, 244–245 Hypothalamic diabetes insipidus, 247
Gonadotropin releasing hormone (GnRH), 235, Hypothalamic paraventicular nucleus (PVN), 127
245 Hypothalamus, 240, 241
Gonadotropins, SCN and, 128–129 anatomy of, 235
G proteins, 99 control of hormonal rhythms by, 235–248
pinopsin and, 81 in mammals, 221
Gq/11a protein, 81 pituitary and, 236
Growth hormone (GH) thyroid gland and, 245
activity of, 240 vasopressin and, 247
circadian rhythms in plasma levels of, 213, 214 Hypothyroidism, 246
release of, 239–240, 240–241 Hypoxia inducible factors (HIFs), 126
Growth hormone releasing hormone (GHRH), 235,
240–241 ICER protein, 234
Growth retardation, 241 Immune system, human circadian rhythms in, 257
Growth rhythms, plant, 198 Indol-3-acetic acid (IAA) levels, 198
GST fusion protein, 78 Inflorescence stem elongation, circadian rhythms in,
Gt1a protein, 81 200
Gynandromorph flies, 36–37, 38 Inhibiting hormones, 236
Input components, entrainment to, 7–9
H1F1a/H1F2a/H1F3a proteins, 125 Input/output pathways, of cyanobacterial circadian
HAIRY protein, 120 system, 163–166
Hepatic leukemia factor (hlf) gene, 133 Input pathways, 7, 20
Heterocysts, 146 Insects. See also Drosophila entries
Histidine kinase-related (HKR) domain, 187 autonomous oscillators in, 218–221
Homeostatic processes, interaction with circadian clock gene expression in, 216
processes, 264–265 Insulin, 240. See also Diabetes
Homeostatic sleep regulation, 261–266 in glucose regulation rhythms, 249–250
Hormonal rhythms, 231–252 Insulin-dependent diabetes, 250
hypothalamic control of, 235–248 Insulinlike growth factor 1 (IGF1), 240, 241
of nighttime melatonin production, 232–235 Intergeniculate leaflet (IGL), 97, 103
Hormone biosynthesis, in plants, 199–200 Interlocked feedback loops, in the Drosophila clock,
Hormone-dependent rhythms, 248–252 50–52. See also Feedback loops
Hormone release regulation, 127–129 Internal masking, 261
Hormones. See also Endocrine function Internal rhythmic desynchronization, 214–215
peripheral oscillators and, 11 Interphotoreceptor retinoid binding protein (IRBP),
released by hypothalamus, 235, 236–237 87
released by pituitary, 235–237 Intracellular clocks, 11
RHYIndex 2/6/04 3:50 PM Page 277

INDEX 277

Intracellular mammalian central oscillator, 103–104 Lipid mutations, 177–178


Intracellular rhythms, of the core circadian oscillator, Lizards, parietal eye of, 76
103–104 LKP2 gene, 197
Intrinsic period, of circadian pacemaker, 263–264 Locomotor activity
Ionophores, 199 of Drosophila, 35
Isolated organs, circadian rhythms in, 215 rhythm of, 95–96
of zebrafish, 82–84
Jet lag, 6, 80, 94, 265 Locomotor behavior regulation
Jrk mutation, 48 by PK2, 129–130
by TGFa, 129, 130
KaiA/KaiB/KaiC clock proteins, 153–159 Luciferase (luc), 23, 41, 42, 50, 60, 155
amino acid sequences of, 154 Luciferase reporter, 148, 149
interactions among, 155–157, 157–159 Luminance, 99
kaiC gene distribution, 156–157, 158 Luteinization, 243–244
kai gene cluster, in cyanobacterial clock, 151–152, Luteinizing hormone (LH), 128–129, 243–244,
153 245–246
kai gene expression, SasA protein and, 160–163 luxAB gene, 148, 150, 166
kai genes, 169 Lymphocytes, 242
deletion or inactivation of, 153–154
distribution of, 156–157 Malpighian tubules, 216, 217, 219–220, 224, 225, 226
mutations mapped to (table), 152 Mammalian central oscillator
Kasalath allele, 206 intracellular, 103–104
Knockout mice, 110, 112–114, 115–116 in the SCN, 103
Knockouts, multiple, 114 Mammalian circadian rhythms, serotonin in, 123
Mammalian circadian system, properties of, 94–97
Lactotrophs, 241 Mammalian clocks
lark mutant, 69–72 alternative components of, 119–124
LARK protein, 69–72 light and, 97–103
“Larks,” 266–267 Mammalian core clock, cryptochromes in, 115–116
Late elongated hypocotyl (LHY), in autoregulatory Mammalian CRYs
loops, 193–194 circadian photoreception and, 115–116
Lateral neurons (Lnvs), 219, 227 in the core circadian clock, 116
Leucine zippers Mammalian per homologs, 110–115
E4BP4 as, 133 Mammals
vrille gene as, 66 chronobiological studies of, 33
LH releasing hormone, 245. See also Gonadotropin circadian clocks of, ix, 10, 11, 221–222
releasing hormone (GnRH) circadian responses to light by, 99–101
Light circadian rhythms of, 93–138
central and peripheral clocks and, 10 clock genes of, 12, 106–108
circadian response to, 5–6, 7–9, 99–101 cryptochromes in, 75
cyanobacteria and, 147–148 endocrine function in, 96–97
detection by the retina, 97–98 humoral signaling in, 251–252
as the dominant human zeitgeber, 256 importance of clocks to, 16
effects on melatonin release, 81 novel photopigments of, 98–99
mammalian clocks and, 97–103 pineal gland of, 76
response of Neurospora clock to, 178–182 reproduction in, 97
as zeitgeber and masking factor, 258–259 rest–activity rhythms in, 95–96
Light–dark (LD) cycles, 6 sleep–wake cycles in, 96
Light-dependent degradation, of TIM and CRY temperature in, 96
proteins, 62–64 Mammary gland, development of, 241, 242–243
Light harvesting complex (LHC) gene family, 205 Masking, 8, 258–259
Light harvesting complex (LHCB) proteins, 191, 205 internal, 261
Light-induced TIM degradation, 57–60 by sleep, 262
Light pulses, circadian clock entrainment to, 8–9 Master–slave oscillator relationship, 132
Light-response elements (LREs), in Neurospora, mCry1/mCry2 genes, 115–116
179–180 Medial SCN (mSCN), 77
Light signaling, in plants, 191–192 Melanopsin, 99
RHYIndex 2/6/04 3:50 PM Page 278

278 INDEX

Melatonin, 225 Multioscillatory communication, interspecies


during total sleep deprivation, 264 variation in, 222–225
masking and, 258–259 Multioscillatory systems
nighttime production of, 232–235 in birds, 76
pineal gland and, 76 molecular evidence for, 215–217
SCN and, 127 organization of, 217–227
Melatonin release, effects of light on, 81 physiological evidence for, 213–215
Melatonin rhythm, significance of, 234–235 Multiple autonomous clocks, plant, 207–208
Melatonin synthesis, 233–234 Multiple oscillators, 252
in zebrafish, 82–84 relationship among, 224
Menopause, 244–245 Mutagenesis screens, random, 18
Metabolic defects, frq-null gene and, 177–178 Mutant backgrounds, assaying expression in, 24–25
Microarrays, 28–29, 120–121, 134 Mutant phenotypes, 18–21
Microchip technology, 227–228 Mutations. See also Circadian mutants
Microcystis, 145 genetically engineered, 25–26
Milk production, 241, 242–243 mapped to kai genes, 152
Mimosa pudica producing arrhythmia, 20
circadian leaf movement of, 198
circadian rhythms in, 4, 186 N-acetyltransferase (AANAT, NAT), 88, 233–234
Mineralocorticoids, 237 NADPH-protochlorophyllide oxidoreductase gene,
Mitogen-activated protein kinase (MAPK), 81–82, 200
99–101 ndhD gene, 165
Molecular analysis Nephrogenic diabetes insipidus, 247–248
of circadian genes, 21–26 N-ethyl-N-nitrosurea (ENU), 106
of nonmammalian vertebrate circadian rhythms, Neural cell adhesion molecule (NCAM), 104
75–91 Neuroanatomic pathways, SCN hormone release
Molecular chronobiology, Drosophila in, 33–73 regulation via, 127–129
Molecular clocks Neurofibromatosis-1 (Nf1) mutant, 71, 72
component variation in 225, 227 Neuropeptide Y, 103
in Drosophila, 33–73 Neurophysin, 247
gene expression and, 13–14 Neurospora. See also Neurospora crassa
Molecular cloning, 38–39 CCGs in, 182–183
Molecular oscillators, function of, 65–66 circadian rhythms in, 171–184
Molecular screens, 28 clock genes in, ix
Mollusks, circadian rhythms in, 215 Neurospora clock, 177
Mop3 knockout mice models for, 12
activity record of, 111 molecular basis of, 172–178
MOP9 protein, 125 response to light and temperature, 178–182
function in peripheral tissues, 126 outputs controlled by, 182–183
MOP (member of PAS) superfamily, 124–126 Neurospora crassa, as genetic system, 171
Morningness, circadian phenotypes and, 266–268 Nicotinamide adenine dinucleotide (NADH),
Mouse. See also Knockout mice 136–137
activity rhythms of, 96, 100 nightblindness b (nbb) mutation, 84
BMAL1 protein in, 110 9-cis-epoxycarotenoid dioxygenase (NCED), 200
Clock gene in, 106–108 Nipponbare allele, 206
in QTL analysis, 121–122 Nitric oxide, 100–101
mPer1/mPer2/mPer3 genes, 108, 109, 110, 112–116, Nitrogenase, 146
118 Nitrogen fixation, in cyanobacteria, 146
m-rigui gene, 111, 112 N-methyl-d-aspartate (NMDA), 99
mRNA Nocturnal animals, 7–8
clock genes and, 12–13 Nocturnin (noc) gene, 91
from per gene, 39–40 Noncell-autonomous entrainment, 65
mTim gene, 124 Noninsulin-dependent diabetes, 250
Multifaceted synchronization, of SCN neurons, Nonmammalian vertebrates, circadian rhythms in,
104 75–91
Multioscillatory circadian organization, in Nonparametric entrainment, 8
vertebrates, 221 Nonphotic zeitgebers, 258
RHYIndex 2/6/04 3:50 PM Page 279

INDEX 279

Norepinephrine, 237 PER homologs, mammalian, 110–115


Nostoc, 146 period (per) allele, 20, 21, 25, 26. See also period (per)
NPAS1 protein, 125 genes
NPAS2 protein, 124–126, 136, 191 in Drosophila, 36–61, 66–68, 70, 72–73
NPAS3 protein, 125 mutants of, 36–38, 39–41, 70
Nuclei, PER protein in, 54–55, 55–57 in zebrafish, 86
Null alleles, experiments using, 178 period (per) gene products, circadian oscillations of,
Null mutations, 20. See also frq-null mutant; sasA 39–47
null background period (per) genes. See also period (per) allele
in Drosophila, 216–217
Obesity, 241 in feedback loops, 105–106
Opaque 2 (O2) gene, 206 in mammals, 75, 99, 101, 111
Opsin, 98 in vertebrates, 217, 225
Organisms, internal rhythmic desynchronization in, period extender (pex) gene, 163
214–215 Peripheral circadian oscillators, 251–252
Organs Peripheral clocks, 10. See also Peripheral oscillators
circadian rhythms in, 215 role in physiology, 227–228
clock-containing, 131–132 Peripheral oscillators, 10–11, 131–137, 224
Oscillatoria, 147 in mammals, 222
Oscillator mechanisms, circadian-clock-controlled restricted feeding and, 135–136
output regulation by, 65–72 Peripheral tissues, MOP9 function in, 126
Oscillator proteins, circadian, 52–57 per-luc reporter gene, 216, 218, 219–220
Oscillators. See also Autonomous oscillators; per mRNA, 39–40
Multioscillatory systems; Peripheral oscillators oscillations in, 42
24-hour, 12 Peroxisome proliferator-activated receptor (PPAR),
revealed by tissue culture, 134 135
Osmoreceptors, 247 per promoter, 60
Output pathways, 7, 20, 126–131 PER protein, 24, 216, 217
Output rhythms, of plants, 198–205 in cytoplasm, 53–54
“Owls,” 266–267 in nuclei, 54–55, 55–57
Oxidative metabolism, increase in, 245–246 TIM protein collaboration with, 44–47
Oxytocin, 235, 247, 248 versus per mRNA, 39–40
PER reporter, temporal expression of, 220
Parametric entrainment, 8 PER/TIM–dCLK/CYC binding, 50, 51
Parathyroid hormone (PTH), 248–249 PER-TIM protein complex, in cyanobacteria, 156
Paraventricular nucleus of thalamus (PVT), 127 PER transcription, 40–41
PAR domain, 131 Pfr phytochromes, 187
Parietal eye, 76 Pharbitis nil, 201, 202
PAR proteins, 133 Phase, 6
Pars tuberalis (PT), 233 entrainment and, 8–9
PAS-containing protein, in Clock cloning, 107–108 Phaseolus coccineus, 207
PAS domain proteins Phaseolus vulgaris, 207
bHLH, 124–126 Phase-response curve (PRC), 9
known (table), 125 Phase shift, 6, 80
PAS domains, 21, 41, 43, 44, 47, 108–109, 114, 187, 208 by photic pathways, 80–82
PASKIN protein, 125 Phenotypes, mutant, 18–21
Pathways, in circadian systems, 7 Phosphoenolpyruvate carboxylase (PEPC), 206
pdf mutant, 69, 71, 72 Phosphoenolpyruvate carboxylase (PEPC) kinase,
Per1/Per2/Per3 genes, 78–80, 102, 111–115 circadian control of, 199, 200
PER1 protein, 113–114, 125 Phospholipase C, 249
Per2 gene, sleep disorders and, 137–138 Phosphorylation, 22, 158, 175
PER2 protein, 113, 125 in plant circadian oscillation, 206
as a tumor suppressor, 138 regulation of circadian oscillator proteins by, 52–57
PER3 protein, 114, 125 TIM proteins and, 62–64
PER-bGAL protein, 44 Photic input
PER-driven feedback, DBT regulation of, 55–57 PACAP in, 101–102
PER expression, 36–37 pathways, 97
RHYIndex 2/6/04 3:50 PM Page 280

280 INDEX

Photic pathways, phase shifting the circadian clock Plant circadian biology, beginning of, 186
by, 80–82 Plant circadian clocks, 185–209
Photic resetting, by frq gene in Neurospora, adaptive significance of, 208
178–179 molecular interactions within, 204
Photoautotrophs, cyanobacteria as, 143–146 Plant circadian system, model of, 187
Photocyanin, 143 Plant genes, effect on clock function and flowering
Photolyases, 61, 116, 189 time, 195–198
Photoperiodism, 186 Plant growth rhythms, mechanisms underlying, 198
Photopigments, 98 Plant photoreceptors, 186–189
novel, 98–99 Plants
Photoreception. See also Photoreceptors central oscillator components in, 192–194
circadian clocks and, 10–11 circadian control of gene expression in, 200–201
CRY mediation of, 64–65 circadian regulation mechanisms in, 205–206
Photoreceptors. See also Photoreception clock-associated genes in, 194–198
cryptochrome, 60–62 enzyme oscillation in, 199–200
extraretinal, 80–81 light signaling in, 191–192
in entrainment pathways, 7 multiple autonomous clocks in, 207–208
pineal gland as, 76–77 output rhythms of, 198–205
plant, 186–189 rhythm entrainment in, 191
Photosynthesis stomatal aperture, gas exchange, and CO2
in cyanobacteria, 146 assimilation in, 198–199
rhythmic expression of genes involved in, 200 Plasma growth hormone (GH), 239–240
Phototropins, 186 Plasminogen activator inhibitor 1 (PAI1), 126
mediation of Arabidopsis responses to blue light PnC401 gene, 201
by, 188–189 PnZIP gene, 201
Phototropism, 189 Polysialic acid (PSA), 104
PHOT proteins Polyubiquitin chains, 62–64
in Arabidopsis, 188–189 Portal veins, 236
in Arabidopsis phytochrome–cryptochrome Pregnancy, 242, 243
interaction, 190–191 Preovulatory LH surge, 243
Phycoerythrin, 143 Progesterone, 242, 243
PHY proteins, in Arabidopsis phytochrome– Prokaryotes, circadian rhythms of, 3, 143, 168–169
cryptochrome interaction, 190–191 Prokineticin 2 (PK2), 130
Physical behavior, human circadian rhythms in, locomotor behavior regulation by, 129–130
257 Prokineticin 2 receptor (PK2R), 130
Physiological processes, rates of, 6–7 Prolactin, 241–242
Physiology, roles of peripheral clocks in, 227–228 mammary glands and, 242–243
Phytochrome–cryptochrome interaction, 190–191 Proline auxotroph, circadian growth in Neurospora,
Phytochrome interacting factor (PIF) proteins, 171
194–195 Promoter elements, in CCGs, 182–183
Phytochromes, 186 Promoters, 13, 22
response to red/far-red light by, 186–187 circadian elements in, 28
Phytochrome signaling model, 188 Promoter trap experiments, 150–151
Pigment dispersing factor (pdf), 69 Proopiomelanocortin (POMC), 238
Pineal gland, 215, 221 Protein kinase A (PKA), 234
in birds, 76–77 Protein kinase A (PKA) mutant, 71, 72
Pinealocytes, 76–77, 81, 232 Protein kinase C (PKC), 234
Pinopsin, 81 Protein–protein interactions, assays for, 26–27
Pituitary adenylate cyclase activating polypeptide Protein regulation, posttranscriptional, 68
(PACAP), involvement in photic input, Proteins
101–102 in clock control, 13–14
Pituitary–adrenal secretion, 237–239 PAS-containing, 107–108
Pituitary gland, 235–237, 241 recombinant, 21
anatomy of, 235–236 Pr phytochromes, 187
thyroid gland and, 245 PSA-NCAM, 104
Pkai promoters, 152, 153 psbAI gene, 148, 150, 165
Plant calcium, circadian rhythm in levels of, 199 Pseudoresponse regulators, 193
RHYIndex 2/6/04 3:50 PM Page 281

INDEX 281

Puberty, 240 Rubisco activase (RCA) gene, 200


purF gene, 150 RUBISCO (RBC) gene, 200

Quantitative trait locus (QTL) analysis, 121–122 S-adenosylmethionine decarboxylase (SAMDC), 201
sasA disruption, effect on circadian regulation,
RAC3 protein, 125 159–163
Race tube assay, 172 sasA gene, as a circadian amplifier, 157–159
Radiotelemetry, 251 sasA null background, 161
Random mutagenesis screens, 18 SasA protein
Ras/MAPK mutations, 72 function in the circadian system, 164
RBCS gene, 201 kai gene expression and, 160–161
RD19a gene, 201 in Synechococcus circadian system, 163, 164
rdta transgene, 98 SCN cell line, 104. See also Suprachiasmatic nucleus
Recombinant proteins, 21 (SCN)
Red (R)/far-red (FR) light, phytochrome response SCN cells, pacemaking properties of, 224, 225, 226
to, 186–187 SCN neurons, multifaceted synchronization of, 104
Redox state, cellular, 136–137 Screens, identification of clock-controlled genes by,
Redundancy, in mammal circadian systems, 99 201–202
Releasing hormones, 236 Secreted factors, SCN output control and, 129
Renilla reniformis, 155 Sella turcica, 235
Reporter genes, 218 Sensory histidine kinase, 157–158
Reproduction, 242–243 Serine hydroymethyltransferase (SHM) genes, 200
mammalian, 97 Serotonin
Respiration, 147 in mammalian circadian rhythms, 123
human circadian rhythms in, 257 in melatonin synthesis, 233
Rest–activity cycles, 3–4 prolactin and, 242
mammalian, 95–96 shaggy (sgg) mutant, 70
Restriction enzyme-mediated integration (REMI), 88 shaggy (sgg) protein kinase, 57, 58
Retina Shiftwork, 266
in birds, 78 SIM1/SIM2 proteins, 125
light detection by, 97–98 Single-gene clock mutations, 36
Retinal degeneration (rd) mutant, 98 Sleep
Retinal degeneration slow (rds) mutant, 98 circadian and homeostatic regulation of, 261–266
Retinal ganglion cells (RGCs), 99 glucose regulation and, 249–250
Retina–SCN pathways, 102–103 gonadotropic axis and, 244
Retinohypothalamic tract (RHT), 77, 97 growth hormone release and, 239–240
Retinoic acid (RA), 135 masking of endogenous circadian rhythms by, 262
Retinoid add receptor (RAR), 134–135 oxytocin and vasopressin secretions during, 248
Retinoid-related orphan receptor b (RORBb), parathyroid hormone levels during, 249
133–134 Sleep deprivation, 261–263
Retinoid X receptor (RXR), 134–135 preserved circadian rhythms during, 263, 264
REV-ERBa receptor, 119, 133 total, 263, 264
rev-erb genes, 87, 119, 134 Sleep disorders, 137–138
Rhodnius prolixus, 220–221 Sleep phase syndromes, advanced and delayed, 268
Rhodopsin, 98 Sleep propensity, 265
Rhodopsin promoter, 41, 42 Sleep–wake cycle, 232, 244, 264–265
Rhythmic desynchronization, internal, 214–215 circadian control of, 258
Rhythms, in glucose regulation, 249–250. See also mammalian, 96
Hormonal rhythms voluntary control of, 260–261, 266
RIGUI gene, 111, 112 S. littoralis, 228
RNA cycling, 12–13 Slow-wave (SW) sleep, 239–240
RNase protection assay (RPA), 24 Somatostatin, 235, 240–241
Robinia, 199 Somatotrophs, 239
Rods, 98 Spatial analysis, 22–23
Root phototropic 2 (rpt2) gene, 190 Species, circadian properties across, 95
ROR family proteins, 134, 135 Sperm production, 227, 228
rpoD2 gene, 151, 165 Spontaneous internal desynchronization, 215
RHYIndex 2/6/04 3:50 PM Page 282

282 INDEX

SRC1 protein, 125 Thermoregulation, human circadian rhythms in, 257.


State parameters/variables, 13 See also Temperature regulation
Steroids, 237 Thyroid gland, 245–247
Stomatal aperture, circadian rhythms in, 198–199 Thyroid hormone receptor (TR), 135
Stromatolites, 144 Thyroid secreting hormone (TSH), 245. See also
Subcellular localization, 23–24 Thyrotropin stimulating hormone (TSH)
Subjective day, 8 Thyroid stimulating hormone (TSH), 241
Subparaventricular zone (SPZ), 127 Thyrotroph embryonic factor (tef) gene, 133
Subtractive hybridization experiments, 27–28 Thyrotrophs, 245
Superovulation, 244 Thyrotropin hormones (TH), regulation of, 245–246
Suppressor of phyA 1 (spa1) gene, 190 Thyrotropin releasing hormones (TRHs), 235,
Suprachiasmatic nucleus (SCN), 11, 215. See also 245–246
Retina–SCN pathways; SCN entries Thyrotropin stimulating hormone (TSH), 245–246
in birds, 77 Thyroxin (T4), 246–247
central oscillator in mammalian, 103 TIF2 protein, 125
connection to distinct brain areas, 127 timeless (tim) gene, in Drosophila, 216–217, 225
hormone release regulation by, 127–129 timeless (tim) gene products
human biological clock in, 256 Arabidopsis homologs of, 198, 204s
humoral signaling and, 251–252 circadian oscillations of, 39, 41–47
in insects, 76 timeless (tim) mutant, 70
in mammals, 76, 94, 99–100, 101–102, 103, 221–222 timeout gene, 124. See also TIM homologs
master clock in, 15–16, 221–222 TIM homologs, in mammals, 123–124
multioscillatory circadian organization and, Timing of CAB1 (toc1) mutants, 192–193
221–222 tim-luc reporter gene, 216, 218, 219
output control by, 126–127, 129 TIM protein, 216, 217
output pathways of, 128 CRY proteins and degradation of, 62–64
peripheral oscillators and, 131–134 light-dependent degradation of, 62–64
Supraoptic nuclei (SON), 131 light-induced degradation of, 57–60
Synechococcus, circadian organization in, 147, 157. PER protein collaboration with, 44–47
See also Synechococcus elongatus Tissue culture, oscillators revealed by, 134
Synechococcus circadian system, SasA protein in, Tissues, molecular clock component variation in,
163, 164 225–227
Synechococcus elongatus, 193 Tobacco, circadian rhythms in, 186
cikA gene of, 164–165 TOC1 protein, 192–193
circadian rhythms in, 143, 166, 167 Total sleep deprivation, 263, 264
as model cyanobacterium, 148–149, 150–152, Trait variability, in circadian rhythms, 267–268
156–157 Transcortin, 238
Synechocystis, 149, 156, 165, 169 Transcription factors, 22
Transcription/translation-based molecular feedback
takeout (to) mutant, 67–68, 70, 227 model, 158
tau gene, 116–119 trans factors, in circadian control, 150–151
cloning of, 117–118 Transforming growth factor a (TGFa), locomotor
cyclic activity and, 118–119 activity regulation by, 129, 130
tau mutant, 103 Transgenes, tissue specific expression of, 25
tau mutant hamster line, 117 Transgenic clock gene expression, 25–26
Temperature Transposase, 18
circadian rhythms and, 6–7 Transposon mobilization, 18
mammalian, 96 Triiodothyronine (T3), 246–247
response of Neurospora clock to, 178–182 Tritordeum, 201
Temperature regulation, by FRQ protein, 180–182. Tumor suppressors, PER2 as, 138
See also Thermoregulation Turntable experiment, 5
Temperature rhythms, 267 24-hour oscillator, 12
Temporal analysis, 22, 24 Type 0/1 phase-response curves, 9
Temporal separation, of nitrogen fixation and
photosynthesis in cyanobacteria, 146–147 UAS promoter, 64, 65
Terminology, chronobiological, 13 Ubiquitin, 63
Tetrapyrrole chromophore, 187 Ubiquitin proteasome pathway, 62–64
RHYIndex 2/6/04 3:50 PM Page 283

INDEX 283

Ubiquitination, TIM proteins and, 62–64 xClock (xClk) gene, 90


Ultradian day studies, 265 xCry/xCry2a/xCry2b genes, 90
Ultraviolet A (UVA) light. See also UV-dependent Xenopus, 58
photolyases clock genes in, 88–91
phototropins and, 186 cry genes in, 90
plant calcium circadian rhythms and, 199 Xenopus eyecup preparation, 88
Urinary potassium, circadian rhythms in levels of, Xenopus laevis, circadian rhythms in, 87–91
213–214, 214–215 xPer1/xPer2 genes, 88–90
UV-dependent photolyases, 115
Yeast, light signaling in, 192
Vas deferens lumen/muscles, 227, 228 Yeast two-hybrid assay, 24, 26, 27
Vasoactive intestinal peptide (VIP) receptors, 102
Vasopressin, 235, 247–248 zCry1a/zCry1b/zCry2a/zCry2b/zCry3/zCry4 genes,
secretion during sleep, 248 86–87
Vertebrates Zeaxanthine epoxidase (ZEP), 200
clock gene expression in, 216–217 Zebrafish
melatonin in, 232–233 circadian system organization in, 82–87
multioscillatory circadian organization in, 221 Clk gene in, 221
nonmammalian, 75–91 cryptochromes in, 86–87
Vibrio harveyi, 148 cycling clock genes in, 85
Visual perception, human circadian rhythms in, locomotor activity and melatonin synthesis in,
257 82–84
Visual SCN (vSCN), 77 per genes in, 86
Vitamin D, 249 Zebrafish clock, molecular components of, 84–87
vivid (vvd) gene, 179–180 Zeitgebers, 256
VPAC1/VPAC2 receptors, 102 freerunning rhythms and, 256–258
VPAC2 protein, 120 light as, 258–259
vrille (vri) mutant, 66–67, 70, 73 nonphotic, 258
VVD protein, 179, 180 Zeitgeber signals, 5–6
Zeitgeber time (ZT), 6
WC-1/WC-2 proteins, as regulators of frq gene, Zeitlupe (ZTL) gene family, 203–204
174–175, 176–177, 179 domain structure of, 196–197, 208
WC complex (WCC), 175 zfClk gene, 84–86
Western blot analysis, 22 zfPer1/zfPer2/zfPer3 genes, 86
wingless (wg) pathway, 57 ZGT gene, 194, 203, 204
Wnt (wg) signaling pathway, 58 Zinc finger, 202

Vous aimerez peut-être aussi