Vous êtes sur la page 1sur 34

Physiol Rev 96: 1093–1126, 2016

Published June 22, 2016; doi:10.1152/physrev.00036.2015

HUMAN INDUCED PLURIPOTENT STEM CELLS AS


A PLATFORM FOR PERSONALIZED AND
PRECISION CARDIOVASCULAR MEDICINE
Elena Matsa, John H. Ahrens, and Joseph C. Wu

Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, and Institute of Stem Cell
Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California

Matsa E, Ahrens JH, Wu JC. Human Induced Pluripotent Stem Cells as a Platform for

L
Personalized and Precision Cardiovascular Medicine. Physiol Rev 96: 1093–1126,
2016. Published June 22, 2016; doi:10.1152/physrev.00036.2015.—Human in-
duced pluripotent stem cells (hiPSCs) have revolutionized the field of human disease
modeling, with an enormous potential to serve as paradigm shifting platforms for
preclinical trials, personalized clinical diagnosis, and drug treatment. In this review, we describe
how hiPSCs could transition cardiac healthcare away from simple disease diagnosis to prediction
and prevention, bridging the gap between basic and clinical research to bring the best science to
every patient.

I. INTRODUCTION 1093 vidual variability (11, 45). Progress in genetic and genomic
II. TOOLS FOR REPROGRAMMING... 1094 research has highlighted some disease-associated genomic
III. GENERATIONS OF FUNCTIONAL... 1097 DNA (gDNA) polymorphisms [e.g., single nucleotide poly-
IV. BIOMEDICAL DATA SCIENCE 1099 morphisms (SNPs) or insertions/deletions (indels)] that pro-
V. hiPSC-BASED MODELS OF HUMAN... 1101 vide causal relationships among genotypes, disease suscep-
VI. ENGINEERED DISEASE MODELS... 1107 tibility, and pharmacokinetics (i.e., drug absorption, distri-
VII. INDUCED MODELS OF CARDIAC... 1110 bution, metabolism, and excretion) or pharmacodynamics
VIII. PATIENT RECRUITMENT VERSUS... 1111 (i.e., the physiological effects of the drug). Such relation-
IX. PHARMACOGENOMICS 1112 ships account for phenotypic variations of clinical impor-
X. POTENTIAL APPLICATIONS OF hiPSCs... 1113 tance in drug therapy and disease progression (173). How-
XI. CONCLUSIONS AND FUTURE... 1116 ever, large-scale studies to establish causal associations be-
tween genetic variations and clinical indications have been
difficult to conduct. The lack of robust methods to experi-
I. INTRODUCTION
mentally test the functional relevance of naturally occurring
polymorphisms in human cells has delayed progress, leav-
The ability to reprogram terminally differentiated human ing the determination of the molecular and cellular basis of
cells into induced pluripotent stem cells (hiPSCs) has fun- human phenotypic variation an unresolved challenge.
damentally altered the previous belief that cellular differen- However, revolutionary and rapidly evolving genome edit-
tiation is a unidirectional, nonreversible developmental
ing technologies such as clustered regularly interspaced
process (84, 257, 259). The discovery of cellular repro-
short palindromic repeats (CRISPR/Cas9), transcription ac-
gramming has opened the possibility to create in vitro mod-
tivator-like effector nucleases (TALEN), and zinc finger nu-
els of human diseases by isolating and reprogramming so-
cleases (ZFN), can significantly decrease the time and effort
matic cells from patients carrying familial disease-associ-
ated mutations. These patient-specific platforms have been it takes to generate human cell lines carrying specific gDNA
used extensively to understand the molecular mechanisms mutations or polymorphisms (46). Thanks to their abilities
leading to pathophysiological perturbations in humans, to undergo indefinite proliferation and single-cell clonal ex-
and test the pathways via which several drugs may amelio- pansion, hiPSCs serve as a particularly useful platform for
rate a disease phenotype or cause cytotoxicity (181, 182). genome editing (82, 97, 308). Therefore, hiPSCs can facili-
tate identification of exactly how disease mutations and
The capacity of hiPSCs to retain patient-specific genomic, polymorphisms result in cellular phenotypes, providing an
transcriptomic, proteomic, metabolomic, and other indi- experimental platform for precision medicine. By advanc-
vidualized big data information makes it possible to extend ing a new model of patient-powered research combined
their application beyond disease modeling, and into the with a patient-focused drug development program, preci-
field of precision medicine which encompasses the adoption sion medicine promises to accelerate biomedical discoveries
of novel prevention and treatment strategies based on indi- and offer clinicians novel tools, knowledge, and therapies

0031-9333/16 Copyright © 2016 the American Physiological Society 1093


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

that will enable them to select optimized treatments for each outline how, in the relatively short time since their discov-
individual patient (36a, 233a). ery, hiPSCs have been married with next generation se-
quencing (NGS) and genome editing technologies to high-
In this review, we focus on the ability of hiPSCs to facilitate light molecular mechanisms leading to heart disease, en-
precision medicine in relation to heart disease. Heart disease, abling us to link disease-associated mutations or
whether familial, congenital, or acquired, remains the leading polymorphisms to disease outcome, severity, and response
cause of mortality in men and women worldwide (150, 198). to therapy. Finally, we highlight the future prospects of
Currently, over 17 million deaths per year are attributed to hiPSCs to ably address the critical need for incorporating a
heart disease, and this number is projected to rise to 30 million human-based platform into drug discovery, disease diagno-
over the following 15 years (300a). Despite decades of exten- sis, and therapeutic pipelines.
sive and expensive research, few interventions have signifi-
cantly improved patients’ survival in heart failure (34). Even
the most commonly prescribed treatments (e.g., ␤-adrenore- II. TOOLS FOR REPROGRAMING SOMATIC
ceptor blockers) act primarily by delaying disease progression, CELLS TO hiPSCs
and can have adverse side effects such as fainting, seizures, or
bradycardia (71, 78, 143). These facts point to the need for The first reprograming methods published by the Ya-
enhancing our understanding of the molecular mechanisms manaka (257) and Thomson (319) labs utilized retroviral
leading to heart failure, and for developing improved predic- and lentiviral vectors to generate hiPSCs from human skin
tive, preventive, and reparative therapies. As detailed later on fibroblasts. They were able to demonstrate that transgenic
in this review, small and large animal models ranging from overexpression of OCT4, SOX2, KLF4, and c-MYC
fruit flies to primates have been utilized extensively to model (OSKM) or OCT4, SOX2, NANOG, and LIN28 (OSNL)
the pathophysiology of human heart disease (192). However, was necessary and sufficient to induce pluripotency in fibro-
species-specific variation has hindered significant progress, blasts. Subsequent interrogation of reprogramming mecha-
making any therapeutic advancements disproportional to the nisms identified that the OSKM or OSNL factors synergis-
amount of time and money invested (226). Thus cutting-edge tically activate the endogenous pluripotency machinery fol-
21st century heart failure research is required to use novel lowing a series of consecutive events (FIGURE 1). The first
human-based and human-relevant research methodologies to step to reprogramming is initiation, and it is characterized
efficiently address the lack of effective therapies against heart by loss of somatic cell gene expression, metabolic changes,
failure. increased cellular proliferation rate, inhibition of apoptosis
and cellular senescence pathways, and initiation of mesen-
We hereby show examples of how hiPSC-derived cardiomy- chymal-to-epithelial transition (MET) (48). These changes
ocytes (hiPSC-CMs) can be used to generate human models are followed by maturation, a stage when cells acquire ex-
of heart diseases in a dish, which faithfully recapitulate the pression of pluripotency-associated genes, but are still de-
human heart’s pathophysiology and respond appropriately pendent on transgene overexpression to become fully repro-
to clinically relevant pharmacological intervention. We also grammed. The final stage of reprogramming, known as the

Initiation Maturation Stabilization


↓ somatic cell gene expression ↑ pluripotency gene expression Epigenetic remodeling

≠ in metabolism (e.g., OCT4, NANOG) ↑ telomere length

↑ proliferation rate X-chromosome reactivation

of apoptosis

of cellular senescence pathways

Mesenchymal-to-epithelial transition Transgene dependence


0 4 8 12 Time (days)
FIGURE 1. Stages of hiPSC reprogramming. Mechanistic analyses have demonstrated that reprogramming
of somatic cells to hiPSCs consists of three major steps. The first step is “initiation,” and it is characterized by
loss of somatic cell gene expression, metabolic changes, increase in cellular proliferation rate, inhibition of
apoptosis and cellular senescence pathways, and initiation of mesenchymal-to-epithelial transition (MET).
These changes are followed by “maturation,” a stage when cells acquire expression of pluripotency-associated
genes, but are still dependent on transgene overexpression to become fully reprogrammed. The final stage of
reprogramming is “stabilization,” and it is achieved when cells undergo X-chromosome reactivation and
telomere elongation, acquire epigenetic characteristics of pluripotent cells, and are transgene independent.
1, Increased; 2, decreased; ⫽, change; ;, inhibition.

1094 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

stabilization phase, is achieved when cells undergo X-chromo- cific backbone mutations that affect key viral proteins, and
some reactivation and telomere elongation, acquire the epige- allow faster and more efficient removal of viral material
netic characteristics of pluripotent cells, and become transgene from the cytoplasm of host cells by a temperature shift (10).
independent. hiPSCs are thought to continue stabilization and This technology decreases the time and increases the effi-
to acquire genetic and epigenetic signatures that more closely ciency of establishing desired hiPSC lines. With the use of
resemble those of embryonic stem cells (ESCs) (270) at late this method, various cell types, including fibroblasts and
passages, typically over passage 16 (14, 221). peripheral blood mononuclear cells (PBMCs), have been
effectively reprogrammed (41, 60, 72).
Reprogramming efficiency (i.e., the number of starting so-
matic cells which transition to hiPSCs) has been reported to
be significantly enhanced by addition of supplementary plu- B. Reprogramming With Episomal Plasmids
ripotency factors (12) or epigenetic modifiers (160) to the
reprogramming mix. Rais et al. (225) and Lujan et al. (168) As an alternative to retroviral or lentiviral-mediated induc-
also demonstrated in both human and mouse cells that al- tion of pluripotency, several DNA-based reprogramming
most 100% of cells in a differentiated population are capa- methods have also been developed using either nonreplicat-
ble of reprogramming following depletion of Mbd3, a core ing (56, 114, 209) or replicating episomal vectors (317).
member of the Mbd3/NuRD (nucleosome remodeling and These techniques are attractive because they reduce biosafety
deacetylation) repressor complex. Even though some con- concerns involved in the production and use of viral particles,
troversy exists around this topic, likely due to due technical as well as the risk of tumorigenesis due to potential reactiva-
differences (64), these studies potentially demonstrate that tion of the KLF4 and c-MYC oncogenes, or due to insertional
while iPSC reprogramming is typically a stochastic process, mutagenesis. Nevertheless, the reprogramming efficiency
it can become deterministic upon epigenetic manipulation. achieved using nonreplicating vectors is relatively low and re-
quires multiple transfections of the target cells (114, 209).
Beyond the investigation of hiPSC reprogramming mecha- Potential explanations for this phenomenon include the low
nisms, clinically relevant advances have been achieved by transfection efficiencies of large polycistronic plasmids and
the development of automated, high-throughput derivation enhanced transgene silencing mechanisms operating on plas-
and characterization protocols for hiPSCs (215, 245), use of mid-based vectors in mammalian cells (167).
chemically defined media (24), and substitution of retrovi-
ral and lentiviral transgene overexpression vectors by inte- To overcome these limitations, the Epstein-Barr virus
gration-free systems that circumvent the risk of spontane- (EBV)-based reprogramming technology was developed.
ous tumor formation due to insertional mutagenesis. Such This potent methodology takes advantage of the EBV-en-
methods include the use of excisable viral vectors, noninte- coded nuclear antigen-1 (EBNA-1) trans-acting element,
grating viruses (250, 251), and even virus-independent ap- and EBV-encoded oriP cis-acting element to enable plas-
proaches such as plasmid, episomal, DNA, protein, or mids to persist by dividing within human cells as multicopy
mRNA mediated transgene overexpression (114, 293, episomes that attach to chromosomes during mitosis (104,
324). The advantages and disadvantages of the most prom- 209). Therefore, the use of episomal vector systems signifi-
inently used nonintegrating reprogramming techniques are cantly increases reprogramming efficiency through pro-
outlined in TABLE 1 and are discussed in detail below. longed transgene expression within transfected cells (209).
Following multiple cell divisions, oriP/EBNA-based vectors
progressively diminish from the targeted cells, thus elimi-
A. Nonintegrating Sendai Virus nating potential tumorigenesis or differentiation deficien-
cies due to transgene reactivation.
One of the most common currently used techniques to in-
duce pluripotency is based on F-deficient Sendai virus (SeV/ Minicircle vectors were also developed as an improvement
⌬F), a nontransmissible negative-sense single-stranded to classical episomal vectors (114). Minicircles are episomal
RNA virus that replicates in the cytoplasm of infected cells vectors devoid of any bacterial plasmid and are therefore
without DNA intermediates or genomic integration into the smaller in size than standard plasmids. This attractive fea-
target cell genome. Despite being one of the most reliably ture enhances their transfection and expression rates (38).
integration-free reprogramming methods currently avail- However, the reprogramming efficiency achieved using
able (72), the use of Sendai virus is expensive and requires minicircle vectors is low, and their production and purifi-
more stringent biosafety containment measures than non- cation methodology is relatively complex and time consum-
viral reprogramming methods. Moreover, residual viral ing (202). To address the aforementioned drawbacks of this
material persists for an extended period of cell culture (up system, a novel, single plasmid reprogramming system
to 10-20 passages), thus delaying the establishment of virus- called 4-in-1 codon optimized mini intronic plasmid
free hiPSC lines for downstream analysis and differentia- (CoMiP) was developed, which carries codon-optimized se-
tion experiments (80). Recently, temperature-sensitive Sen- quences of the canonical OKSM reprogramming factors to
dai virus particles have been developed by engineering spe- achieve more accurate and faster translation of transgenes.

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1095


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

Table 1. Tools for reprogramming somatic cells to hiPSCs


Reference
Tool Subtype Vector Transgenes Efficiency, % ⴙ ⴚ Nos.

Viral Integrating Retrovirus OSKC, OSK, OS, 0.001–0.1 1Efficiency, robust Risk for random 58, 213,
OK, O integration into 257
somatic
genome
Lentivirus OSKC, OSNL, 0.0001–0.1 Can infect nondividing Same as above 103, 201
OSN, OSL cells
Excisable Cre-LoxP Lentivirus OSKC, OSK 0.005–0.01 2Risk for IM Labor intensive, 246
may leave
footprint
Nonintegrating Adenovirus OSKC 0.001 No risk for IM 2Efficiency 325
Sendai virus OSKC 1 Same as above 1Cost 72
Nonviral, DNA Plasmids EBV Episome OSKCNL & 0.00003–0.02 2Risk for IM 2Efficiency, need 316, 318
based SV40LT for additional
factors
Minicircle OSNL 0.0005–0.005 Inexpensive, easy to use, 2Efficiency, rapid 114
2Risk for IM transgene
silencing may
require
multiple
transfections
CoMIP OSKC 0.02–0.03 Same as above Same as above 56, 57
Transposons PiggyBac OSKC 0.1 1Efficiency May leave 123
footprint, risk
for random
insertion
Sleeping Beauty OSKC 0.02 2Risk for IM Same as above 50
HAC OSKC & p53 0.00007–0.00014 Built-in safeguard system 2Efficiency 94
shRNA based on herpes
simplex virus
Nanoparticle OSKC, SKN & 0.001–0.049 Stable transgene source Labor intensive 29, 151
NR5A2
Nonviral, non-DNA CPP OSKC 0.001 No risk of IM 2Efficiency 135
based
RNA replicon VEE OSKC, OSKG 0.25 1Efficiency, easy, 2risk Possibility of RT 315
for IM to cDNA,
leading to IM
Synthetic Cationic lipid OSKC & VPA, & 1.4–4.4 Same as above Expensive, labor 293
mRNAs hypoxia intensive
miRNAs Cationic lipid miR-200c, 302, 0.1 Same as above Same as above 194
369 s
Small molecules N/A O & VC6T 0.00005 Minimal genetic Greatly 158
(mouse) modification, easy to 2efficiency,
use not
demonstrated
in humans
VC6TFZ 0.2 No genetic modification, Not 98
no risk for IM, easy to demonstrated
use in human cells

A summary of the plethora of methods available for the reprogramming of somatic cells to hiPSCs, with
reference to their individual efficiencies, advantages (⫹), and disadvantages (⫺). O, OCT4; S, SOX2; K, KLF4;
C, c-MYC; N, NANOG; L, LIN28; SV40LT, simian vacuolating virus 40 large T antigen; IM, insertional
mutagenesis; VPA, valproic acid; EBV, Epstein Barr virus; CoMIP, codon optimized mini intronic plasmid; RT,
reverse transcription; VEE, Venezuelan equine encephalitis self-replicating RNA virus; HAC, human artificial
chromosome; CPP, cell penetrating peptides; VC6T; VPA, CHIR99021, 616452, and tranylcypromine;
VC6TFZ, VC6T, forskolin and 3-deazaneplanocin A; 1, high; 2, low.

Similar to minicircles, the CoMiP vector system overcomes potential to integrate into the host genome. Careful down-
transgene silencing limitations that are often observed with stream screening methods must be used to confirm the der-
regular plasmids, and provides at least 2–10 times higher ivation of integration-free pluripotent cells to guarantee the
levels of transgene expression, as demonstrated in vitro safety of these systems (80).
with HEK293 cells and in vivo with mouse liver cells (166).
Furthermore, the 4-in-1 CoMiP vector is a highly efficient,
integration-free, and cost-effective methodology applicable C. Non-DNA Approaches for the Induction of
for hiPSC reprograming in a wide variety of cell types (57). Pluripotency

One area of potential concern when using episomal DNA- One of the first non-DNA tools used for the generation of
based reprogramming methods is that they maintain a small hiPSCs was transient transfection of modified mRNAs. This

1096 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

approach not only guarantees the derivation of integration- of the population, normal 46 XX or 46 XY karyotype,
free hiPSCs due to the lack of any DNA intermediates, but identical short tandem repeat (STR) profiles between paren-
also obviates further time-consuming screening experi- tal and hiPSC lines, sterility, absence of mycoplasma or
ments (177, 293). Initial reprogramming experiments using endotoxin (⬍0.5 EU/ml) contamination, lack of episomal
mRNA transfection required a laborious series of consecu- or other vector integration into gDNA, viral panel clearance
tive mRNA transfections for 14 days, as well as pretreat- (e.g., retroviruses, adeno-associated viruses, HIV, EBV,
ment of target cells with the expensive interferon-␣ antag- ETC, and bovine or porcine viruses), and over 50% cell
onist B18R (177) to prevent cell death from repeated viability per cryopreserved vial (9). These assays should be
mRNA transfections (44). Another limitation of initial pro- conducted prior to the use of hiPSCs for clinical applica-
tocols for mRNA-mediated reprogramming for clinical ap- tions.
proaches was the requirement of mouse embryonic fibro-
blast (MEF) feeder cells and conditioned media, which po-
tentially increases the risk of transmitting undetected III. GENERATION OF FUNCTIONAL HUMAN
animal pathogens to human cells (4). Recent optimizations CARDIOMYOCYTES FROM hiPSCs
of the established mRNA reprogramming factors addressed
some of the aforementioned problems, allowing the deriva-
tion of footprint-free hiPSCs from human fibroblasts with-
A. Genetic and Epigenetic Control of Cardiac
out the use of feeder cells or other potentially xeno-contam- Specification
inated reagents (294), using only a single mRNA transfec-
tion (315). Still, mRNA-based reprogramming has thus far Cardiac specification during development is thought to be a
only allowed reprogramming of adherent cells, which limits four-stage process tightly controlled by transcription factor
the types of differentiated cells that can be reprogrammed. networks, signaling pathways, and epigenetic modifications
(25). On the basis primarily of mouse studies, it is thought
Another successful DNA-free method for hiPSC generation that the first stage of cardiac specification during develop-
involves the use of transmembrane-permeable fusion pro- ment consists of cardiac mesoderm formation around the
teins (135, 152). However, due to slow kinetics, low effi- time of gastrulation (261). This process is facilitated by
ciency, and high cost (80), this technique has not been upregulation of NODAL and BMP4 signaling (6). Subse-
widely adopted. In addition, an exciting alternative ap- quent induction of WNT signaling by BMP4 activates the
proach has been the elimination of classical reprogramming expression of essential mesoderm transcription factors,
factors by exclusively using small-molecule compounds to such as brachyury (T), which in turn stimulate Mesp1, a
reprogram mouse somatic cells (98). Reproducing this tech- master regulator and marker of cardiogenic mesoderm for-
nique using human somatic cells would greatly broaden its mation (15). During the second stage of cardiac specifica-
clinical interest. tion, Mesp1⫹ cardiac mesoderm progenitors give rise to
two distinct progenitor cell populations known as the first
heart field (FHF) and second heart field (SHF) progenitors
D. Selecting the Right Reprogramming Tools (154). This process is controlled by intrinsic signaling as
for Clinical Applications well as complex extrinsic signaling by BMP4 from the un-
derlying anterior ectoderm, and BMP4, FGF, and WNT
Recent evidence suggests that the number and size of karyo- signaling from the overlying anterior endoderm (247).
type abnormalities, such as copy number variations
(CNVs), are significantly higher in isogenic hiPSC lines gen- In the FHF, WNT signaling inhibition through the Mesp1-
erated using integrating reprogramming methods compared activated gene Dkk1 leads to upregulation of the cardiac
with nonintegrating methods (126). This confirms the need progenitor marker Nkx2-5 and the FHF-specific marker
for adopting safe reprogramming methods for clinical ap- T-box transcription factor Tbx5 (70). This incorporates the
plications. It is important to note that not all reprogram- third stage of cardiac specification, known as patterning of
ming routes may lead to the same pluripotency state. Al- the heart fields, which is followed by the fourth and final
though genetic background and cell type of origin contrib- stage of terminal cardiac differentiation. The latter is
ute to heterogeneity among hiPSC lines (231), the type of brought about by Nkx2.5 and Tbx5-mediated activation of
reprogramming strategy used can also affect the stoichiom- connexin 40 (Gja5), Nppa, and sarcomere-specific genes
etry and expression levels of individual transgenes, thus (18) which induce the formation of atrial cardiomyocytes
leading to transcriptional diversity amongst reprogrammed (CMs), left ventricular CMs, and cardiac bundle of His
populations (327). Carefully assessing the quality of the pacemaker cells from the FHF (95). Nkx2.5 and Tbx5
reprogrammed state is vital as new technologies emerge. physically interact to coregulate their downstream targets
Quality control (QC) testing and release assays that ensure during cardiac differentiation and morphogenesis (18),
the identity, safety, purity, and viability of new hiPSCs lines whereas their independent binding activity also serves to
include the expression of pluripotency-associated markers prevent transcription factors from distributing to ectopic
(e.g., OCT4, SSEA4, TRA-1-81, TRA-1-160) in over 70% loci and activate lineage-inappropriate genes (169). Positive

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1097


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

regulation of Nkx2.5 by polycomb repressive complex 1 IWR1) that modulate BMP and WNT signaling (22, 24, 81,
(PRC1)-mediated chromatin remodeling (241) and negative 159). Small molecules are advantageous compared with
regulation by histone 3 trimethylation (205) add an addi- growth factors in that they are cheaper to produce, simpler
tional level of complexity to these temporally and spatially to store, more stable, and more amenable to quality control
controlled gene regulatory pathways. (137). Finally, the labor intensive process of embryoid body
formation has been replaced by hiPSC differentiation in
In the SHF, Isl1 and Mef2c act downstream of Mesp1 (125) monolayer (145) or suspension cultures (37) that generate
to activate cardiac-specific factors such as Smyd1, Hand1, over 90% pure CM populations, and are amenable to au-
Irx4, and Fgf10 (217, 295) and ultimately give rise to atrial tomation and scale-up of 1.5–2 billion cells per liter of
CMs, right ventricular CMs, and sinoatrial node pace- culture. These remarkable advancements in cardiac specifi-
maker cells. Mef2c is known to be regulated by DNA meth- cation of hiPSCs have greatly enhanced their potential for
ylation and histone acetylation (33, 260), further support- clinical application.
ing the importance of epigenetic modifications during car-
diac specification. Even though many details of this process
are yet to be untangled, our current understanding of car- C. Generation and Characterization of
diac specification during development has greatly facili- hiPSC-CMs at a High-Throughput Scale
tated in vitro cardiac differentiation of hiPSCs, as described
below. The clinical application of hiPSC-CMs necessitates their
production and functional characterization in large scale
(52). In parallel to the evolution of cardiac differentiation
B. Protocols for Cardiac Differentiation protocols in suspension bioreactors as mentioned above
(66, 132, 235, 252), automated liquid handling platforms
Cardiac differentiation of pluripotent stem cells has pro- (e.g., Tecan Freedom Evo 100, Sartotious CompacT SelecT,
gressed from an original efficiency of 5–10% to over 90% and Beckman Coulter Biomek FXP) for robotic scale-up of
efficiency within the past 15 years (23). Original protocols mouse and human pluripotent stem cells and their differen-
were based on random differentiation of three-dimensional tiated cardiac derivatives have been evaluated and demon-
pluripotent stem cell aggregates, known as embryoid bod- strated to improve the consistency and quality of cell cul-
ies, to CMs. Techniques including the enrichment with Per- tures (105, 139, 215, 267).
coll separation gradients (305), flow cytometry sorting us-
ing the mitochondrial dye tetramethylrhodamine methyl es- Industrial-scale phenotyping of hiPSC-CM properties is
ter perchloratethe (TMRM) (92), and the use of the surface also advancing. High content imaging (HCI) platforms
markers EMILIN2 (280), SIRPA (62), or VCAM1 (277) have been used to perform 96, 384, or 1,536-well screening
have been shown to selectively enrich CM populations. for small molecules to improve reprogramming efficiency,
However, the most widely reproduced method for CM en- hiPSC culture, and cardiac differentiation and maturation
richment has been based on the distinct metabolic differ- (52). Importantly, HCI has been used to assess drug-in-
ences in glucose and lactate metabolism between CMs and duced cardiotoxicity in hiPSC-CMs. For example, Sirenko
other differentiated or undifferentiated cell types, where et al. (242) have used HCI platforms to perform Calcei-
glucose-deprived culture media containing abundant lac- nAM, Hoechst, and MitoTracker imaging in hiPSC-CMs to
tate was found to favor the survival of CMs over other cell evaluate the cardiotoxicity of 131 drugs affecting Na⫹, K⫹,
types (272). and Ca2⫹ channels, as well as adreno-, dopamine, and his-
tamine receptors. Mioulane et al. (193) also used the
Advances have also been made in cardiac specification of Thermo Scientific Arrayscan imaging platform to study cel-
hiPSCs based on the observation that this process is highly lular stress induced by chelerythrine, a cell-permeable pro-
dependent on precise modulation of both endogenous (131, tein kinase C inhibitor, by performing combined TMRM,
212) and exogenous BMP and WNT signaling pathways caspase-3, and BOBO-1 staining to measure mitochondrial
(122, 307). Targeted protocols that use key signaling content, apoptosis, and cell death, respectively. More de-
growth factors such as BMP4 and Activin A enabled con- tailed high content assessment of hiPSC-CM mitochondrial
version of pluripotent stem cells to CMs with an improved function has been performed by measuring glycolysis and
efficiency (26, 321). This discovery also emphasized the oxidative phosphorylation (OXPHOS) using the Seahorse
importance of using hiPSC reprogramming strategies that Extracellular Flux Analyzer (63, 289).
do not allow for continued overexpression of reprogram-
ming factors, because OCT4, SOX2, and NANOG each Importantly, the development of 96-well noninvasive
have known roles in inhibiting differentiation by modulat- multi-electrode array (MEA) systems (e.g., Multichannel
ing BMP4-mediated mesendoderm induction (188, 292). Systems and Axion Biosystems Maestro) and contractility
analysis platforms (e.g., Cellogy Pulse and SONY SI8000)
In protocols developed more recently, growth factors were enable recording of field potentials and contraction profiles
also replaced by small molecules (e.g., CHIR99021 and of hiPSC-CMs in an automated fashion. Besides enabling

1098 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

high-throughput functional analysis of drug-induced effects ligation to the sequencing primer, and the specificity of
in hiPSCs, such platforms also enable monitoring of chronic ligation is analyzed by interrogating the first and second
toxicity using long-term recordings over several hours, bases that are incorporated in each ligation reaction. Mul-
days, or weeks. For example, Gilchrist et al. (79) performed tiple cycles of ligation and detection finally enable reading
high-content MEA recordings at 0.5, 1, 2, and 4 h post of the full fragment. Another platform marketed by Ther-
exposure to compounds with known proarrhythmic prop- moFisher is the Ion Torrent, which encompasses the Ion
erties (e.g., verapamil, ranolazine, flecainide, amiodarone, PGM, Ion Proton, and Ion S5 systems. This platform uti-
ouabain, cisapride, and terfenadine) and found that cardio- lizes yet another unique detection technology, which relies
toxicity could be recapitulated in vitro using hiPSC-CMs. on identification of proton release when a nucleotide is
Similar data were presented using video-edge contractility incorporated into a DNA fragment by the polymerase en-
analysis by Maddah et al. (174). zyme, resulting in a detectable local shift in pH for each
dNTP introduced. The Ion Torrent also uses a water-in-oil
The evolution of hiPSC-CM technologies which enable emulsified droplet method for initial PCR-based fragment
high-throughput culture and integration of these cells into amplification, which is a methodology shared with Roche’s
high-content industrial platforms that assess structure, mi- 454 platforms (http://www.454.com). However, in the 454
tochondrial function, electrophysiology, calcium tran- platforms, reads are detected using a method known as
sients, and contractility are paramount towards their poten- sequencing-by-synthesis or pyrosequencing, which relies on
tial applications in precision medicine. the recognition of pyrophosphate release on nucleotide in-
corporation, rather than chain termination with dNTPs or a
IV. BIOMEDICAL DATA SCIENCE change in pH. Revolutionary technology is also incorpo-
rated into Pacific Biosciences’ RS and Sequel tools (http://
www.pacb.com) which utilize a proprietary Single Mole-
Our ability to generate pure hiPSC-CM populations in large
cule, Real-Time (SMRT) technology to deliver sequencing
scale has opened the route towards performing in vitro
of the longest reads on the market without the requirement
functional characterization assays (e.g., structural, electro-
of a PCR amplification step. Increased read length can im-
physiological, contractile, and metabolic profiling) using
prove the alignment process, especially in stretches of highly
automated high-throughput screening platforms (52, 128).
repetitive elements. SMRT is based on zero-mode wave-
The combination of these platforms with next generation
biomedical data science is transforming hiPSC-based explo- guides that permit light to specifically illuminate the bottom
ration of molecular mechanisms that underlie cellular func- of a well in which a DNA polymerase/template complex is
tion and contribute to human health. High-throughout se- immobilized. Phospholinked nucleotides then enable detec-
quencing information is becoming increasingly important tion of the immobilized complex as the DNA polymerase
in biomedical research (286), and it has gradually become enzyme produces a natural DNA strand based on the im-
more cost effective and easier to perform thanks to contin- mobilized template. Other NGS platforms include SeqLL’s
uous improvement in related technologies (178, 228, 269). HeliScope (http://seqll.com), the first microscope-based
The most prominent NGS technologies are compared and NGS tool that eliminates the need for a PCR amplification
contrasted in TABLE 2 and are outlined in detail below. step and, therefore, avoids any associated amplification
bias. The advantages and disadvantages of each technology
are outlined in TABLE 2.
A. Prominent High-Throughput Sequencing
Technologies Despite their differences in sequencing chemistries, most
platforms rely on a common experimental workflow, which
A key vendor for NGS platforms is Illumina (http:// consists of random fragmentation of gDNA or RNA, and
www.illumina.com), whose bridge amplification tech- ligation of platform-specific adapters to the flanking ends of
nology allows billions of reads to be sequenced per run. the fragments generated to produce a library. Libraries are
The bridge amplification method relies on segregation of then amplified through platform-specific PCR (e.g., on glass
identical amplified sequences into millions of small clus- or beads) and undergo coupled DNA synthesis and detec-
ters, which are immobilized on a flow cell surface to tion processes. As multiple sequencing reactions are run
facilitate enzymatic access to the DNA. Amplified se- simultaneously, this process is also known as Massively
quences are then detected using fluorescence-labeled po- Parallel Sequencing (42). Desynchronization of reads dur-
lymerization terminator dNTPs that allow parallel se- ing a sequencing and detection cycle is a source of NGS
quencing of millions of clusters by detecting the fluores- errors, which can range from base substitutions to inser-
cence emission following addition of each dNTP. A tions or deletions, depending on the sequencing platform
different detection chemistry is employed by the SOLiD used (286). The average number of times a given region is
System, which is currently marketed by Thermo Fisher Sci- sequenced by independent reads in a sequencing experiment
entific (http://www.thermofisher.com). In this platform, a (known as read “depth”) can be increased to boost confi-
set of four fluorescently labeled di-base probes compete for dence in the accuracy of sequencing information (3).

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1099


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

Table 2. Overview of high-throughput platforms available for biomedical science applications


Reads/ Time/ Reference
Vendor Chemistry Platform run run Applications ⴙ ⴚ Nos.

Illumina Reversible termination MiSeq 15 M 4–55 h A 2Equipment cost 1Cost per sample 222
MiSeq FGx Same as above Same as above
NextSeq 500 130–400 M 11–29 B Same as above Same as above
h
HiSeq 2500 300 M-4 B 7 h-6 C 2Cost per sample, 1Instrument cost, 165
days 1scale and 1time, 2read
throughput length
HiSeq 3000 2.5 B 1–3.5 Same as Same as above Same as above
days above
HiSeq 4000 2.5–5 B Same as Same as above Same as above
above
Thermo Fisher Proton detection Ion PGM 400 K-6 M 2.3– A 2Cost and time 2Read no. and 222
7.3 h sample
throughput
Ion Proton I 60–80 M 2–4 h B Same as above Same as above 190
Ion S5 3–80 M 2.5–4 Same as Same as above Same as above
h above
Ion S5 XL Same as 1Sample 1Equipment cost
above throughput
Ligation SOLiD 1.4–2.8 B 8h C 2Error rate, 2cost, 2Read length, 1run 165
5500/5500xl 1throughput time
SOLiD 2h Same as 2Time 1Instrument cost
5500W/5500xlW above
PacBio Real-time sequencing RS I 23 K 1–12 h B Longest read length, 1Error rate 222, 229
no PCR step,
simple
preparation
RS II 62 K Same as Same as above 1Cost 229
above
Sequel 400 K 1–6 h A Same as above Same as above
Roche Pyro-sequencing 454 GS Junior 20–100 K 3.5 h A 1Read length 1Error rate, 1cost, 165
2throughput
454 GS FLX⫹ 20 K-1 M 6h Same as above Same as above
SeqLL Single molecule HeliScope 600–800 M 8 days C Nonbiased 1Run time, cost, 268
representation of and error rate
reads
Oxford Nanopore Exonuclease gridION* 4–10 M Flexible C Greatly 1read 1Error rate 88
sequencing length, 2run time

A summarized cross-comparison of the sequencing abilities, applications, advantages (⫹) and disadvantages
(⫺) of next generation sequencing instruments commercialized by leading vendors in the field. WG, whole
genome; K, thousand; M, million; B, billion; PacBio; Pacific Biosciences; PGM, Personal Genome Machine; 1,
high; 2, low; no., number; A, targeted gene panels; B, exome and RNA sequencing and targeted gene panels;
C, de novo sequencing; WG, exome, RNA, ChIP, and methyl sequencing, and targeted gene panels; *Instru-
ment is under development.

B. Biological Information Obtained Via High- proach to create a cost-effective and rapid clinical-grade
Throughput Technologies NGS platform that enables detection of thousands of mu-
tations in over 50 genes (e.g., TNNT2, MYH7, and
NGS platforms enable the deciphering of complex yet valu- LMNA/C) that are associated with familial cardiomyopa-
able biological information. For example, whole genome thies. Such platforms promise to facilitate the identification
(WG) or exome sequencing can be used to identify muta- of underlying disease mutations, and gather personalized
tions that may cause genetic disorders, or genomic poly- information that will expedite precision medicine endeav-
morphisms that may underlie disease progression, disease ours.
severity, and response to pharmacotherapy. A read depth of
30 –100⫻ is recommended to enable accurate detection of RNA-sequencing (RNA-seq) is a widely used method to
gDNA variation (146). However, as the read depth in- generate dynamic and quantitative information regarding
creases, so does the sequencing cost (279). Using targeted transcribed mRNA present in a tissue or cell population at
capture arrays that focus on a subgroup of target genes the time of sample collection. Its high sensitivity allows
involved in a specific pathway or disease can provide a detection of low abundance transcripts and splice variants,
balance between performance and cost. Notably, in a recent which can contribute towards unraveling the molecular
publication, Wilson et al. (298a) used a padlock probe ap- mechanisms of cellular processes (144). As mentioned

1100 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

above for gDNA sequencing, several gene targeted plat- the interactive personal omics profiling (iPOP) initiative (155),
forms (e.g., Ion AmpliSeq panels for genes involved in de- which is intended to assemble an individual’s exome, tran-
velopmental, heart, or metabolic disease) are available for scriptome, proteome, metabolome, microbiome, and other
RNA-seq, all of which are designed to reduce the cost of omes to create a dynamic profile of the physiology and
experimentation, and increase sample throughput. RNA- health state of the individual. Such collectives can become
seq is also not limited to the quantification of mRNA tran- stepping stones towards precision health by improving risk
scripts; depending on the methods used for RNA isolation prediction as well as disease diagnosis, monitoring, and
and library preparation, it can sequence other RNA popu- treatment.
lations such as miRNA, tRNA, or ribosome-bound RNA to
detect mRNA actively being translated into proteins (108).
The ability to measure the abundance of different RNAs has C. The Combination of hiPSC and NGS
in recent years refined the investigation of a variety of bio- Technologies
logical problems, including optimizing experimental condi-
tions (e.g., drug dose and timing), time-course experiments In hiPSCs, transcriptional and epigenetic variation originat-
(e.g., to monitor developmental processes, or disease pro- ing from genetic background have been shown to dominate
gression), population-specific response profiling (e.g., in over cell type of origin or derivation method in regards to
healthy and diseased individuals), studying response to the variation observed among lines from different patients
treatment (e.g., to several different drugs), and performing (40). This demonstrates that hiPSCs preserve patient-spe-
large-scale RNA interference (RNAi) studies (249). cific omic information and, therefore, provide a useful ex-
perimental platform to study the pathophysiological effects
Because gene expression can also be greatly influenced by of individualized biomedical data. An interesting study re-
epigenetic modifications such as histone methylation or lated to hypertrophic cardiomyopathy (HCM) was pub-
acetylation, as well as gDNA methylation of cytosine gua- lished by Zhi et al. (323), who performed whole exome
nine dinucleotides (CpGs) (112), chromatin immunopre- sequencing in seven sibling trios to identify missense or
cipitation sequencing (ChIP-seq) and DNA methylation se- nonsense mutations that were associated with increased left
quencing (Methyl-Seq) can also provide valuable informa- ventricular (LV) mass. Functional correlation of these mu-
tion regarding biological processes. ChIP-seq involves tations to hypertrophy-related gene signaling changes in
cross-linking of histone proteins to gDNA (e.g., using form- hiPSC-CMs led to the conclusion that a predicted conserved
and damaging nonsense variant in exon 13 (A2099G) of the
aldehyde), followed by an immunoprecipitation reaction
THBS1 gene was significantly associated with pathologi-
with antibodies against histone modifications associated
cally increased LV mass. It is anticipated that, in combina-
with actively transcribed genes (e.g., H3K4Me3 and
tion with NGS tools, hiPSC technology will serve as a next-
H3K9ac) or silenced genes (e.g., H3K27Me3). The result-
generation biomedical interface enabling us to establish
ing immunoprecipitated DNA is then sequenced in an NGS
strong phenotype-to-genotype correlations and understand
platform to identify genes associated with each type of his-
complex biological systems (90). Further examples of stud-
tone epigenetic modification (224). On the other hand,
ies which provide proof-of-principle that hiPSCs can be
Methyl-seq relies on the comparison of native gDNA to
used to understand how complex gene networks and poly-
bisulfite-treated gDNA, in which non-CpG cytidine nucle-
genic modifications may lead to heart disease are discussed
otides are converted to uracil nucleotides (141). Regions in more detail in the following section.
lacking bisulfite conversion are thought to be methylated
and silenced, since DNA methylation is strongly correlated
with the suppression of gene expression (278). Epigenetic V. hiPSC-BASED MODELS OF HUMAN
modifications influence developmental processes (107), and INNATE DISEASE
a variety of disorders, including heart disease (1, 47), and
their study helps us understand the physiological and path- The most widely studied hiPSC-based models of human
ological mechanisms involved. cardiovascular disease are those caused by familial or con-
genital mutations. Following clinical consultation and ob-
Large international projects aim to capture genome-wide taining informed consent, patients carrying familial muta-
relationships using large population samples and a variety tions associated with cardiovascular disease are recruited
of platforms and sequencing approaches. These include the for hiPSC generation (FIGURE 2). Subsequent cardiac spec-
1000 Genomes Project Consortium and the 100,000 Ge- ification of hiPSCs generates beating cells (24) which, de-
nome Project (100kGP) (282). De-identified genomic data are spite their relatively immature structural and electrophysi-
freely available and publicly accessible through resources such ological state (25, 184), have been shown to recapitulate the
as the ClinVar (http://www.nh.gov/clinvar/cbi.nlm.ni) or the clinical phenotypes observed in the donors. Disease model
ClinGen (http://www.clinicalgenome.org) databases. In addi- derivation has also been achieved in integration-free chem-
tion to the collection of samples, large efforts are underway to ically defined conditions, making them more clinically rel-
integrate different classes of omic information. These include evant (20). The growing body of research regarding innate

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1101


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

Innate Engineered Induced

Healthy donor Diseased donor Healthy donor Healthy donor

Reprogramming
to hiPSCs Genome Disease-
Healthy inducing
editing
conditions conditions

hiPSC
differentiation

Phenotypic
& functional
characterization

Structure Protein assay Electrophysiology Molecular pathway interrogation


FIGURE 2. hiPSC-based disease modeling approaches. hiPSC modeling of cardiovascular disease has mainly
been based on three approaches: 1) the generation of patient-specific disease lines using somatic cells from
patients with familial mutations associated with heart disease (innate model); 2) the introduction of pathogenic
mutations in healthy hiPSC-CMs via genome editing approaches; and 3) the induction of heart disease in
hiPSC-CMs from healthy individuals, either via drug treatment, pathogen infection, media supplementation, or
exposure to stress-inducing conditions (induced model).

models of cardiac diseases is summarized in TABLE 3 and and allele-specific RNA interference (183), were also able to
below. ameliorate the disease phenotypes. These findings were
among the first to provide proof-of-principle that hiPSC-
CMs can recapitulate the human pathophysiology of LQT
A. Cardiac Channelopathies
syndrome.
Among the most extensively researched diseases using
Other arrhythmia syndromes have also been studied, in-
hiPSC-based models are arrhythmic disorders caused by
cluding LQT syndrome type 3 (LQT3), which is caused by
mutations in cardiac ion channels, which are known as
gain-of-function mutations in the sodium ion channel-en-
cardiac channelopathies. This category of disorders in-
cludes congenital long QT (LQT) syndrome, catecholamin- coding gene SCN5A. These hiPSC-CMs showed markedly
ergic polymorphic ventricular tachycardia (CPVT), Timo- prolonged APD (due to decreased voltage-dependent inac-
thy syndrome, and Brugada syndrome, which are clinically tivation of Na⫹ channels) and late or persistent Na⫹ cur-
characterized by arrhythmias, ventricular fibrillation, sei- rents. These phenomena were reversed by a combination of
zures, and even sudden death (248). As summarized in increased pacing rate and NaV1.5 sodium channel block-
TABLE 3, characterization via MEA and patch-clamp elec- ade (e.g., with mexiletine). This treatment regimen closely
trophysiological techniques has enabled identification of mirrors clinical management of LQT3 (172, 175, 209,
prolonged field potential duration (FPD) and action poten- 264). Similarly, hiPSC-CMs generated from patients carry-
tial duration (APD) in hiPSC-CMs from patients with LQT ing mutations in the L-type calcium channel Ca(v)1.2 that
syndrome type 1 and type 2 loss-of-function autosomal are associated with LQT syndrome type 8, also known as
dominant mutations in potassium ion channels (184, 197). Timothy syndrome, showed pro-arrhythmia, prolonged
These abnormalities were found to be due to diminished IKs APD, excess Ca2⫹ influx, and abnormal Ca2⫹ transients.
or IKr currents resulting from trafficking defects in the pro- Roscovitine, a Ca(v)1.2 activator, ameliorated the electro-
teins responsible for the respective ion channel formation. physiological and Ca2⫹ handling perturbations observed in
␤-Adrenergic stimulation or pharmacological blockade of these hiPSC-CMs (311).
cardiac repolarization currents (e.g., with E4031) caused
LQT syndrome hiPSC-CMs to develop arrhythmias in the Finally, hiPSC-CMs from patients with CPVT types 1 and
form of early afterdepolarizations (EADs), which could be 2, carrying mutations in the ryanodine receptor (RYR2) or
corrected by treatment with ␤-blockers (e.g., propranolol cardiac calsequestrin (CASQ2) genes, showed pro-arrhyth-
and nadolol). Novel therapeutic avenues, such as small mol- mia in the form of delayed afterdepolarizations (DADs),
ecule-mediated modulation of chaperone proteins with N- which were reversed following administration of flecainide,
[N-(N-acetyl-L-leucyl)-L-leucyl]-L-norleucine (ALLN) (187) a Nav1.5 sodium channel and RYR2 blocker that is clini-

1102 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

Table 3. Innate and engineered models of cardiac disease


Model Class Disorder Gene Mutation Reference Nos. Key Phenotypes

Innate Arrhythmia LQT1 KCNQ1 R190Q 197 Protein trafficking defects


syndromes
Exon 7 del 171 1FPD and APD in atrial
and ventricular CMs
LQT2 KCNH2 A561T 184 Serve 2 or absence of Ik,
(LQT1) or Ik (LQT2)
17 Arrhythmias charactrized
by EADs
A614V 110 1Sensitivity to
proarrhythmic drugs
A561P 120 2repolarzation reserve
LQT3 SCN5A F1473C 264 Defective Na⫹ channel
caused by deficiency in
open-state inactivation
of the Na⫹ channel
V1763M 172
V240M 67 Enhanced late Na⫹
channel current (INaL)
R535Q
1644H 175 Delayed repolarization
and prolonged APD
CPVT1 RYR2 F2483I 68 1Diastolic [Ca2⫹]
322 2SR Ca2⫹content,
indicating Ca2⫹ leakage
S406L 121 1Frequency and duraton
of elemantary Ca2⫹
release events (sparks)
M4109R 109
P2328S 142 Arrhy thmias
characterized by
DADs, broad and
double-humped
transists
Q2311D 54 1Senstivity to
proarrhythmic drugs
R420Q 206 Immature ultrastructural
features
CPVT2 CASQ2 D307H 206 1Diastolic [Ca2⫹]
Oscillatory arrhythmic
prepotentials, DADs
207 Myofibrillar disarray,
1SR cisternae size,
1caveolae number
TS CACNA1C G406R 312 Irregular contractility and
electrical activity,
1APD
1Ca2⫹ influx
Cardiomyopathies DCM TNNT2 R173W 254 Abnormal Ca2⫹
regulation,
2contractility
301 Myofibrillar disarray-
RBM20 R636S 304
LMNA/C R225X 243 1Nuclear blebbing and
micronucleation
1Nuclear senescence
and apoptosis
DES A285V 275 Abnormal DES
aggregations
2 Max rate of Ca2⫹ re-
uptake and 2beating
rate
2Tolerance to inotropic
stress
HCM MYH7 R663H 147 1Cellular size and
sarcomere
disorganization
R442G 89 Contractile arrhythmias
Continued

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1103


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

Table 3.—Continued
Model Class Disorder Gene Mutation Reference Nos. Key Phenotypes

MYBPC3 G999-Q1004del 262 Dysregulation of Ca2⫹


cycling and 1[Ca2⫹]i
FXN Silencing 153 FRDA-associated HCM
1Rate of iron
accumulation
2Ca2⫹ uptake and
release kinetics
Disorganized and
fragmented
mitochondrial network
1ROS accumulation
LS PTPN11 T468M- 31 1Nuclear NFATC4
localization
163 1Expression of genes
associated with cardiac
hypertrophy
A324fs335X 32 1Lipogenesis and
apoptosis
ARVD PKP2 c.2484C⬎T 134 2PKP2 and 1PPAR-␥
gene expression
c.2013delC 2PKP2, plakoglobin, and
Cx43 protein density
c.1841T⬎C 170 2␤-Catenin activity
Desmosome
disorganization
DMD DMD exon 45-52del 164 Dystrophin deficiency
1Levels of resting Ca2⫹
Mitochondrial damage
and 1apoptosis
Structural HLHS Not identified Not identified 115 2Ability for cardiac
differentiation
73 2Myofibrillar organization
1Fetal gene expression
pattern
Calcium transient and
electrophysiological
abnormalities
Metabolic ALHD deficiency ALDH2 E487K 63 1Levels of ROS and toxic
aldehydes
1Cell cycle arrest and
activation of apoptotic
signaling
BTHS TAZ c.517delG c.517ins 289 1Levels of ROS,
2mitochondrial
function
1Sparse and irregular
sarcomeres, weak
contractility
Pompe disease GAA D645E 101
exon 18del 227 1Levels of glycogen,
1441delT defective respiration
Deficit of Golgi-based
protein glycosylation
c.796C⬎T 233 Ultrastructural
c.1316T⬎A aberrances, lysosomal
enlargement
Engineered Arrhythmias LQT1 KCNQ1 R190Q 290 Protein trafficking defects
G269S 1APD in atrial and
ventricular CMs
G345E Severe 2 or absence of
IKs (LQT1) or IKr (LQT2)
LQT2 KCNH2 A614V Arrhythmias
characterized by EADs
N996I 13
Continued

1104 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

Table 3.—Continued
Model Class Disorder Gene Mutation Reference Nos. Key Phenotypes

Cardiomyopathies DCM PLN R14del corrected to 130 Correction of:


WT
Ca2⫹ handling
abnormalities
Electrical instability
Abnormal cytoplasmic
distribution of PLN
protein
1Expression cardiac
hypertrophy molecular
markers
TTN W976Rfs 93 2Contractile
A22352fs performance due to
P22582fs A-band truncations
Sarcomere insufficiency
Impaired responses to
mechanical and ␤-
adrenergic stress
Attenuated growth factor
and cell signaling
activation
Metabolic BTHS TAZ c.517delG 289 1Levels of ROS,
c.517ins 2mitochondrial
function
Sparse and irregular
sarcomeres, weak
contractility

Together, Tables 3 and 4 provide a detailed outline of hiPSC-based models of cardiac disease published to date,
with correlation to the gDNA mutations studies, and the key phenotypes observed for each disorder. APD,
action potential duration; FPD; field potential duration; 1, increased; 2, decreased; EAD, early afterdepolar-
ization; DAD, delayed afterdepolarization; hiPSC, human induced pluripotent stem cell; CMs, cardiomyocytes;
SM, small molecule; GF, growth factor; SR, sarcoplasmic reticulum; VPA, valproic acid; ET-1, ETA-b, endo-
thelin receptor type A blocker; ROS, reactive oxygen species; CDI, Cellular Dynamics International; LQT1, 2, 3,
long-QT syndrome type 1, 2, 3; JLNS, Jervell and Lange-Nielsen syndrome;CPVT1, 2, catecholaminergic
polymorphic ventricular tachycardia type 1, 2; TS, Timothy syndrome; DCM, dilated cardiomyopathy; HCM,
hypertrophic cardiomyopathy; FRDA, Friedreich’s ataxia; DFP, deferiprone; LS, LEOPARD syndrome; ARVD,
arrhythmogenic right ventricular dysplasia; DMD, Duchenne muscular dystrophy; HLHS, hypoplastic left heart
syndrome; BTHS, Barth syndrome.

cally used against CPVT (109). Thapsigargin, a sarcoplas- athy (ARVC), LV noncompaction (LVNC), and restrictive
mic reticulum calcium ATPase pump inhibitor, also amelio- cardiomyopathy (RCM) (91). Most cardiomyopathies are
rated the CPVT phenotype in hiPSC-CMs by depleting in- characterized by disrupted sarcomeric alignment, a phe-
ternal Ca2⫹ stores. This supported the mechanistic nomenon known as myocardial disarray that is linked to
explanation that increased diastolic Ca2⫹ can lead to the deterioration in myocardial function, heart failure, and car-
development of arrhythmias. In a separate study, ultra- diac sudden death (111). However, each cardiomyopathy
structural characterization demonstrated that hiPSC-CMs has its own distinct pathophysiological features: DCM is
from CPVT1 patients also have enlarged mitochondrial, identified by ventricular dilation and systolic dysfunction;
glycogen, and lysosome clusters, as well as thinner Z-bands, HCM manifests as thickening primarily of the left ventric-
reduced number of sarcomeres per cell, and increased sar- ular wall; ARVC is distinguished by cardiac enlargement,
comere disarray (206). These phenotypes were more pro- aneurysm formation, and fatty infiltration of the ventricles;
nounced in CPVT2 hiPSC-CMs, demonstrating that hiPSCs and RCM manifests as atrial arrhythmias, abnormal right-
provide a setting for the investigation of the similarities and and left-sided filling pressures, and other clinical signs and
differences in the pathophysiological consequences of dif- symptoms characteristic of right ventricular failure (210).
ferent mutations.
hiPSC-CMs derived from patients with familial cardiomy-
B. Cardiomyopathies opathy have been shown to recapitulate the human heart
pathophysiology. For example, hiPSC-CMs carrying a
A second class of cardiac disorders that have been widely DCM-related cardiac troponin T mutation (TNNT2
studied using the hiPSC in vitro platform are cardiomyop- p.R173W) showed characteristic sarcomeric disarray and
athies. These include familial HCM, dilated cardiomyopa- other ultrastructural abnormalities such as scattered distri-
thy (DCM), arrhythmogenic right ventricular cardiomyop- bution pattern of Z bodies, as well as reduction in contrac-

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1105


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

tile force, altered regulation of Ca2⫹, and decreased toler- were evident following iron loading-induced stress. Treatment
ance to ␤1-adrenergic challenge (254, 301). These pheno- with molecules that are currently under clinical trial for
types were attenuated by clinically relevant ␤1-blockade FRDA, such as the iron chelator deferiprone (DFP), signifi-
(e.g., with metoprolol), or more experimental pharmaco- cantly suppressed ROS synthesis, modulated mitochondrial
logical inhibition of PDE2A and PDE3A (e.g., with Bay-60- stress, and improved electrical coupling during hiPSC-CM
7550 and milrinone), or transgenic overexpression of sar- contraction.
coplasmic reticulum Ca2⫹ adenosine triphosphatase
(SERCA2a). Microarray analysis in hiPSC-CMs that had Finally, hiPSC-CMs carrying ARVC-associated mutations
undergone SERCA2a gene therapy showed that Ca2⫹ sig- in desmosome proteins have also recapitulated disease pa-
naling, protein kinase A (PKA) signaling, and G protein- thologies (e.g., increased lipogenesis and apoptosis, and
coupled receptor (GPCR) signaling were mainly responsible desmosome disorganization) following induction of adult-
for the phenotypic rescue. It should be noted that the like metabolic energetics from glycolysis to fatty acid oxi-
R173W mutation identified in these DCM patients had not dation, therefore demonstrating the ability of hiPSC-CMs
been previously reported, and was discovered in this family to model adult-onset cardiac disorders (134, 170).
cohort following exome sequencing of the skin fibroblasts
used to generate hiPSCs. In separate studies, hiPSC-CMs Collectively, these studies not only demonstrate that hiPSC-
carrying DCM-associated mutations in the spliceosome CMs can serve as disease modeling and drug discovery plat-
RNA-binding motif protein 20 (RBM20), lamin A/C forms, but can also be used to assess the unique interactions
(LMNA/C), or desmin (DES) genes (243, 275, 304) also between individual patients’ genetic backgrounds and envi-
demonstrated abnormalities that closely recapitulate the ronmental factors, and serve as biomedical data interfaces
morphological and functional phenotypes of the affected to unravel complex molecular pathways implicated in cel-
human heart, as outlined in TABLE 3. lular dysfunction and human cardiac or syndromic disease.

hiPSC models of HCM associated with thick myofilament C. Structural Defects


myosin heavy chain 7 (MYH7) (89, 148) or myosin binding
protein C3 (MYBPC3) (262) mutations showed that in vitro-
Developmental heart defects such as hypoplastic left heart
derived hiPSC-CMs are significantly enlarged, as seen in the
syndrome (HLHS), characterized by severe underdevelop-
diseased human myocardium. hiPSC-CMs also demonstrated
ment of the left ventricle, are another class of cardiac disor-
increased myofibril content, contractile arrhythmias (DADs),
ders that have been modeled in hiPSC-CMs. hiPSCs from
Ca2⫹ cycling perturbations, and intracellular Ca2⫹ elevation,
HLHS patients were found to have a reduced ability to form
which were worsened by adrenergic stimulation (e.g., with
beating CMs in vitro, whereas hiPSC-CMs also demon-
isoproterenol) or environmental exposure to the potent vaso-
strated myofilament disorganization, induction of fetal
constrictor endothelin-1 (ET-1) (310). These disease pheno-
gene expression profiles (e.g., HIF1␣), Ca2⫹ handling ab-
types were reversed either by ␤-blockade (e.g., with propran- normalities, and increased susceptibility to ␤-adrenergic
olol or metoprolol), treatment with endothelin receptor type A stimulation and caffeine treatment (73, 116). Further stud-
blockers (e.g., BQ-123), or treatment with L-type Ca2⫹ chan- ies in hiPSC-CMs revealed that disease pathologies could be
nel blockers (e.g., verapamil). The latter observation provided associated with alternative Ca2⫹ release mechanisms from
mechanistic validation that Ca2⫹ handling defects and ele- the sarcoplasmic reticulum via the inositol trisphosphate
vated intracellular Ca2⫹ levels are underlying disease mecha- receptor rather than the ryanodine receptor, thus providing
nisms for HCM. Whole transcriptome sequencing and path- evidence that hiPSC-CMs can be used for mechanistic in-
way enrichment analysis in HCM hiPSC-CMs also showed a vestigations of developmental heart defects. Although no
widespread enrichment of genes associated with cellular pro- known mutations linked to HLHS were identified in these
liferation (e.g., WNT1), Notch signaling (e.g., DLL1/4), and studies, hiPSC-CMs offer an opportunity for further re-
FGF signaling (e.g., FGF8/FGFR4) which could serve as po- search regarding causative gDNA alterations.
tential therapeutic targets (89). HCM manifestations associ-
ated with the multi-organ condition known as LEOPARD
syndrome were likewise studied in a hiPSC-CM platform, D. Metabolic Disorders
showing that irregularities in RAS-MAPK signal transduction
are implicated in development of disease pathology (31). In Familial metabolic disorders, also known as inborn errors of
addition, hiPSC-CMs from patients with HCM associated metabolism (IEMs), represent more than 15% of monogenic
with the recessive neurodegenerative disorder Friedreich’s disorders (8). Initiating during early infancy or childhood,
ataxia (FRDA) showed an increase in reactive oxygen species over 40% of IEMs patients manifest a cardiovascular pheno-
(ROS) levels caused by silencing of the mitochondrial gene type that includes limited exercise capacity, HCM, DCM,
frataxin (FXN) (153). Additional disease phenotypes, includ- LVNC, RCM, and fatal arrhythmias (162). Due to the high
ing disorganization of the mitochondrial network, intracellu- dependence of cardiac muscle on aerobic metabolism (up to
lar iron accumulation, and abnormal Ca2⫹ handling kinetics 70% of energy in the heart muscle stems from oxidative me-

1106 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

tabolism) (85), defects in mitochondrial oxidative phosphor- bility towards ischemic heart damage and coronary artery
ylation (e.g., complex I, II, III, IV, and V disorders, Barth, disease (CAD), was associated with elevated accumulation
Leigh, and MELAS syndromes) or fatty acid ␤-oxidation (e.g., of ROS and toxic aldehydes (e.g., 4-hydroxynonenal;
CPT2, VLCAD, and CTD syndromes) are more frequently 4HNE) in heterozygous hiPSC-CMs. These pathological
associated with cardiac findings (19). Other major groups of phenotypes were connected with the induction of cell cycle
metabolic disorders associated with cardiomyopathy include arrest and activation of apoptotic signaling genes (e.g.,
lysosomal diseases (e.g., Fabry disease), glycogenesis-related JUN) and were aggravated during exposure to an ischemia-
disorders (e.g., Pompe and Danon diseases), organic acidurias, mimetic solution and hypoxic conditions. Alda-1-mediated
and glycosylation disorders. Although metabolic disorders ac- activation of ALDH2, treatment with the independent ROS
count for a minority of cardiomyopathy cases, identifying an scavenger catalase, or inhibition of the Jun regulator Jun
IEM as the underlying cause of disease has significant prog- NH2-terminal kinase (JNK) were all able to decrease path-
nostic and therapeutic implications (297). ological ROS levels. Therefore, the authors were able to
shed light onto the underlying molecular bases of a genetic
Regardless of the clinical avenues used for therapy, the mech- polymorphism often found in East Asians that is associated
anisms by which IEMs lead to cardiomyopathy remain poorly with adverse cardiac function in response to ischemia.
understood. To bridge this gap, hiPSCs have been used as
platforms to study the mechanisms and physiological pertur- VI. ENGINEERED DISEASE MODELS
bations underlying cardiomyopathy related to metabolic dis- THROUGH TARGETED GENOME
eases. Infantile-onset Pompe disease, an autosomal recessive EDITING
IEM caused by mutations of the lysosomal glycogen-hydrolyz-
Targeted gene editing has been historically achieved by homol-
ing enzyme acid ␣-glucosidase (GAA), is one of the disorders
ogous recombination (HR) (157); however, the low efficiency
studied in a hiPSC-CM setting. Pompe and control hiPSC-
of HR events in human cells impeded extended application of
CMs demonstrated no significant differences in contractile
this approach. Recent advances in endonuclease-based gene
strength, kinetics, or clearance rates of autophagosomes.
editing systems have transformed the field of genome engineer-
However, similar to the patients’ myocardium, Pompe hiPSC-
ing (46). Following discovery that induction of DNA double-
CMs had diminished GAA activity, lysosomal glycogen accu-
strand breaks (DSBs) increases the frequency of HR events by
mulation, and lysosome enlargement (227). MALDI-TOF-MS
several orders of magnitude, targeted nucleases such as ZFN,
analysis of N-linked glycans revealed that Pompe cardiomy-
TALEN, and CRISPR/Cas9 have emerged as the preferred
opathy could be caused by a glycan-processing abnormality,
methods for improving the efficiency of HR-mediated gene
as often seen in congenital glycosylation disorders with an modification (74). Once endonuclease enzymes induce a DSB
HCM phenotype, therefore providing mechanistic insight into in the DNA, the cell’s natural repair mechanism initiates one
the disease pathological mechanisms. Gene therapy by lentivi- of two different processes: 1) the more likely but also more
ral GAA transfer was able to reverse the disease-specific error prone nonhomologous end joining (NHEJ) pathway,
hiPSC-CM phenotypes (233). whereby the two broken strands are modified by either adding
or removing sheared nucleotides and then physically ligating
Another metabolic disorder to be studied in a hiPSC setting is the two strands back together; or 2) the more precise homol-
Barth syndrome (BTHS), a mitochondrial disorder caused by ogy directed repair (HDR) pathway, kinetically favored over
mutations in the tafazzin (TAZ) gene. Engineered muscular NHEJ only if a DNA strand with homology to the cut gDNA
thin film (MTF) chips (276) constructed from BTHS hiPSC- is present (199). This process can utilize an exogenously pro-
CMs had defined metabolic (e.g., excess accumulation of vided homologous sequence or plasmid as a template from
ROS), structural (e.g., sparse and irregular sarcomeres), and which a complementary strand is synthesized. The newly syn-
functional (e.g., weak contractility of engineered heart muscle) thesized strand then ligates with the original, resulting in a
abnormalities (289). These were reversed either by gene re- repaired double strand. Endonuclease-based techniques take
placement therapy or genome editing, demonstrating that the advantage of this repair system by providing a template of
TAZ mutation under investigation was necessary and suffi- semi-homologous sequence with specific and intended muta-
cient for these phenotypes. Therefore, the combination of tis- tions that will be copied into the cell’s DNA.
sue engineering, genome editing, and hiPSC technologies in
this study provided new insights into the pathogenesis of The next section of the review provides more details regard-
BTHS, delivering a biomedical platform to confirm the phe- ing novel genome editing technologies, by comparing the
notype-to-genotype correlation of a gDNA mutation. ZFN, TALEN, and CRISPR/Cas9 systems, and highlighting
the future research applications of these technologies.
Taking a step further to demonstrate the association of
gDNA alterations to disease manifestation, Ebert et al. (63) A. DNA Binding Designs
showed that the aldehyde dehydrogenase 2 (ALDH2) ge-
netic polymorphism E487K, which is found in nearly 8% of In addition to the induction of DSBs, current nuclease-based
the human population and is linked to increased suscepti- approaches distinguish themselves from previous gene editing

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1107


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

systems by the ease with which their DNA recognition do- ing. crRNA sequences are easy to design, making the CRISPR/
mains are customized, a feature that ultimately improves ge- Cas9 more versatile than either ZFN or TELEN tools.
nome editing efficiency (214). ZFN and TALEN technologies
share a mechanism where repeating domains in their protein- While it is tempting to further stratify ZFN, TALEN, and
based DNA binding modules recognize individual nucleotides. CRISPR/Cas9 by ranking them based on binding efficien-
The zinc finger domain, first discovered in 1997, is the core cies, it would be potentially misleading to do so. A clear and
component of a zinc finger protein (ZFP), which recognizes a fixed ranking system is impossible to construct because each
three nucleotide sequence. ZFNs typically comprise 3– 6 zinc system binds differently to different targets. Currently no
finger proteins and bind 9 –18 nucleotides of the target DNA. molecular basis explains this discrepancy. However, it has
ZFNs may be combined through a modular assembly to rec- been suggested that DNA methylation, chromatin state,
ognize nearly any genetic sequence; however, the codon-based genomic position, and DNA sequence may affect DNA
gene editing limits flexibility and the ability to potentially tar- binding and thus explain why different editing efficiencies
get any gDNA sequence. Additionally, even though ZFNs rely are reported for different targets (100).
on a very modular assembly, their production remains labor
intensive and time consuming (136). B. Off-Target Binding
Similar to the role of the ZFPs, the core TALEN domain Recent analysis of whole genome sequencing applied to
binds to DNA based on a protein-DNA interaction. The hiPSCs reveals minimal differences in off-target activity
TALEN binding domain is a sequence of 33–35 repeating among ZFN, TALEN, and CRISPR/Cas9 technologies
amino acids that forms a repeat-variable di residue (RVD). (244, 255, 281). In addition, the frequency of off-target
The 12th and 13th amino acids in each RVD, known as activity is typically lower than the rate of naturally accruing
hypervariable positions, are responsible for binding differ- mutations that result from culture maintenance (51). Stud-
ent nucleotides. Based on the affinity of A, T, G, and C to ies specific to TALEN and CRISPR/Cas9 identified signifi-
the respective RVDs NI, HD, NN, and NG, TALENs may cantly more mutations as a result of cell maintenance than
be utilized to recognize any sequence of DNA. This one-to- those causally linked to the application of gene editing
one interaction is a large advantage over ZFN in that it (106). Although some reports indicate high rates of off-
provides increased specificity and ease of customization target mutations, these numbers stem from analysis of
(83). However, like ZFNs, to target a new DNA sequence transgenic mammalian cell lines such as HEK293, and thus
one must reengineer the primary binding domains. Thanks cannot be put into context relevant to hiPSC culture and the
to intensive research in recent years, customized TALENS development of disease models (274). Nevertheless, off-tar-
can be constructed in ⬍1 mo, following minimally labor get binding remains a significant hurdle to clinical adoption
intensive procedures that also reduce or eliminate the num- of gene editing techniques. More research is required to
ber of cuts and ligations required, thus reducing the possi- develop improved computational algorithms able to iden-
bility of orientation error (59, 234). tify sequences with low risk of off-target binding, and to
predict off-target binding even when DSBs do not occur
The most prominent technology for modern genome editing is (291).
the CRISPR/Cas9 system, first reported in 1987. A form of the
CRISPR/Cas9 system can be found throughout bacteria and Recently, research has expanded beyond the optimization
prokaryotes. However, the most thoroughly researched and of binding domains to explore other creative approaches for
most applicable to DNA modification is the modular type 2 reducing off target mutations. For example, engineered nu-
CRISPR/Cas9 system found only in bacteria. Cas9 is an endo- clease mutations that induce DNA “nicks” or single-
nuclease specific to the bacteria Streptococcus pyogenes and is stranded breaks (SSBs) are advantageous because SSB cel-
the only nuclease involved in silencing of foreign DNA (117). lular repair circumvents NHEJ repair mechanisms, which
Within bacteria, different CRISPR systems exhibit slight mod- would normally increase the probability of mutagenesis.
ifications in length and DNA recognition, but all can be engi- Multiple reports identify that both ZFNickases and
neered through modular assembly of three main components: TALENickases induce fewer off-target mutations while
1) the nuclease that cuts DNA; 2) a specificity determining demonstrating equal if not better on-target mutation rates
RNA strand, known as the CRISPR RNA (crRNA); and 3) the (303). Likewise, nickase-based CRISPR systems (CRISPR/
trans-activating crRNA (tracrRNA) (118). Unlike the ZFN Cas9n) result in negligible off-target effects without a sac-
and TALEN recognition of DNA via protein interactions in- rifice of on-target mutation frequency (240).
volving complex ␣-helix recognition of nucleotide triplets or
repeating amino acids with hypervariable regions, the C. Genome Editing in hiPSC-Based Cardiac
CRISPR-Cas9 system binds to DNA in the same fashion as an Disease Modeling
RNA oligo (302). Twenty nucleotides on the crRNA bind with
the 20 nucleotides of the target DNA, recruiting the Cas9 In hiPSCs, genome editing has thus far been utilized for the
endonuclease, which on its own has no sequence-specific bind- correction of genetic disorders as mentioned earlier for

1108 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

BTHS (289). Karakikes et al. (130) also used genome edit- D. Future Directions of Genome Editing
ing to correct the PLN gene mutation R14del which has Technology
been associated with ventricular dilation, ventricular ar-
rhythmias, and heart failure. In this study, hiPSC-CMs The last decade of research has seen rapid progress in the
heterozygous for the R14del mutation exhibited Ca2⫹ han- field of genome editing. It is clear that off-target effects
dling abnormalities, electrical instability, abnormal cyto- exist, and resolving them will be important for future clin-
plasmic distribution of PLN protein, and increased expres- ical applications of this technology. As research continues
sion of molecular markers of cardiac hypertrophy, which to optimize current protocols, more creative approaches
were ameliorated following TALEN-based mutation cor- such as the use of recombinant fusion proteins provide new
rection. opportunities (75, 76, 189). Fusing nuclease DNA-binding
domains to alternate protein complexes can create genome-
Similarly, genome editing has been utilized for the gener- editing systems with reporter and therapeutic applications.
ation of hiPSC-based disease models, such as LQT syn- Depending on which protein complex is used to replace the
drome. In two studies by Wang et al. (290) and Bellin et normal endonuclease domain, ZFN and TALEN systems
al. (13), hiPSCs from healthy donors were engineered to can transcriptionally upregulate or downregulate genes to
introduce dominant negative mutations in the ion chan- alter entire genetic pathways, remodel chromatin networks,
nel genes KCNQ1 (p.R190Q, p.G269S, and p.G345E) or even act as reporters for gene activity.
and KCNH2 (p.A614V and p.N996I) into the safe har-
bor AAVS1 locus. Compared with isogenic controls, ge- A wide range of successful experiments have validated these
nome-edited lines displayed characteristic LQT syn- supplementary additions to the genetic toolbox and proto-
cols that are readily available through publicly available
drome phenotypes, which included significant APD pro-
repositories (e.g., Addgene). In the case of both ZFNs and
longation, as outlined in TABLE 3. These disease
TALENs, the Folk1 endonuclease domain is replaced by
phenotypes were corrected by drug treatment with the
transcriptional activators or repressor domains. Similar
L-type calcium channel blocker nifedipine, or the KATP
lists of protocols and gene regulatory activities are possible
channel opener pinacidil, therefore providing validation for CRISPR applications, where the Cas9 must be rendered
that engineered isogenic hiPSC lines can be used for dis- catalytically inactive (CRISPR/Cas9d). This allows other
ease modeling and drug testing. protein complexes to be fused with the CRISPR/Cas9d,
which is guided to the target sequence via the guide RNA,
In combination with genome editing, hiPSC technology has and can consequently provide targeted gene inhibition
also been utilized in functional genomic screening for iden- (CRISPRi) (149), activation (CRISPRa) (77), as well as spa-
tification of the roles essential genes play in specific cellular tiotemporal (204) or conditional (306) gene regulation with
process. For example, Hinson et al. (93) were able to gen- a higher specificity and reproducibility compared with
erate cardiac microtissues using hiPSC-CMs to evaluate the other technologies such as RNA interference (RNAi) or
pathogenic mechanism of truncating titin gene variants Cre-LoxP systems. Modified CRISPR/Cas9 systems can
(TTNtvs), which are commonly associated with DCM. also inflict alterations of single or multiple genes at a time.
TTNtvs engineered into healthy hiPSCs resulted in micro- Notably, some groups have synthesized CRISPR/Cas9 li-
tissues with diminished contractile abilities (e.g., reduced braries consisting of 70,290 guides targeting all human Ref-
contractile force) and impaired responses to mechanical Seq coding isoforms (138), demonstrating that this technol-
and ␤-adrenergic stress. The investigators also found re- ogy constitutes a powerful genetic perturbation tool, with
duced expression of growth factors (e.g., TGF␤1, VEGF, wide applications in precision medicine.
HGF, EGF, and FGF2), hypoxia regulators (e.g., HIF1A
and EPAS1 or HIF2A), and MAP kinases (e.g., MEK and VII. INDUCED MODELS OF CARDIAC
ERK). VEGF supplementation of hiPSC-CM culture media DISEASE
ameliorated these phenotypes. The pathogenic mechanisms
of TTN-induced DCM were identified as the disruption of Aside from genetic composition, several environmental
critical linkages between sarcomerogenesis and adaptive re- factors (e.g., diet, stress, drugs, or pathogens) as well as
modeling when TTNtvs are present in the A-band domain secondary factors that occur due to failure in other or-
of this protein that is responsible for thick filament-binding. gans can lead to cardiac disease, or affect disease progres-
Another interesting finding of this study was that hiPSC- sion and response to pharmacotherapy in heart failure
CMs carrying engineered A-band TTNtvs showed less se- patients (179) (see TABLE 4). Factoring in the effects of all
vere phenotypes compared with patient-specific hiPSCs car- these parameters is an important part of precision medi-
rying the same mutations, thereby raising the possibility cine. Very few studies to date, however, have established
that genetic background can influence the functional sever- direct mechanistic links regarding the complex effects of
ity of TTNtvs. This observation has important functional environmental and secondary factors on heart failure.
implications for precision medicine. hiPSC-CMs that lack known familial or engineered

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1109


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

Table 4. Induced models of cardiac disease


Reference
Model Class Disorder Stimulus Nos. Key Phenotypes

Induced Drug Myopathy Doxorubicin 21, 96 1Contractile ability


Cardiac troponin T and lactate
dehydrogenase leakage
Pathogen Myocarditis CSV-B3 virus 239 Irregular or ceased beating
Calcium handling
abnormalities
1Cell death
Secondary Hypertension ET-1 2 Investigation of differentially
expressed miRNAs and
mRNAs upon induction of
hypertrophy
T2D Glucose, ET-1, cortisol 61 Myofibrillar disarray
Lipid accumulation
1Oxidative stress
Hypertrophy
1Cytosolic [Ca2⫹]

Together, Tables 3 and 4 provide a detailed outline of hiPSC-based models of cardiac disease published to date,
with correlation to the gDNA mutations studies, and the key phenotypes observed for each disorder. ET-1,
endothelin-1; CSV-B3, Coxsackievirus B3; T2D, type 2 diabetes.

gDNA mutations can be exposed in vitro to a variety of B. Drug Induced


environmental conditions (e.g., ischemic injury, hypoxic
conditions, dietary supplements, electromechanical Pluripotent stem cell-derived CMs have been shown to re-
stress, and infectious agents) to enable determination of spond appropriately to a wide variety of drug classes, in-
their effects on CM function. Existing studies of induced cluding ␤-adrenergic receptor agonists and class I, II, III,
hiPSC-models are discussed below. and IV anti-arrhythmic agents (186, 314). As discussed in
this review, clinically relevant and experimental drugs have
also been used in vitro for the recovery of diseased pheno-
A. Pathogen Induced types in models of cardiovascular disease. However, only a
few studies have investigated the effects of known cardio-
hiPSC-CMs can provide a suitable platform for the investi- toxic drugs in a pluripotent stem cell-based platform that
gation of pathological mechanisms and therapeutic targets lacks known mutations. One example was given by Hol-
for life-threatening infectious diseases affecting the heart, mgren et al. (96), who found severe in vitro abnormalities
such as Chagas cardiomyopathy caused by the T. cruzi try- following a 2-day treatment with varying doses of doxoru-
panosome (176), or viral myocarditis caused by the B3 bicin, a chemotherapeutic agent indicated for the treatment
strain of coxsackievirus (CVB3) (5). In a recent study, of a variety of cancer types (e.g., leukemia, lymphomas, and
Sharma et al. (239) demonstrated that hiPSC-CMs lacking solid tumors) that is also associated with severe cardiotox-
icity, especially in children and adolescent patients (36).
any known mutations associated with cardiac disease can
CMs treated with the drug exhibited concentration-depen-
be infected by CVB3 particles and undergo pathological
dent alterations in cellular morphology, reduced contractile
phenotypic changes such as increasingly irregular beating
ability, lactate dehydrogenase leakage, cardiac troponin T
patterns, increase in Ca2⫹ transient duration and time to
release, and increased apoptosis. Transcriptome profiling
transient peak, and eventual beating arrest (239). The au- performed by RNA-seq revealed that differentially ex-
thors also showed that these phenotypes were ameliorated pressed genes due to doxorubicin exposure were associated
by treatment with antiviral drugs (e.g., interferon-␤1, riba- with cellular defense mechanisms such as p53 signaling
virin, pyrrolidine dithiocarbamate, and fluoxetine), which (e.g., TP53I3), DNA damage response (e.g., GADD45A),
acted by triggering viral RNA and protein clearance path- and cellular senescence and response to stress (e.g.,
ways and reducing the proliferation capacity of the CVB3 HIST1H4H). These genes could, therefore, serve as sensi-
virus. This study demonstrated that hiPSC-CMs can be used tive novel biomarkers for doxorubicin-induced toxicity in
to predict antiviral drug efficacy and serve as a sensitive human CMs. In a separate study, Burridge et al. (21) dem-
platform to screen novel antiviral therapeutics for their ef- onstrated that doxorubicin treatment caused higher suscep-
fectiveness in a high-throughput fashion. tibility to cell death, metabolic dysfunction, and calcium

1110 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

handling abnormalities in hiPSC-CMs from breast cancer RNA-sequencing gene expression data showed significant
patients who experienced doxorubicin-induced cardiotox- upregulation in metabolic genes involved in the tricarbox-
icity (DIC) in the clinic compared with hiPSC-CMs from ylic acid (TCA) cycle (e.g., PDK1), mitochondrial electron
breast cancer patients who had not experienced DIC. These transport chain (e.g., CYCS), glucose metabolism (e.g.,
data support that hiPSC-CMs are a suitable platform for ENO2), cell adhesion (e.g., integrins), and extracellular
identifying patient-specific predilection to drug-induced matrix deposition (e.g., collagens), whereas downregula-
cardiotoxicity. tion was observed in genes related to protein synthesis and
the cellular response to dysfunctional protein production
(e.g., ATF4 and CHOP10). This secondary cardiomyopa-
C. Secondary Cardiomyopathy thy model was consistent with one generated using hiPSC-
CMs from patients with type II diabetes (T2D), therefore
Cardiomyopathy arising as a secondary feature to medical lending support to the hypothesis that clinical chemistry can
conditions initiated at a different organ has also been the induce a phenotypic surrogate of diabetic cardiomyopathy.
subject of hiPSC-based studies. For example, induced car- The use of NPPB-based high-throughput phenotypic assays
diac hypertrophy was developed in hiPSC-CMs following enabled the investigation of therapeutic targets by screening
5-day exposure to endothelin 1 (ET-1), a potent vasocon- a library of 480 compounds. Beneficial molecules included
strictor peptide secreted by endothelial cells which leads to regulators of Ca2⫹ homeostasis, such as inhibitors of volt-
pulmonary hypertension (39) and also has known implica- age-gated Ca2⫹ channels (e.g., fluspirilene), molecules that
tions in biochemical and structural remodeling of the heart deplete intracellular Ca2⫹ stores (e.g., thapsigargin), and
(30). Mechanistic investigation performed by high- inhibitors of Ca2⫹-regulated proteins such as calmodulin
throughput transcriptome analysis identified a pathological (e.g., W7). The use of Na⫹ and K⫹ channel blockers (e.g.,
increase in the expression of fetal genes (e.g., ACTA1, phenamil and penitrem A), which reduced the frequency of
NPPA, and NPPB). Pathological upregulation of NPPB CM Ca2⫹ transients, also showed a therapeutic benefit.
was also confirmed at the protein level, and served as the
basis for development of a 384-well HCI phenotypic screen- Overall, by combining the hiPSC technology with high-
ing tool for the assessment of compounds suspected to tar- throughput screening assays, these studies demonstrate a
get the NPPB pathway, and therefore modulate the hyper- close correlation between hiPSC-CMs and human heart or
trophic response. Several compounds, including the Ca2⫹ animal models, provide mechanistic insight into the patho-
channel blocker verapamil, were shown to reverse the ef- logical gene regulatory pathways of HCM and T2D devel-
fects of cellular hypertrophy induced by ET-1. Other bene- opment, and demonstrate nicely that hiPSCs are valuable
ficial compounds included BEZ-235 (a dual PI3K-mTOR platforms in predictive toxicology and safety pharmacology
inhibitor), fenofibrate (a PPAR␣ activator), and SAHA (a applications that can benefit the early phases of drug dis-
broad-spectrum histone deacetylase inhibitor). covery and development.

In a separate study, microRNA (miRNA) sequencing


showed that ET-1 treatment in hiPSC-CMs leads to differ- VIII. PATIENT RECRUITMENT VERSUS
ential expression of 250 known (e.g., miR-23a-3p and miR- GENE EDITING
208a-3p) and 34 novel miRNAs, the majority of which
were associated with corresponding mRNA expression Recent improvements in gene editing may supplement the
changes (2). Some of the complex RNA regulatory mecha- traditional system of patient recruitment that identifies and
nisms associated with cardiac hypertrophy were involved in compares a group of diseased individuals to a control group
cardiovascular development (e.g., MEF2C), cell prolifera- that lacks disease phenotypes (FIGURE 2). This cause-and-
tion and transformation (e.g., MYC, FOS, TGFB2, and effect based discovery is hampered by multiple issues, such
TGFBR3), and mitogen-activated protein kinase signal as the possibility that phenotypic differences between pa-
transduction (e.g., MAP3K9 and MAP2K6). Differentially tient and control groups are not due to disease-causing mu-
expressed miRNA-mRNA pairs were consistent with sev- tations, but arise due to other differences in genetic back-
eral animal models of cardiac hypertrophy (28, 102). In ground (TABLE 5). To reduce the genetic background vari-
addition, bioinformatics principal component analysis ability between patient and control cells, hiPSC lines
(PCA) comparing in vitro hiPSC-based findings to human generated from family members of the patient unaffected by
myocardial biopsies from unrelated patients with left ven- the disease have been used (148, 184, 254). However, this
tricular hypertrophy detected overlapping expression approach can limit, but not abolish, genetic background
changes between the two groups. variability, given that even siblings or parent-child pairs
only have 50% genetic overlap. Gene editing techniques
Induced type II diabetic cardiomyopathy was also modeled circumvent this limitation, but present other challenges
in hiPSC-CMs following prolonged exposure to “diabetic such as off-target effects and clonal heterogeneity (180),
milieu” consisting of a combination of glucose, ET-1, and which need to be sufficiently addressed to allow confirma-
cortisol (61). Gene set enrichment analysis (GSEA) using tion of casual relationships between the underlying genetics

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1111


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

Table 5. Comparison of hiPSC-based disease modeling approaches


Innate Engineered Induced

⫹ Disease modeled against patients’ genetic 2Need for patient recruitment Enables modeling of noncongenital
background conditions
Isogenic disease and control lines
⫺ Patient recruitment difficult Time consuming Induced conditions may not accurately
recapitulate disease
Nonisogenic disease and control lines Costly
Potential risk for genomic instability

The advantages (⫹) and disadvantages (⫺) of innate, induced, and engineered hiPSC-based disease models. 2,
Reduced.

and disease phenotypes. To limit the effects of clonal vari- drugs. Human genetic variations can result from SNPs, in-
ation, some studies have attempted to generate iPSCs in dels, or duplication of gDNA sequences. SNPs are by far the
bulk culture (55, 298). More recently, it has also been most common type of variation, with more than 14 million
shown that underlying genetic background variation is re- SNPs identified in the human genome (173). Over 60,000 of
sponsible for most heterogeneity between hiPSC lines, while these SNPs are located within gene coding regions, and it esti-
cell type of origin and clonality have minor effects on hiPSC mated that more than 90% of human genes contain at least
gene expression and methylation profiles (27, 231, 232a). one SNP. Genetic polymorphisms of proteins involved in
pharmacodynamics and pharmacokinetics are known to be
Another problem that restricts the paradigm of patient recruit- important determinants of individual variability in drug effi-
ment involves patient and family consent. Not only must a cacy. Over 30 drugs are currently prescribed only under ful-
patient fit a correct disease profile, but the individual and their fillment of a genetic testing prerequisite due to their associa-
family must consent to donate human tissue. As a way to tion with deleterious gDNA polymorphisms. Such informa-
circumvent these problems, ZFNs, TALENs, and CRISPR/ tion is shared through large publicly available (e.g.,
Cas9 genome editing tools may be applied to create control PharmGKB) or private (e.g., PGMD) databases, with the aim
groups from diseased cell lines, and vice versa. This reduces the of contributing towards more effective, and safer medications
effort needed to recruit additional patients. Nevertheless, even and doses that will be tailored to a person’s genetic make-up.
this approach does not account for the possibility that multiple
polymorphisms may contribute towards a disease phenotype,
or address the fact that some disorders and clinical abnormal- A. Examples of Known Polymorphisms
ities are not yet correlated to any known mutations. Implicated in Drug-Induced Toxicity

It is clear that there are roles for both patient recruitment One example of a pharmacogenomic study focused on the
and genome editing, and it is becoming the general consen- therapeutic efficacy of simvastatin, a HMG CoA reductase
sus that both approaches should complement each other to inhibitor with cholesterol-lowering abilities that is used to
establish large biorepository banks that can serve as imme- lower the risk of stroke, heart attack, and other heart com-
diate resources for the academic and pharmaceutical com- plications in people with diabetes or coronary heart disease.
munities (299). This study involved a cohort of 156 human subjects with
low-density lipoprotein (LDL) cholesterol levels of 160 mg/
dl, who were treated with simvastatin at varying doses (40,
IX. PHARMACOGENOMICS 80, and 160 mg/day) over the period of 6 weeks. Simvasta-
tin treatment led to reduction of LDL cholesterol by 41, 47,
When a patient is administered a new drug, three outcomes and 53%, respectively, therefore providing strong evidence
are possible: 1) the patient experiences a beneficial effect in that this drug has a high therapeutic value in reducing LDL
relation to the condition they are being treated for, 2) the cholesterol (49). However, a small subset of the cohort (⬃5%)
patient’s condition does not improve, or 3) the patient ex- showed little to no reduction in LDL cholesterol levels, even at
periences adverse cytotoxic effects or even death. In many 160 mg/day. Furthermore, 0.7 and 2.1% of patients devel-
cases, the same medication can elicit any of these three oped liver toxicity, as measured by elevated activity of alanine
responses in different patients, and our understanding of aminotransferase in the plasma, at the 80 and 160 mg/day
the parameters that influence this outcome is greatly lack- doses. Importantly, one subject developed myopathy (0.7%)
ing. The field of pharmacogenomics aims to bridge this gap, at the 160 mg/day dose. This variation in clinical outcome was
by combining pharmacology and genomics to determine later linked with genetic polymorphisms in the HMG CoA
how individuals’ genomic variation can affect responses to reductase gene HMGCR (e.g., p.A12T and p.T29G), as well

1112 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

as in the solute carrier drug transporter SLCO1B1 (e.g., ation. In a study that generated a library of hiPSC-CMs
p.V174A), and the ATP-binding cassette transporter ABCG2 from healthy patients as well as patients carrying muta-
(e.g., c.421C⬎A) genes that have varying frequencies among tions related to LQT syndrome, DCM, or HCM, it was
different ethnic groups. The latter genes are known to regulate found that LQT and HCM hiPSC-CMs were more sus-
efflux of statins and statin metabolites and, therefore, contrib- ceptible to development of cardiotoxicity (e.g., EADs and
ute to the variability in efficacy and side effects of cholesterol- DADs) compared with DCM and control hiPSC-CMs
lowering drugs such as simvastatin, pravastatin, or rosuvasta- when treated with cisapride (161), a gastroprokinetic
tin (35, 200, 273). agent that was voluntarily withdrawn from the United
States market in 2000 due to arrhythmic and sudden
Another example is concerned with drug-induced LQT syn- cardiac death episodes in patients with preexisting heart
drome. In a cohort of 98 patients with drug-induced LQTS, conditions such as LQT syndrome and heart failure
the KCNE2 variant c.T8A, which is found in ⬃1.6% of the (223). In a separate study, Braam et al. (17) observed that
general population, was linked to adverse QT interval prolon- two potent blockers of the IKs current (HMR1556 and
gation following administration of trimethoprim/sulfame- JNJ303) were cardiotoxic in LQT hiPSC-CMs, but not in
thoxazole for the treatment of urinary tract infections (237). controls. These studies have important clinical implica-
Screening for KCNE2-blocking activity is now routinely per- tions showing that genetic predisposition to toxicity is
formed during the development process of any new drug to reflected in disease-specific hiPSC-CMs and may predict
predict any risk for drug-induced QT prolongation that could adverse drug responses more accurately than tests in
lead to sudden death in humans. However, testing is typically transgenic lines overexpressing a single ion channel or
performed in transgenic cell lines overexpressing KCNE2, even tests using a single control hiPSC line.
which lack the functional complexity of human CMs such as
the interaction between opposing ion currents, as well as the
ability to reflect deleterious effects associated with SNPs and X. POTENTIAL APPLICATIONS OF hiPSCs
other types of individualized gDNA variation. IN PRECISION MEDICINE

Patient-specific gDNA variation has also been shown to


affect cardiotoxicity induced by the drug doxorubicin, an
A. Safety and Efficacy Assessment of Novel
anthracycline antibiotic with broad antineoplastic activity Compounds in Preclinical Trials
which is currently one of the most common and most effec-
tive chemotherapy agents (265). Despite its anti-cancer ef- Despite strict guidelines by regulatory agencies, several
ficacy, doxorubicin treatment is associated with fatal heart drugs continue to present adverse side effects subsequent
failure in 5–50% of patients (depending on administered to their release to the clinic, with deleterious cardiovas-
dose) that was not predicted in preclinical drug-testing cular events (e.g., torsades de pointes, ventricular tachy-
stages (113). Increased susceptibility to cardiotoxicity has cardia, ventricular fibrillation, and sudden cardiac ar-
been linked to polymorphisms in genes related to mitochon- rest) being implicated in 28% of drug withdrawals (16,
drial function (e.g., NADPH oxidase subunits) (283, 300). 86). For example, milrinone, a phosphodiesterase-4
Specific examples include the 212A⬎G variant of the gene (PDE4) inhibitor, is an inotropic drug thought to have
encoding the NCF4 subunit of NADPH oxidase (230), and great clinical promise against heart failure, but was
the rs17863783 variant in the UDP glucuronosyltransferase found during clinical trials to be associated with increase
UGT1A6 (284). Interestingly, some SNPs found in efflux in mortality and lack of efficacy due to calcium overload
transporters (e.g., ABCB1/MDR1, ABCC2, ABCC3, and DADs in some patients (69). Cardiotoxicity has been
ABCG2, and RALBP1) improve doxorubicin pharmacoki- observed not only with drugs targeted to act on the heart,
netics/clearance and have been shown to be cardioprotec- but also with drugs aimed at other organs. For example,
tive against this drug (271). Further study regarding the anthracycline chemotherapy agents (e.g., doxorubicin,
cardiotoxic or cardioprotective effects of such variants in daunorubicin, idarubicin, and epirubicin) have been fre-
patient-specific or engineered hiPSC models could lead to a quently linked to cardiac complications such as arrhyth-
deeper understanding of the genetic determinants of chemo- mias, or dilated cardiomyopathy which develop either
therapeutic cardiotoxicity. acutely or chronically (216, 285).

It is possible that unanticipated drug-induced cardiotoxic-


B. Use of hiPSCs in Pharmacogenomic ity may be in part attributed to the lack of appropriate
Investigations preclinical screening platforms. Current drug testing is reli-
ant on preclinical evaluation using human or animal trans-
Preclinical drug testing in hiPSC-CMs can circumvent the genic cell lines [e.g., Chinese hamster ovary (CHO) cells and
limitations of drug testing in transgenic lines (196). Al- human embryonic kidney (HEK) cells overexpressing the
ready, hiPSC platforms have been utilized to highlight human ether-à-go-go-related gene (hERG)], followed by in
differences in drug response underlined by genomic vari- vivo evaluation in small and large animal models (99, 232)

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1113


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

(FIGURE 3). Despite having several advantages, including mals and humans. For example, mice have a heart beat rate
ease of maintenance and simplicity in developing assay for that is eight times faster than that of humans (500 vs. 60
transgenic lines and the ability to study systemic effects for beats/min), and cardiac repolarization in murine CMs relies
animal models, these platforms also have several draw- primarily on Ito, IK,slow1, IK,slow2, and ISS ion currents,
backs. Transgenic lines are only engineered to overexpress whereas in humans, repolarization is mostly dependent on
certain human proteins (e.g., the IKr protein hERG) and the potassium channels IKs and IKr (226). Calcium handling
lack the structural and functional complexity of relevant (e.g., phospholamban; PLN), myofilament (e.g., MYH6/
human cell types (218). For example, transgenic lines over- MYH7), and surface (e.g., SIRPA) proteins also have differ-
expressing a single ion channel failed to predict the false- ent expression patterns and pathophysiological roles in
positive outcome related to the drug verapamil, because mouse versus human hearts. As an example, while trunca-
while it blocks outward ion flow through the rapid delayed tion mutations have been shown to result in considerably
rectifier current, IKr, it also blocks inward ion flow through reduced myocardial PLN protein content and loss of PLN
the L-type calcium channel ICa-L, therefore producing a neu- inhibition of SERCA2a, leading to development of heart
tral outcome in functional CMs (191). Notably, Navarette failure and early mortality in homozygous humans, in mice
et al. (203) showed no cardiotoxicity in response to vera- PLN deficiency enhances myocardial inotropy and lusi-
pamil in hiPSC-CMs. In addition, these transgenic immor- tropy without adverse effects (87). Large animals such as
talized lines accumulate several karyotype aneuploidies that rats, pigs, sheep, or dogs have hearts with closer cardiac
may alter their response to drugs (253, 256). As even SNP electromechanical properties to those of humans, but are
changes are known to contribute to variability in drug re- more difficult to handle, and more expensive to maintain in
sponse (253), aneuploidies causing whole gene duplications animal facilities. In addition, they are known to have differ-
or deletions as seen in transgenic lines may significantly ent drug dose sensitivity compared with humans. For exam-
alter recorded phenotypes. ple, relative to humans, canines can tolerate up to 100-fold
higher concentrations of chemotherapy agents such as thio-
In vivo testing in small and large animal models is also TEPA, myleran, actinomycin D, mitomycin C, mithramy-
limited by significant interspecies variation between ani- cin, and fludarabine (220). It is estimated that animal mod-

Predicted Paradigm Shift in Clinical Trial Model

Transgenic lines
Traditional model

Costly
Slow
Small animals
Low predictive
value
Large animals

Selection of therapeutic
Pre-clinical testing Clinical testing
molecule
New model

Fast

Increased
confidence prior to
hiPSC-CMs from patients with diverse costly clinical trials
ethnicity, gender, cardiovascular history

FIGURE 3. Use of hiPSCs to transform the current clinical trial model. Of great importance is the application
of hiPSCs in drug discovery and toxicology testing as a part of clinical trials. In the current drug development
model, novel compounds are initially tested on human or animal transgenic lines (e.g., HEK and CHO) which not
only lack the complexity of functionally relevant human cell types, but also carry several aneuploidies that may
affect their response to drugs. This often results in false-positive or false-negative data. In vivo testing in small,
and later on, large animal models is also an FDA requirement prior to approval of novel compounds for human
clinical testing. Although this allows for systemic characterization of drug function, the vast species differences
between animal models and humans often lead to the release of cytotoxic compounds to the clinic. Introduction
of hiPSCs to the current drug development model will allow testing of novel compounds on functionally relevant
human cells derived from individuals with diverse ethnicities, gender, and disease history (e.g., cardiovascular
disease). Preclinical drug testing against a diverse human genetic background may prevent the costly release
of cytotoxic drugs to the clinic, and will enable informed decisions about drug prescriptions based on gender,
ethnic group, disease status, and other relevant categories.

1114 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

els have a combined predictability of human behavior of dicates that hiPSC-CMs can offer the pharmaceutical indus-
only 10% (185, 238). try a powerful, human, and physiologically relevant tool for
validating new drug candidates in early drug development
In contrast to transgenic and animal platforms, in vitro stages (65).
derived hiPSC-CMs display many of the complex charac-
teristics associated with in vivo development of the heart,
including gene and protein expression, ion channel forma- B. Clinical Management
tion, electrophysiological responsiveness, calcium han-
dling, and excitation-contraction coupling (320). Although Another goal of precision medicine is to enable physicians
their current relatively early maturation state warrants ad- to prescribe appropriate medications at the right dose and
ditional attempts to drive further maturation, hiPSC-CMs schedule for individual patients to achieve maximal thera-
provide an excellent platform to further improve preclinical peutic benefit with minimal, tolerable adverse effects (133).
drug testing (FIGURE 3). Beyond their properties as a func- In current clinical protocols, clinicians typically rely on the
tionally relevant human platform for preclinical drug test- patient’s history and test indications to estimate the best
ing, the introduction of hiPSCs to the current drug develop- pharmacotherapy, which can be conditional upon careful
ment model may allow testing of novel compounds on hu- subsequent monitoring of patients to assess the effective-
man cells which will be derived from individuals with ness and toxicity of the drug. Often, the clinician may pre-
diverse ethnicities, gender, and disease history (e.g., cardio- scribe several different drugs to identify the optimum treat-
vascular disease). This will take into account the expanding ment for each patient (FIGURE 4). During this lengthy pro-
role of pharmacogenomics in the management of cardiovas- cess, patients may experience unwanted side effects, and
cular disorders (313). Preclinical drug testing against a di- their condition may deteriorate. Furthermore, side effects
verse human genetic background has the potential to refine may be induced by polypharmacy (i.e., the use of multiple
drug response-to-genotype correlations, prevent the release medications by a patient). Avoidable adverse drug events
of cytotoxic chemicals to the clinic, and enable informed are among the most serious consequences of inappropriate
decisions about drug prescriptions based on gender, ethnic drug prescribing (219). However, effective protocols for
group, disease status, etc. A growing body of evidence in- predicting such events are lacking. A hiPSC-based model for

Predicted Paradigm Shift in Drug Treatment

Patient history
Traditional model

Slow
Clinical examinations
Ineffective

Serial testing of multiple drugs

Diseased patient Investigation of possible therapies Selection of best drug

Patient-specific
New model

hiPSC-CMs Fast

Selection of
optimum drug
treatment
Drug screen

FIGURE 4. hiPSC-based personalized medicine as a novel model for clinical pharmacotherapy. The hiPSC field
is growing at an unparalleled pace, with the promise that these cells will become major tools in the advance-
ment of personalized medicine. When treating a patient, clinicians nowadays typically rely on the patient’s
history and clinical test indications to choose the presumed best pharmacotherapy. Since there is limited
predictive confidence in these indications, the clinician may need to prescribe several different drugs before
they can identify one that best suits each patient. During this sometimes lengthy process, the patient may
experience unwanted and potentially serious side effects, and their condition may deteriorate. In contrast, a
hiPSC-based model for personalized medicine may allow safe and patient-specific testing of several drugs on
hiPSC-differentiated derivatives (e.g., cardiomyocytes), and a pharmacogenomics analysis using hiPSC-derived
cells may enable precise prediction of the patient’s response to each drug. This paradigm-shifting model of
clinical treatment could vastly increase our confidence in the prescription of appropriate medication, saving
patients from dangerous side effects or toxicities, and optimally addressing their disease symptoms.

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1115


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

precision medicine has the potential to provide patient-spe- Similar principles can be applied to disease prognosis by
cific information about drug response by taking into ac- utilizing patient-specific hiPSC derivatives to enable exten-
count individualized information such as genomic, tran- sive phenotypic screening and early prediction of disease
scriptomic, metabolomic, pharmacokinetic, and pharmaco- outcome and severity (FIGURE 5). Whereas current clinical
dynamic profiles that are reflected in hiPSCs. This may protocols for prognosis rely on family history and long-
allow safe and patient-specific laboratory-based testing of term monitoring, hiPSCs may provide a fast track to diag-
several drugs on hiPSC differentiated derivatives (e.g., nosis in certain clinical scenarios, thus enabling implemen-
CMs) to enable precise prediction of the patient’s response tation of prevention or early treatment plans that lead to the
to a drug or to polypharmacy, as well as to guide the dosing best outcome.
and frequency of drug administration. Overall, this model
of clinical treatment could provide increased confidence in One limitation towards clinical applications of hiPSCs is the
the prescription of appropriate medication in the future, time required (⬃3 months) to reprogram somatic cells to
which could protect patients from side effects or toxicities, hiPSCs and subsequently differentiate them to functional
and optimally address their disease symptoms. cell types, such as CMs. Optimization of differentiation
protocols has already shortened the time needed for hiPSC
In a hiPSC-based study in which the proband had a de novo differentiation to CMs from ⬃18 –25 to ⬃9 –11 days (24).
SCN5A LQT3-associated mutation (F1473C) and a poly- As further improvements in this field are under develop-
morphism (K897T) in KCNH2, the gene associated with ment, large-scale bio-repositories of both hiPSCs and their
LQT2 syndrome, biophysical and molecular analysis of ion differentiated derivatives provide an optimal way to reduce
channels expressed in hiPSC-CMs demonstrated that phe- the time required to obtain patient-specific cells for func-
notypic abnormalities (e.g., arrhythmias) observed in vitro tional assays. It is also possible that hiPSC derivation and
were attributed to the SCN5A mutation, whereas the bio-banking could become routine, to enable population-
KCNH2 polymorphism was not a contributing factor to wide individualized medicine in the future.
disease severity (264). The abnormally augmented INaL cur-
rent was corrected either by faster pacing of hiPSC-CMs or
treatment with the Na⫹ channel blocker mexiletine. There- XI. CONCLUSIONS AND FUTURE
fore, in vitro testing in hiPSCs was able to provide func- PERSPECTIVES
tional information as to the mechanisms of disease, indicat-
ing that Na⫹ channel blockers, rather than K⫹ channel Precision medicine approaches in which the combination of
activators, would serve as a better pharmacological treat- any given patient’s unique clinical, genomic, and in vitro
ment for this specific patient. cellular characteristics can fine-tune decisions regarding the

Predicted Paradigm Shift in Disease Diagnosis

Family history
Traditional model

? Slow

? Ineffective

Health monitoring

Familial disease Investigation of possible diagnosis Disease outcome

Fast
Patient-specific
New model

hiPSC-CMs

Predict disease
development &
Phenotypic screen progression

FIGURE 5. hiPSC-based personalized diagnostics for disease prediction. Familial disease often has a large
social impact on family life. In the absence of a known disease-associated mutation, clinicians adopt a disease
prediction strategy for young offspring that involves long-term monitoring of biomarkers and heart function.
However, this strategy is time-consuming and delays implementation of prevention or early treatment plans.
Generation of hiPSC-CMs from family members may enable extensive phenotypic screening and early prediction
of disease severity and outcome, to improve patient care.

1116 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

diagnosis, treatment, and prevention of human disease are types, thus far none has become standard practice. Improv-
starting to take shape thanks to in vitro hiPSC-CM models. ing our ability to generate specific CM subtypes from hiP-
To facilitate this field and to promote more efficient devel- SCs will also enable more precise modeling of diseases that
opment of novel therapies, biobanking of hiPSCs has preferentially affect either the ventricles (e.g., HCM, DCM,
bloomed over the past 5 years, with participation of several ARVD), or atria (e.g., atrial arrhythmias), in addition to
academic groups (e.g., Stanford Cardiovascular Institute) opening new prospects for creating patient-specific bioen-
and commercial institutions (e.g., Coriell Institute, Cellular gineered pacemakers.
Dynamics International, Stem Cell Theranostics, Pfizer, and
Roslin Cells) which are typically funded in partnership with Significant limitations for the genome editing technology
government entities (e.g., NIH, CIRM, etc.). The Interna- include not only the length and cost of the process, but more
tional Stem Cell Banking Initiative (ISCBI) alongside the importantly, its efficiency, and off-target effects, as dis-
International Stem Cell Initiative (ISCI) have also coordi- cussed extensively in previous sections. In addition, the
nated teams of stem cell biologists, bio-banking centers, NGS field needs to address the issue of storage and manage-
stem cell society representatives (e.g., ISSCR, ISCT), eth- ment of the vast datasets generated, as well as to develop
ics committees (e.g., ISCF Ethics Working Party), and more effective methods of data analysis (288). Sequencing
regulators from over 20 countries to standardize the der- throughputs have rapidly outpaced even Moore’s Law for
ivation, culture, and characterization methods of hiPSCs computing power, which states that the number of transis-
(7). These large multinational efforts have one goal in tors in a dense integrated circuit doubles approximately
common: to expedite the clinical uses promised by the every two years (195). Improvement of data compression
hiPSC technology. algorithms would help to address the growing data-to-cen-
tral processing unit (CPU) ratio (287). To date, thousands
While hiPSCs avoid the main controversies associated with of WG and hundreds of thousands of exomes have been
hESCs (i.e., the destruction of human embryos during hESC sequenced. However, meaningful biomedical contribution
derivation), new ethical concerns have not only clouded the of these data has been greatly outpaced by the rate at which
use of hiPSCs, but also the generation and privatization of new samples are sequenced (156). Therefore, an additional
challenge for the NGS technology is to accelerate the rate of
patient-specific “omic” data. For example, when the com-
data analysis. Efficient data analysis would lead to the iden-
pany 23andMe initiated sales of a saliva-based Personal
tification of new disease-associated mutations, epigenetic
Genome Service (PGS) that provides genotyping reports on
modifications, or gene expression pathways which could in
254 diseases and conditions without prior marketing clear-
turn generate innovative therapeutic targets, improve the
ance or approval from the Food and Drug Administration,
predictive abilities of genetic testing, and ultimately trans-
this was considered a risk. The concerns were not only that
late into public health benefits. In the future, clinical and
this kit should have been regulated and marketed as a med-
hiPSC-based sequencing of patients suffering from disease
ical device, but also that patients were receiving complex
is expected to guide diagnosis and treatment decisions. The
information about their possible predisposition to diseases
precision medicine initiative is here to ensure that outstand-
without appropriate genetic counseling, and without con-
ing issues associated with the technologies involved are suf-
sideration for false-positive or false-negative outcomes ficiently addressed so that these tools can serve to benefit
(11). This and other examples illustrate the need to set patients. Precision health represents a revolution in patient
ethical boundaries and rules for generation and distribution care, by moving beyond incremental improvements in diag-
of patient-specific NGS data related to hiPSC generation nosis and treatments for acute or chronic disease to achieve
and analysis (43). Careful regulation of genome editing fundamental understanding of underlying disease causes
technologies will also be necessary to prevent eugenic mis- and treating individuals based on their unique genetic com-
use (140). position (45). By combining the strengths of basic scientific
research, biomedical data science, genome editing, and
In addition to ethical issues, technical challenges also need transformative biomedical platforms such as hiPSCs (299),
to be addressed for each technology. For the hiPSC technol- we can truly begin to understand individual variability in
ogy, one major concern is the immature state of differenti- disease development, progression, and response to therapy.
ated derivatives. This topic has been extensively discussed Although precision health requires big thinking and bold
elsewhere (128, 309), and several physical, chemical, and action, its promise of a revolution in healthcare away from
electrical signals are under investigation to enable these cells after-the-fact diagnosis, towards prediction and prevention,
to mature to a state more closely resembling the adult hu- is approaching fast.
man heart (124, 208, 266, 276, 309). Subtype specification
of CMs (i.e., into pacemaker, atrial, or ventricular cells of ACKNOWLEDGMENTS
either the right or left side of the heart) is also under inves-
tigation. Although a few protocols claim to enable enhance- We thank Dr. Joseph Gold and Blake Wu for critical read-
ment of the specific generation of ventricular (129, 296), ing of the manuscript. Due to space limitations, we are
atrial (53, 119, 263), and pacemaker (127, 326) CM sub- unable to include all of the important papers relevant to the

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1117


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

use of hiPSCs in cardiovascular research; we apologize to 13. Bellin M, Casini S, Davis RP, D’Aniello C, Haas J, Ward-van Oostwaard D, Tertoolen
LG, Jung CB, Elliott DA, Welling A, Laugwitz KL, Moretti A, Mummery CL. Isogenic
the investigators who have made significant contributions human pluripotent stem cell pairs reveal the role of a kcnh2 mutation in long-qt
to this field. syndrome. EMBO J 32: 3161–3175, 2013.

14. Bilic J, Izpisua Belmonte JC. Concise review: induced pluripotent stem cells versus
Address for reprint requests and other correspondence: J. C. embryonic stem cells: Close enough or yet too far apart? Stem Cells 30: 33– 41, 2012.
Wu, 265 Campus Dr., Rm. G1120B, Stanford, CA 94305-
5454 (e-mail: joewu@stanford.edu) or E. Matsa (e-mail: 15. Bondue A, Lapouge G, Paulissen C, Semeraro C, Iacovino M, Kyba M, Blanpain C.
Mesp1 acts as a master regulator of multipotent cardiovascular progenitor specifica-
ematsa@stanford.edu). tion. Cell Stem Cell 3: 69 – 84, 2008.

GRANTS 16. Braam SR, Passier R, Mummery CL. Cardiomyocytes from human pluripotent stem
cells in regenerative medicine and drug discovery. Trends Pharmacol Sci 30: 536 –545,
2009.
This work was supported by the American Heart Associa-
tion; Burroughs Wellcome Foundation; National Institutes 17. Braam SR, Tertoolen L, Casini S, Matsa E, Lu HR, Teisman A, Passier R, Denning C,
of Health (NIH) Grants R01 HL123968, R01 HL126527, Gallacher DJ, Towart R, Mummery CL. Repolarization reserve determines drug re-
sponses in human pluripotent stem cell derived cardiomyocytes. Stem Cell Res 10:
R01 HL128170, and R01 HL130020 (to J. C. Wu); NIH 48 –56, 2013.
Progenitor Cell Biology Jump Start Award, Stanford Car-
18. Bruneau BG, Nemer G, Schmitt JP, Charron F, Robitaille L, Caron S, Conner DA,
diovascular Institute Seed Grant, and American Heart As-
Gessler M, Nemer M, Seidman CE, Seidman JG. A murine model of holt-oram syn-
sociation Beginning Grant in Aid (to E. Matsa). drome defines roles of the t-box transcription factor tbx5 in cardiogenesis and dis-
ease. Cell 106: 709 –721, 2001.

DISCLOSURES 19. Brunel-Guitton C, Levtova A, Sasarman F. Mitochondrial diseases and cardiomyopa-


thies. Can J Cardiol 31: 1360 –1376, 2015.

J. Wu is a co-founder and Scientific Advisory Board member 20. Burridge PW, Diecke S, Matsa E, Sharma A, Wu H, Wu JC. Modeling cardiovascular
for Stem Cell Theranostics. diseases with patient-specific human pluripotent stem cell-derived cardiomyocytes.
Methods Mol Biol. 1353: 119 –130, 2016.

21. Burridge PW, Fuga Y, Matsa E, Wu H, Ong G, Sharma A, Holmström A, Chang AC,
REFERENCES Coronado MJ, Ebert AD, Knowles JW, Telli ML, Witteles RM, Blau HM, Bernstein D,
Altman RB, Wu JC. Human induced pluripotent stem cells recapitulate breast cancer
1. Abi Khalil C. The emerging role of epigenetics in cardiovascular disease. Ther Adv patients’ predilection to doxorubicin-induced cardiotoxicity. Nature Med. 22: 547–
Chronic Dis 5: 178 –187, 2014. 556, 2016.
2. Aggarwal P, Turner A, Matter A, Kattman SJ, Stoddard A, Lorier R, Swanson BJ, Arnett 22. Burridge PW, Holmstrom A, Wu JC. Chemically defined culture and cardiomyocyte
DK, Broeckel U. RNA expression profiling of human ipsc-derived cardiomyocytes in differentiation of human pluripotent stem cells. Curr Protocol Hum Genet 87: 21 3 1 3
a cardiac hypertrophy model. PLoS One 9: e108051, 2014.
15547–21, 2015.
3. Alex Buerkle C, Gompert Z. Population genomics based on low coverage sequencing:
23. Burridge PW, Keller G, Gold JD, Wu JC. Production of de novo cardiomyocytes:
How low should we go? Mol Ecol 22: 3028 –3035, 2013.
Human pluripotent stem cell differentiation and direct reprogramming. Cell Stem Cell
4. Amit M, Winkler ME, Menke S, Bruning E, Buscher K, Denner J, Haverich A, Itskovitz- 10: 16 –28, 2012.
Eldor J, Martin U. No evidence for infection of human embryonic stem cells by feeder
24. Burridge PW, Matsa E, Shukla P, Lin ZC, Churko JM, Ebert AD, Lan F, Diecke S,
cell-derived murine leukemia viruses. Stem Cells 23: 761–771, 2005.
Huber B, Mordwinkin NM, Plews JR, Abilez OJ, Cui B, Gold JD, Wu JC. Chemically
5. Andreoletti L, Leveque N, Boulagnon C, Brasselet C, Fornes P. Viral causes of human defined generation of human cardiomyocytes. Nat Methods 11: 855– 860, 2014.
myocarditis. Arch Cardiovasc Dis 102: 559 –568, 2009.
25. Burridge PW, Sharma A, Wu JC. Genetic and epigenetic regulation of human cardiac
6. Ang SL, Constam DB. A gene network establishing polarity in the early mouse em- reprogramming and differentiation in regenerative medicine. Annu Rev Genet 49:
bryo. Semin Cell Dev Biol 15: 555–561, 2004. 20.1.24855–20, 2015.

7. Aoi T, Stacey G. Impact of national and international stem cell banking initiatives on 26. Burridge PW, Thompson S, Millrod MA, Weinberg S, Yuan X, Peters A, Mahairaki V,
progress in the field of cell therapy: Iabs-jst joint workshop: summary for session 5. Koliatsos VE, Tung L, Zambidis ET. A universal system for highly efficient cardiac
Biologicals 43: 399 – 401, 2015. differentiation of human induced pluripotent stem cells that eliminates interline vari-
ability. PLoS One 6: e18293, 2011.
8. Applegarth DA, Toone JR, Lowry RB. Incidence of inborn errors of metabolism in
British Columbia, 1969 –1996. Pediatrics 105: e10, 2000. 27. Burrows CK, Banovich NE, Pavlovic BJ, Patterson K, Gallego Romero I, Pritchard JK,
Gilad Y. Genetic variation, not cell type of origin, underlies the majority of identifiable
9. Baghbaderani BA, Tian X, Neo BH, Burkall A, Dimezzo T, Sierra G, Zeng X, Warren
regulatory differences in ipscs. PLoS Genet 12: e1005793, 2016.
K, Kovarcik DP, Fellner T, Rao MS. cGMP-manufactured human induced pluripotent
stem cells are available for pre-clinical and clinical applications. Stem Cell Reports 5: 28. Callis TE, Pandya K, Seok HY, Tang RH, Tatsuguchi M, Huang ZP, Chen JF, Deng Z,
647– 659, 2015.
Gunn B, Shumate J, Willis MS, Selzman CH, Wang DZ. MicroRNA-208a is a regulator
10. Ban H, Nishishita N, Fusaki N, Tabata T, Saeki K, Shikamura M, Takada N, Inoue M, of cardiac hypertrophy and conduction in mice. J Clin Invest 119: 2772–2786, 2009.
Hasegawa M, Kawamata S, Nishikawa S. Efficient generation of transgene-free human
29. Cao X, Deng W, Qu R, Yu Q, Li J, Yang Y, Cao Y, Gao X, Xu X, Yu J. Non-viral
induced pluripotent stem cells (ipscs) by temperature-sensitive sendai virus vectors.
co-delivery of the four yamanaka factors for generation of human induced pluripotent
Proc Natl Acad Sci USA 108: 14234 –14239, 2011.
stem cells via calcium phosphate nanocomposite particles. Adv Functional Materials 23:
11. Barlas S. Precision medicine initiative aims for a new generation of diagnostics and 5403–5411, 2013.
treatments: but is the promise of genetic targeting overinflated? PT 40: 340 –352,
2015. 30. Carlson C, Koonce C, Aoyama N, Einhorn S, Fiene S, Thompson A, Swanson B, Anson
B, Kattman S. Phenotypic screening with human ips cell-derived cardiomyocytes:
12. Bayart E, Cohen-Haguenauer O. Technological overview of ips induction from human Hts-compatible assays for interrogating cardiac hypertrophy. J Biomol Screen 18:
adult somatic cells. Curr Gene Ther 13: 73–92, 2013. 1203–1211, 2013.

1118 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

31. Carvajal-Vergara X, Sevilla A, D’Souza SL, Ang YS, Schaniel C, Lee DF, Yang L, Kaplan foetal fibroblasts using the sleeping beauty transposon gene delivery system. Differ-
AD, Adler ED, Rozov R, Ge Y, Cohen N, Edelmann LJ, Chang B, Waghray A, Su J, entiation 86: 30 –37, 2013.
Pardo S, Lichtenbelt KD, Tartaglia M, Gelb BD, Lemischka IR. Patient-specific induced
pluripotent stem-cell-derived models of leopard syndrome. Nature 465: 808 – 812, 51. Denning C, Allegrucci C, Priddle H, Barbadillo-Munoz MD, Anderson D, Self T, Smith
2010. NM, Parkin CT, Young LE. Common culture conditions for maintenance and cardio-
myocyte differentiation of the human embryonic stem cell lines, bg01 and hues-7. Int
32. Caspi O, Huber I, Gepstein A, Arbel G, Maizels L, Boulos M, Gepstein L. Modeling of J Dev Biol 50: 27–37, 2006.
arrhythmogenic right ventricular cardiomyopathy with human induced pluripotent
stem cells. Circ Cardiovasc Genet 6: 557–568, 2013. 52. Denning C, Borgdorff V, Crutchley J, Firth KSA, George V, Kalra S, Kondrashov A,
Hoang MD, Mosqueira D, Patel A, Prodanov L, Rajamohan D, Skarnes WC, Smith
33. Chamberlain AA, Lin M, Lister RL, Maslov AA, Wang Y, Suzuki M, Wu B, Greally JM, JGW, Young LE. Cardiomyocytes from human pluripotent stem cells: from labo-
Zheng D, Zhou B. DNA methylation is developmentally regulated for genes essential ratory curiosity to industrial biomedical platform. Biochim Biophys Acta 4889:
for cardiogenesis. J Am Heart Assoc 3: e000976, 2014. 367–375, 2015.

34. Chandrasekera PC, Pippin JJ. The human subject: an integrative animal model for 53. Devalla HD, Schwach V, Ford JW, Milnes JT, El-Haou S, Jackson C, Gkatzis K, Elliott
21(st) century heart failure research. Am J Transl Res 7: 1636 –1647, 2015. DA, Chuva de Sousa Lopes SM, Mummery CL, Verkerk AO, Passier R. Atrial-like
cardiomyocytes from human pluripotent stem cells are a robust preclinical model for
35. Chasman DI, Posada D, Subrahmanyan L, Cook NR, Stanton VP Jr, Ridker PM. assessing atrial-selective pharmacology. EMBO Mol Med 7: 394 – 410, 2015.
Pharmacogenetic study of statin therapy and cholesterol reduction. JAMA 291: 2821–
2827, 2004. 54. Di Pasquale E, Lodola F, Miragoli M, Denegri M, Avelino-Cruz JE, Buonocore M,
Nakahama H, Portararo P, Bloise R, Napolitano C, Condorelli G, Priori SG. CaMKII
36. Chatterjee K, Zhang J, Honbo N, Karliner JS. Doxorubicin cardiomyopathy. Cardiology inhibition rectifies arrhythmic phenotype in a patient-specific model of catecholamin-
115: 155–162, 2010. ergic polymorphic ventricular tachycardia. Cell Death Dis 4: e843, 2014.

36a.Chen IY, Matsa E, Wu JC. Induced pluripotent stem cells: at the heart of cardiovascular 55. Dick E, Matsa E, Bispham J, Reza M, Guglieri M, Staniforth A, Watson S, Kumari R,
precision medicine. Nat Rev Cardiol 13: 333–349, 2016. Lochmuller H, Young L, Darling D, Denning C. Two new protocols to enhance the
production and isolation of human induced pluripotent stem cell lines. Stem Cell Res 6:
37. Chen VC, Ye J, Shukla P, Hua G, Chen D, Lin Z, Liu JC, Chai J, Gold J, Wu J, Hsu D, 158 –167, 2011.
Couture LA. Development of a scalable suspension culture for cardiac differentiation
from human pluripotent stem cells. Stem Cell Res 15: 365–375, 2015. 56. Diecke S, Lisowski L, Kooreman NG, Wu JC. Second generation codon optimized
minicircle (comic) for nonviral reprogramming of human adult fibroblasts. Methods
38. Chen ZY, He CY, Ehrhardt A, Kay MA. Minicircle DNA vectors devoid of bacterial Mol Biol 1181: 1–13, 2014.
DNA result in persistent and high-level transgene expression in vivo. Mol Ther 8:
495–500, 2003. 57. Diecke S, Lu J, Lee J, Termglinchan V, Kooreman NG, Burridge PW, Ebert AD,
Churko JM, Sharma A, Kay MA, Wu JC. Novel codon-optimized mini-intronic plasmid
39. Chester AH, Yacoub MH. The role of endothelin-1 in pulmonary arterial hyperten- for efficient, inexpensive, and xeno-free induction of pluripotency. Sci Rep 5: 8081,
sion. Glob Cardiol Sci Pract 2014: 62–78, 2014. 2015.

40. Choi J, Lee S, Mallard W, Clement K, Tagliazucchi GM, Lim H, Choi IY, Ferrari F, 58. Dimos JT, Rodolfa KT, Niakan KK, Weisenthal LM, Mitsumoto H, Chung W, Croft GF,
Tsankov AM, Pop R, Lee G, Rinn JL, Meissner A, Park PJ, Hochedlinger K. A compar- Saphier G, Leibel R, Goland R, Wichterle H, Henderson CE, Eggan K. Induced pluri-
ison of genetically matched cell lines reveals the equivalence of human ipscs and escs. potent stem cells generated from patients with als can be differentiated into motor
Nat Biotechnol 33: 1173–1181, 2015. neurons. Science 321: 1218 –1221, 2008.

41. Churko JM, Burridge PW, Wu JC. Generation of human ipscs from human peripheral 59. Ding Q, Lee YK, Schaefer EA, Peters DT, Veres A, Kim K, Kuperwasser N, Motola
blood mononuclear cells using non-integrative sendai virus in chemically defined con- DL, Meissner TB, Hendriks WT, Trevisan M, Gupta RM, Moisan A, Banks E, Friesen
ditions. Methods Mol Biol 1036: 81– 88, 2013. M, Schinzel RT, Xia F, Tang A, Xia Y, Figueroa E, Wann A, Ahfeldt T, Daheron L,
Zhang F, Rubin LL, Peng LF, Chung RT, Musunuru K, Cowan CA. A talen genome-
42. Churko JM, Mantalas GL, Snyder MP, Wu JC. Overview of high throughput sequenc- editing system for generating human stem cell-based disease models. Cell Stem Cell
ing technologies to elucidate molecular pathways in cardiovascular diseases. Circ Res 12: 238 –251, 2013.
112: 1613–1623, 2013.
60. Dowey SN, Huang X, Chou BK, Ye Z, Cheng L. Generation of integration-free human
43. Clarke AJ. Managing the ethical challenges of next-generation sequencing in genomic induced pluripotent stem cells from postnatal blood mononuclear cells by plasmid
medicine. Br Med Bull 111: 17–30, 2014. vector expression. Nature Protocols 7: 2013–2021, 2012.

44. Colamonici OR, Domanski P, Sweitzer SM, Larner A, Buller RM. Vaccinia virus b18r 61. Drawnel FM, Boccardo S, Prummer M, Delobel F, Graff A, Weber M, Gerard R, Badi
gene encodes a type i interferon-binding protein that blocks interferon alpha trans- L, Kam-Thong T, Bu L, Jiang X, Hoflack JC, Kiialainen A, Jeworutzki E, Aoyama N,
membrane signaling. J Biol Chem 270: 15974 –15978, 1995. Carlson C, Burcin M, Gromo G, Boehringer M, Stahlberg H, Hall BJ, Magnone MC,
Kolaja K, Chien KR, Bailly J, Iacone R. Disease modeling and phenotypic drug screening
45. Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med 372: for diabetic cardiomyopathy using human induced pluripotent stem cells. Cell Rep 9:
793–795, 2015. 810 – 821, 2014.

46. Corrigan-Curay J, O’Reilly M, Kohn DB, Cannon PM, Bao G, Bushman FD, Carroll D, 62. Dubois NC, Craft AM, Sharma P, Elliott DA, Stanley EG, Elefanty AG, Gramolini A,
Cathomen T, Joung JK, Roth D, Sadelain M, Scharenberg AM, von Kalle C, Zhang F, Keller G. Sirpa is a specific cell-surface marker for isolating cardiomyocytes derived
Jambou R, Rosenthal E, Hassani M, Singh A, Porteus MH. Genome editing technolo- from human pluripotent stem cells. Nat Biotechnol 29: 1011–1018, 2011.
gies: Defining a path to clinic. Mol Ther 23: 796 – 806, 2015.
63. Ebert AD, Kodo K, Liang P, Wu H, Huber BC, Riegler J, Churko J, Lee J, de Almeida
47. Corwin EJ. The concept of epigenetics and its role in the development of cardiovas- P, Lan F, Diecke S, Burridge PW, Gold JD, Mochly-Rosen D, Wu JC. Characterization
cular disease: Commentary on “new and emerging theories of cardiovascular dis- of the molecular mechanisms underlying increased ischemic damage in the aldehyde
ease.” Biol Res Nurs 6: 11–16, 2004. dehydrogenase 2 genetic polymorphism using a human induced pluripotent stem cell
model system. Sci Transl Med 6: 255ra130, 2014.
48. David L, Polo JM. Phases of reprogramming. Stem Cell Res 12: 754 –761, 2014.
64. Ebrahimi B. Reprogramming barriers and enhancers: Strategies to enhance the effi-
49. Davidson MH, Stein EA, Dujovne CA, Hunninghake DB, Weiss SR, Knopp RH, Illing- ciency and kinetics of induced pluripotency. Cell Regen 4: 10, 2015.
worth DR, Mitchel YB, Melino MR, Zupkis RV, Dobrinska MR, Amin RD, Tobert JA.
The efficacy and six-week tolerability of simvastatin 80 and 160 mg/day. Am J Cardiol 65. Edwards AM, Arrowsmith CH, Bountra C, Bunnage ME, Feldmann M, Knight JC,
79: 38 – 42, 1997. Patel DD, Prinos P, Taylor MD, Sundstrom M, Barker P, Barsyte D, Bengtson MH,
Bell C, Bowness P, Boycott KM, Buser-Doepner C, Carpenter CL, Carr AJ, Clark K,
50. Davis RP, Nemes C, Varga E, Freund C, Kosmidis G, Gkatzis K, de Jong D, Szuhai K, Das AM, Dhanak D, Dirks P, Ellis J, Fantin VR, Flores C, Fon EA, Frail DE, Gileadi O,
Dinnyes A, Mummery CL. Generation of induced pluripotent stem cells from human O’Hagan RC, Howe T, Isaac JT, Jabado N, Jakobsson PJ, Klareskog L, Knapp S, Lee

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1119


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

WH, Lima-Fernandes E, Lundberg IE, Marshall J, Massirer KB, MacKenzie AE, Maruy- 83. Guilinger JP, Pattanayak V, Reyon D, Tsai SQ, Sander JD, Joung JK, Liu DR. Broad
ama T, Mueller-Fahrnow A, Muthuswamy S, Nanchahal J, O’Brien C, Oppermann U, specificity profiling of talens results in engineered nucleases with improved DNA-
Ostermann N, Petrecca K, Pollock BG, Poupon V, Prinjha RK, Rosenberg SH, Rouleau cleavage specificity. Nat Methods 11: 429 – 435, 2014.
G, Skingle M, Slutsky AS, Smith GA, Verhelle D, Widmer H, Young LT. Preclinical
target validation using patient-derived cells. Nat Rev Drug Discov 14: 149 –150, 2015. 84. Gurdon JB. Adult frogs derived from the nuclei of single somatic cells. Dev Biol 4:
256 –273, 1962.
66. Elanzew A, Sommer A, Pusch-Klein A, Brustle O, Haupt S. A reproducible and ver-
satile system for the dynamic expansion of human pluripotent stem cells in suspen- 85. Guzun R, Kaambre T, Bagur R, Grichine A, Usson Y, Varikmaa M, Anmann T, Tepp K,
sion. Biotechnol J 10: 1589 –1599, 2015. Timohhina N, Shevchuk I, Chekulayev V, Boucher F, Dos Santos P, Schlattner U,
Wallimann T, Kuznetsov AV, Dzeja P, Aliev M, Saks V. Modular organization of cardiac
67. Fatima A, Kaifeng S, Dittmann S, Xu G, Gupta MK, Linke M, Zechner U, Nguemo F, energy metabolism: Energy conversion, transfer and feedback regulation. Acta Physiol
Milting H, Farr M, Hescheler J, Saric T. The disease-specific phenotype in cardiomy- 213: 84 –106, 2015.
ocytes derived from induced pluripotent stem cells of two long qt syndrome type 3
patients. PLoS One 8: e83005, 2013. 86. Gwathmey JK, Tsaioun K, Hajjar RJ. Cardionomics: a new integrative approach for
screening cardiotoxicity of drug candidates. Expert Opin Drug Metab Toxicol 5: 647–
68. Fatima A, Xu G, Shao K, Papadopoulos S, Lehmann M, Arnaiz-Cot JJ, Rosa AO, 660, 2009.
Nguemo F, Matzkies M, Dittmann S, Stone SL, Linke M, Zechner U, Beyer V, Hennies
87. Haghighi K, Kolokathis F, Pater L, Lynch RA, Asahi M, Gramolini AO, Fan GC, Tsiapras
HC, Rosenkranz S, Klauke B, Parwani AS, Haverkamp W, Pfitzer G, Farr M, Cleemann
D, Hahn HS, Adamopoulos S, Liggett SB, Dorn GW 2nd, MacLennan DH, Kremasti-
L, Morad M, Milting H, Hescheler J, Saric T. In vitro modeling of ryanodine receptor
nos DT, Kranias EG. Human phospholamban null results in lethal dilated cardiomy-
2 dysfunction using human induced pluripotent stem cells. Cell Physiol Biochem 28:
opathy revealing a critical difference between mouse and human. J Clin Invest 111:
579 –592, 2011.
869 – 876, 2003.
69. Feldman MD, Copelas L, Gwathmey JK, Phillips P, Warren SE, Schoen FJ, Grossman
88. Han D, Pal S, Yang Y, Jiang S, Nangreave J, Liu Y, Yan H. DNA gridiron nanostructures
W, Morgan JP. Deficient production of cyclic amp: Pharmacologic evidence of an
based on four-arm junctions. Science 339: 1412–1415, 2013.
important cause of contractile dysfunction in patients with end-stage heart failure.
Circulation 75: 331–339, 1987. 89. Han L, Li Y, Tchao J, Kaplan AD, Lin B, Li Y, Mich-Basso J, Lis A, Hassan N, London B,
Bett GC, Tobita K, Rasmusson RL, Yang L. Study familial hypertrophic cardiomyopa-
70. Foley AC, Mercola M. Heart induction by wnt antagonists depends on the homeodo-
thy using patient-specific induced pluripotent stem cells. Cardiovasc Res 104: 258 –269,
main transcription factor hex. Genes Dev 19: 387–396, 2005.
2014.
71. Frishman WH. 〉-adrenergic blockade in cardiovascular disease. J Cardiovasc Pharma-
90. Hankowski KE, Hamazaki T, Umezawa A, Terada N. Induced pluripotent stem cells
col Ther 18: 310 –319, 2013.
as a next-generation biomedical interface. Lab Invest 91: 972–977, 2011.
72. Fusaki N, Ban H, Nishiyama A, Saeki K, Hasegawa M. Efficient induction of transgene-
91. Hannah-Shmouni F, Seidelmann SB, Sirrs S, Mani A, Jacoby D. The genetic challenges
free human pluripotent stem cells using a vector based on sendai virus, an rna virus
and opportunities in advanced heart failure. Can J Cardiol 31: 1338 –1350, 2015.
that does not integrate into the host genome. Proc Jpn Acad Ser B Phys Biol Sci 85:
348 –362, 2009. 92. Hattori F, Chen H, Yamashita H, Tohyama S, Satoh YS, Yuasa S, Li W, Yamakawa H,
Tanaka T, Onitsuka T, Shimoji K, Ohno Y, Egashira T, Kaneda R, Murata M, Hidaka K,
73. Gaber N, Gagliardi M, Patel P, Kinnear C, Zhang C, Chitayat D, Shannon P, Jaeggi E, Morisaki T, Sasaki E, Suzuki T, Sano M, Makino S, Oikawa S, Fukuda K. Nongenetic
Tabori U, Keller G, Mital S. Fetal reprogramming and senescence in hypoplastic left method for purifying stem cell-derived cardiomyocytes. Nat Methods 7: 61– 66, 2010.
heart syndrome and in human pluripotent stem cells during cardiac differentiation. Am
J Pathol 183: 720 –734, 2013. 93. Hinson JT, Chopra A, Nafissi N, Polacheck WJ, Benson CC, Swist S, Gorham J, Yang
L, Schafer S, Sheng CC, Haghighi A, Homsy J, Hubner N, Church G, Cook SA, Linke
74. Gaj T, Gersbach CA, Barbas CF 3rd. Zfn, talen, and crispr/cas-based methods for WA, Chen CS, Seidman JG, Seidman CE. Heart disease. Titin mutations in ips cells
genome engineering. Trends Biotechnol 31: 397– 405, 2013. define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science 349:
982–986, 2015.
75. Gaj T, Mercer AC, Gersbach CA, Gordley RM, Barbas CF 3rd. Structure-guided
reprogramming of serine recombinase DNA sequence specificity. Proc Natl Acad Sci 94. Hiratsuka M, Uno N, Ueda K, Kurosaki H, Imaoka N, Kazuki K, Ueno E, Akakura Y,
USA 108: 498 –503, 2011. Katoh M, Osaki M, Kazuki Y, Nakagawa M, Yamanaka S, Oshimura M. Integration-
free ips cells engineered using human artificial chromosome vectors. PLoS One 6:
76. Gaj T, Mercer AC, Sirk SJ, Smith HL, Barbas CF 3rd. A comprehensive approach to
e25961, 2011.
zinc-finger recombinase customization enables genomic targeting in human cells.
Nucleic Acids Res 41: 3937–3946, 2013. 95. Hiroi Y, Kudoh S, Monzen K, Ikeda Y, Yazaki Y, Nagai R, Komuro I. Tbx5 associates
with nkx2-5 and synergistically promotes cardiomyocyte differentiation. Nat Genet
77. Gersbach CA, Perez-Pinera P. Activating human genes with zinc finger proteins,
28: 276 –280, 2001.
transcription activator-like effectors and crispr/cas9 for gene therapy and regenera-
tive medicine. Expert Opin Ther Targets 18: 835– 839, 2014. 96. Holmgren G, Synnergren J, Bogestal Y, Ameen C, Akesson K, Holmgren S, Lindahl A,
Sartipy P. Identification of novel biomarkers for doxorubicin-induced toxicity in hu-
78. Gersh BJ, Maron BJ, Bonow RO, Dearani JA, Fifer MA, Link MS, Naidu SS, Nishimura
man cardiomyocytes derived from pluripotent stem cells. Toxicology 328: 102–111,
RA, Ommen SR, Rakowski H, Seidman CE, Towbin JA, Udelson JE, Yancy CW. 2011
2015.
ACCF/AHA guideline for the diagnosis and treatment of hypertrophic cardiomyopa-
thy: a report of the american college of cardiology foundation/american heart associ- 97. Hotta A, Yamanaka S. From genomics to gene therapy: Induced pluripotent stem cells
ation task force on practice guidelines. Circulation 124: e783– 831, 2011. meet genome editing. Annu Rev Genet. 49: 47–70, 2015.

79. Gilchrist KH, Lewis GF, Gay EA, Sellgren KL, Grego S. High-throughput cardiac safety 98. Hou P, Li Y, Zhang X, Liu C, Guan J, Li H, Zhao T, Ye J, Yang W, Liu K, Ge J, Xu J,
evaluation and multi-parameter arrhythmia profiling of cardiomyocytes using micro- Zhang Q, Zhao Y, Deng H. Pluripotent stem cells induced from mouse somatic cells
electrode arrays. Toxicol Appl Pharmacol 288: 249 –257, 2015. by small-molecule compounds. Science 341: 651– 654, 2013.

80. Gonzalez F, Boue S, Izpisua Belmonte JC. Methods for making induced pluripotent 99. Houser SR, Margulies KB, Murphy AM, Spinale FG, Francis GS, Prabhu SD, Rockman
stem cells: reprogramming a la carte. Nat Rev Genet 12: 231–242, 2011. HA, Kass DA, Molkentin JD, Sussman MA, Koch WJ, American Heart Association
Council on Basic Cardiovascular Sciences. Animal models of heart failure: a scientific
81. Gonzalez R, Lee JW, Schultz PG. Stepwise chemically induced cardiomyocyte speci- statement from the american heart association. Circ Res 111: 131–150, 2012.
fication of human embryonic stem cells. Angew Chem Int Ed Engl 50: 11181–11185,
2011. 100. Hsu PD, Lander ES, Zhang F. Development and applications of crispr-cas9 for ge-
nome engineering. Cell 157: 1262–1278, 2014.
82. Grobarczyk B, Franco B, Hanon K, Malgrange B. Generation of isogenic human ips cell
line precisely corrected by genome editing using the crispr/cas9 system. Stem Cell Rev 101. Huang HP, Chen PH, Hwu WL, Chuang CY, Chien YH, Stone L, Chien CL, Li LT,
11: 774 –787, 2015. Chiang SC, Chen HF, Ho HN, Chen CH, Kuo HC. Human pompe disease-induced

1120 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

pluripotent stem cells for pathogenesis modeling, drug testing and disease marker stem cell-derived atrial-like cardiomyocytes based on sarcolipin expression. PLoS One
identification. Hum Mol Genet 20: 4851– 4864, 2011. 9: e101316, 2014.

102. Huang ZP, Chen J, Seok HY, Zhang Z, Kataoka M, Hu X, Wang DZ. Microrna-22 120. Jouni M, Si-Tayeb K, Es-Salah-Lamoureux Z, Latypova X, Champon B, Caillaud A,
regulates cardiac hypertrophy and remodeling in response to stress. Circ Res 112: Rungoat A, Charpentier F, Loussouarn G, Baro I, Zibara K, Lemarchand P, Gaborit N.
1234 –1243, 2013. Toward personalized medicine: Using cardiomyocytes differentiated from urine-de-
rived pluripotent stem cells to recapitulate electrophysiological characteristics of type
103. Huangfu D, Osafune K, Maehr R, Guo W, Eijkelenboom A, Chen S, Muhlestein W, 2 long qt syndrome. JAMA 4: 9, 2015.
Melton DA. Induction of pluripotent stem cells from primary human fibroblasts with
only oct4 and sox2. Nat Biotech 26: 1269 –1275, 2008. 121. Jung CB, Moretti A, Schnitzler MM, Iop L, Storch U, Bellin M, Dorn T, Ruppenthal S,
Pfeiffer S, Goedel A, Dirschinger RJ, Seyfarth M, Lam JT, Sinnecker D, Gudermann T,
104. Hung SC, Kang MS, Kieff E. Maintenance of epstein-barr virus (ebv) orip-based epi- Lipp P, Laugwitz KL. Dantrolene rescues arrhythmogenic ryr2 defect in a patient-
somes requires ebv-encoded nuclear antigen-1 chromosome-binding domains, which specific stem cell model of catecholaminergic polymorphic ventricular tachycardia.
can be replaced by high-mobility group-i or histone h1. Proc Natl Acad Sci USA 98: EMBO Mol Med 4: 180 –191, 2012.
1865–1870, 2001.
122. Kadari A, Mekala S, Wagner N, Malan D, Koth J, Doll K, Stappert L, Eckert D, Peitz M,
105. Hussain W, Moens N, Veraitch FS, Hernandez D, Mason C, Lye GJ. Reproducible Matthes J, Sasse P, Herzig S, Brustle O, Ergun S, Edenhofer F. Robust generation of
culture and differentiation of mouse embryonic stem cells using an automated micro- cardiomyocytes from human ips cells requires precise modulation of bmp and wnt
well platform. Biochem Eng J 77: 246 –257, 2013. signaling. Stem Cell Rev 11: 560 –569, 2014.

106. Hussein SM, Batada NN, Vuoristo S, Ching RW, Autio R, Narva E, Ng S, Sourour M, 123. Kaji K, Norrby K, Paca A, Mileikovsky M, Mohseni P, Woltjen K. Virus-free induction
Hamalainen R, Olsson C, Lundin K, Mikkola M, Trokovic R, Peitz M, Brustle O, of pluripotency and subsequent excision of reprogramming factors. Nature 458: 771–
Bazett-Jones DP, Alitalo K, Lahesmaa R, Nagy A, Otonkoski T. Copy number variation 775, 2009.
and selection during reprogramming to pluripotency. Nature 471: 58 – 62, 2011.
124. Kamakura T, Makiyama T, Sasaki K, Yoshida Y, Wuriyanghai Y, Chen J, Hattori T,
107. Illingworth R, Kerr A, Desousa D, Jorgensen H, Ellis P, Stalker J, Jackson D, Clee C, Ohno S, Kita T, Horie M, Yamanaka S, Kimura T. Ultrastructural maturation of
Plumb R, Rogers J, Humphray S, Cox T, Langford C, Bird A. A novel cpg island set human-induced pluripotent stem cell-derived cardiomyocytes in a long-term culture.
identifies tissue-specific methylation at developmental gene loci. PLoS Biol 6: e22, Circ J 77: 1307–1314, 2013.
2008.
125. Kang HB, Kim JS, Kwon HJ, Nam KH, Youn HS, Sok DE, Lee Y. Basic fibroblast growth
108. Ingolia NT, Brar GA, Rouskin S, McGeachy AM, Weissman JS. The ribosome profiling factor activates erk and induces c-fos in human embryonic stem cell line mizhes1.
strategy for monitoring translation in vivo by deep sequencing of ribosome-protected Stem Cells Dev 14: 395– 401, 2005.
mrna fragments. Nat Protoc 7: 1534 –1550, 2012.
126. Kang X, Yu Q, Huang Y, Song B, Chen Y, Gao X, He W, Sun X, Fan Y. Effects of
integrating and non-integrating reprogramming methods on copy number variation
109. Itzhaki I, Maizels L, Huber I, Gepstein A, Arbel G, Caspi O, Miller L, Belhassen B, Nof
and genomic stability of human induced pluripotent stem cells. PLoS One 10:
E, Glikson M, Gepstein L. Modeling of catecholaminergic polymorphic ventricular
e0131128, 2015.
tachycardia with patient-specific human-induced pluripotent stem cells. J Am Coll
Cardiol 60: 990 –1000, 2012. 127. Kapoor N, Liang W, Marban E, Cho HC. Direct conversion of quiescent cardiomyo-
cytes to pacemaker cells by expression of tbx18. Nat Biotech 31: 54 – 62, 2013.
110. Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O, Winterstern A, Feldman O,
Gepstein A, Arbel G, Hammerman H, Boulos M, Gepstein L. Modelling the long qt 128. Karakikes I, Ameen M, Termglinchan V, Wu JC. Human induced pluripotent stem
syndrome with induced pluripotent stem cells. Nature 471: 225–229, 2011. cell-derived cardiomyocytes: insights into molecular, cellular, and functional pheno-
types. Circ Res 117: 80 – 88, 2015.
111. Jacoby D, McKenna WJ. Genetics of inherited cardiomyopathy. Eur Heart J 33: 296 –
304, 2012. 129. Karakikes I, Senyei GD, Hansen J, Kong CW, Azeloglu EU, Stillitano F, Lieu DK, Wang
J, Ren L, Hulot JS, Iyengar R, Li RA, Hajjar RJ. Small molecule-mediated directed
112. Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome inte- differentiation of human embryonic stem cells toward ventricular cardiomyocytes.
grates intrinsic and environmental signals. Nat Genet 33 Suppl: 245–254, 2003. Stem Cells Transl Med 3: 18 –31, 2014.
113. Jensen BC, McLeod HL. Pharmacogenomics as a risk mitigation strategy for chemo- 130. Karakikes I, Stillitano F, Nonnenmacher M, Tzimas C, Sanoudou D, Termglinchan V,
therapeutic cardiotoxicity. Pharmacogenomics 14: 205–213, 2013. Kong CW, Rushing S, Hansen J, Ceholski D, Kolokathis F, Kremastinos D, Katoulis A,
Ren L, Cohen N, Gho JM, Tsiapras D, Vink A, Wu JC, Asselbergs FW, Li RA, Hulot JS,
114. Jia F, Wilson KD, Sun N, Gupta DM, Huang M, Li Z, Panetta NJ, Chen ZY, Robbins RC,
Kranias EG, Hajjar RJ. Correction of human phospholamban r14del mutation associ-
Kay MA, Longaker MT, Wu JC. A nonviral minicircle vector for deriving human ips
ated with cardiomyopathy using targeted nucleases and combination therapy. Nat
cells. Nat Methods 7: 197–199, 2010.
Commun 6: 6955, 2015.
115. Jiang Y, Habibollah S, Tilgner K, Collin J, Barta T, Al-Aama JY, Tesarov L, Hussain R,
131. Kattman SJ, Witty AD, Gagliardi M, Dubois NC, Niapour M, Hotta A, Ellis J, Keller G.
Trafford AW, Kirkwood G, Sernagor E, Eleftheriou CG, Przyborski S, Stojkovic M,
Stage-specific optimization of activin/nodal and bmp signaling promotes cardiac dif-
Lako M, Keavney B,Armstrong L. An induced pluripotent stem cell model of hypoplas- ferentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell 8: 228 –
tic left heart syndrome (hlhs) reveals multiple expression and functional differences in 240, 2011.
hlhs-derived cardiac myocytes. Stem Cells Transl Med 3: 416 – 423, 2014.
132. Kempf H, Olmer R, Kropp C, Ruckert M, Jara-Avaca M, Robles-Diaz D, Franke A,
116. Jiang Y, Habibollah S, Tilgner K, Collin J, Barta T, Al-Aama JY, Tesarov L, Hussain R, Elliott DA, Wojciechowski D, Fischer M, Roa Lara A, Kensah G, Gruh I, Haverich A,
Trafford AW, Kirkwood G, Sernagor E, Eleftheriou CG, Przyborski S, Stojkovic M, Martin U, Zweigerdt R. Controlling expansion and cardiomyogenic differentiation of
Lako M, Keavney B, Armstrong L. An induced pluripotent stem cell model of hyp- human pluripotent stem cells in scalable suspension culture. Stem Cell Reports 3:
oplastic left heart syndrome (hlhs) reveals multiple expression and functional differ- 1132–1146, 2014.
ences in hlhs-derived cardiac myocytes. Stem Cells Transl Med, 2014.
10.5966/sctm.2013-0105. 133. Khoury MJ, Iademarco MF, Riley WT. Precision public health for the era of precision
medicine. Am J Prevent Med 50: 398 – 401, 2016.
117. Jinek M. A programmable dual-rna-guided DNA endonuclease in adaptive bacterial
immunity. Science 337: 816 – 821, 2012. 134. Kim C, Wong J, Wen J, Wang S, Wang C, Spiering S, Kan NG, Forcales S, Puri PL,
Leone TC, Marine JE, Calkins H, Kelly DP, Judge DP, Chen HS. Studying arrhythmo-
118. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable genic right ventricular dysplasia with patient-specific ipscs. Nature 494: 105–110,
dual-rna-guided DNA endonuclease in adaptive bacterial immunity. Science 337: 816 – 2013.
821, 2012.
135. Kim D, Kim CH, Moon JI, Chung YG, Chang MY, Han BS, Ko S, Yang E, Cha KY, Lanza
119. Josowitz R, Lu J, Falce C, D’Souza SL, Wu M, Cohen N, Dubois NC, Zhao Y, Sobie EA, R, Kim KS. Generation of human induced pluripotent stem cells by direct delivery of
Fishman GI, Gelb BD. Identification and purification of human induced pluripotent reprogramming proteins. Cell Stem Cell 4: 472– 476, 2009.

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1121


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

136. Kim JS, Lee HJ, Carroll D. Genome editing with modularly assembled zinc-finger 156. Li C, Liakata M, Rebholz-Schuhmann D. Biological network extraction from scientific
nucleases. Nat Methods 7: 91–92, 2010. literature: state of the art and challenges. Brief Bioinform 15: 856 – 877, 2014.

137. Kim WH, Jung DW, Williams DR. Making cardiomyocytes with your chemistry set: 157. Li X, Heyer WD. Homologous recombination in DNA repair and DNA damage
Small molecule-induced cardiogenesis in somatic cells. World J Cardiol 7: 125–133, tolerance. Cell Res 18: 99 –113, 2008.
2015.
158. Li Y, Zhang Q, Yin X, Yang W, Du Y, Hou P, Ge J, Liu C, Zhang W, Zhang X, Wu Y,
138. Konermann S, Brigham MD, Trevino AE, Joung J, Abudayyeh OO, Barcena C, Hsu PD, Li H, Liu K, Wu C, Song Z, Zhao Y, Shi Y, Deng H. Generation of ipscs from mouse
Habib N, Gootenberg JS, Nishimasu H, Nureki O, Zhang F. Genome-scale transcrip- fibroblasts with a single gene, oct4, and small molecules. Cell Res 21: 196 –204, 2011.
tional activation by an engineered crispr-cas9 complex. Nature 517: 583–588, 2015.
159. Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, Raval KK, Zhang J, Kamp
139. Kowalski MP, Yoder A, Liu L, Pajak L. Controlling embryonic stem cell growth and TJ, Palecek SP. Robust cardiomyocyte differentiation from human pluripotent stem
differentiation by automation: Enhanced and more reliable differentiation for drug cells via temporal modulation of canonical wnt signaling. Proc Natl Acad Sci USA 109:
discovery. J Biomol Screen 17: 1171–1179, 2012. E1848 –1857, 2012.

140. Krishan K, Kanchan T, Singh B. Human genome editing and ethical considerations. Sci 160. Liang G, Zhang Y. Embryonic stem cell and induced pluripotent stem cell: an epige-
Eng Ethics 22: 597–599, 2016. netic perspective. Cell Res 23: 49 – 69, 2013.

141. Krueger F, Kreck B, Franke A, Andrews SR. DNA methylome analysis using short 161. Liang P, Lan F, Lee AS, Gong T, Sanchez-Freire V, Wang Y, Diecke S, Sallam K,
bisulfite sequencing data. Nature Methods 9: 145–151, 2012. Knowles JW, Nguyen PK, Wang PJ, Bers DM, Robbins RC, Wu JC. Drug screening
using a library of human induced pluripotent stem cell-derived cardiomyocytes re-
142. Kujala K, Paavola J, Lahti A, Larsson K, Pekkanen-Mattila M, Viitasalo M, Lahtinen AM, veals disease specific patterns of cardiotoxicity. Circulation 127: 1677–1691, 2013.
Toivonen L, Kontula K, Swan H, Laine M, Silvennoinen O, Aalto-Setala K. Cell model
of catecholaminergic polymorphic ventricular tachycardia reveals early and delayed 162. Limongelli G, Tome-Esteban M, Dejthevaporn C, Rahman S, Hanna MG, Elliott PM.
afterdepolarizations. PLoS One 7: e44660, 2012. Prevalence and natural history of heart disease in adults with primary mitochondrial
respiratory chain disease. Eur J Heart Fail 12: 114 –121, 2010.
143. Kushwaha SS, Fallon JT, Fuster V. Restrictive cardiomyopathy. N Engl J Med 336:
267–276, 1997. 163. Lin B, Kim J, Li YX, Pan HY, Carvajal-Vergara X, Salama G, Cheng T, Li Y, Lo CW,
Yang L. High-purity enrichment of functional cardiovascular cells from human ips cells.
144. Labaj PP, Leparc GG, Linggi BE, Markillie LM, Wiley HS, Kreil DP. Characterization Cardiovasc Res 95: 327–335, 2012.
and improvement of rna-seq precision in quantitative transcript expression profiling.
Bioinformatics 27: i383–391, 2011. 164. Lin B, Li Y, Han L, Kaplan AD, Ao Y, Kalra S, Bett GC, Rasmusson RL, Denning C, Yang
L. Modeling and studying mechanism of dilated cardiomyopathy using induced pluri-
145. Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA, Dupras SK, Reinecke potent stem cells derived from duchenne muscular dystrophy (dmd) patients. Dis
H, Xu C, Hassanipour M, Police S, O’Sullivan C, Collins L, Chen Y, Minami E, Gill EA, Model Mech 8: 457– 466, 2015.
Ueno S, Yuan C, Gold J, Murry CE. Cardiomyocytes derived from human embryonic
stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat Biotech- 165. Liu L, Li Y, Li S, Hu N, He Y, Pong R, Lin D, Lu L, Law M. Comparison of next-
nol 25: 1015–1024, 2007. generation sequencing systems. J Biomed Biotechnol 2012: 251364, 2012.

146. Lam HY, Clark MJ, Chen R, Chen R, Natsoulis G, O’Huallachain M, Dewey FE, 166. Lu J, Zhang F, Kay MA. A mini-intronic plasmid (mip): a novel robust transgene
expression vector in vivo and in vitro. Mol Ther 21: 954 –963, 2013.
Habegger L, Ashley EA, Gerstein MB, Butte AJ, Ji HP, Snyder M. Performance com-
parison of whole-genome sequencing platforms. Nat Biotechnol 30: 78 – 82, 2012. 167. Lu J, Zhang F, Xu S, Fire AZ, Kay MA. The extragenic spacer length between the 5’ and
3’ ends of the transgene expression cassette affects transgene silencing from plasmid-
147. Lan F, Lee AS, Liang P, Sanchez-Freire V, Nguyen PK, Wang L, Han L, Yen M, Wang
based vectors. Mol Ther 20: 2111–2119, 2012.
Y, Sun N, Abilez OJ, Hu S, Ebert AD, Navarrete EG, Simmons CS, Wheeler M, Pruitt
B, Lewis R, Yamaguchi Y, Ashley EA, Bers DM, Robbins RC, Longaker MT, Wu JC. 168. Lujan E, Zunder ER, Ng YH, Goronzy IN, Nolan GP, Wernig M. Early reprogramming
Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy regulators identified by prospective isolation and mass cytometry. Nature 521: 352–
pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell 12: 101– 356, 2015.
113, 2013.
169. Luna-Zurita L, Stirnimann CU, Glatt S, Kaynak BL, Thomas S, Baudin F, Samee AH, He
149. Larson MH, Gilbert LA, Wang X, Lim WA, Weissman JS, Qi LS. Crispr interference D, Small EM, Mileikovsky M, Nagy A, Holloway AK, Pollard KS, CMüller CW, Bruneau
(crispri) for sequence-specific control of gene expression. Nat Protocols 8: 2180 – BG. Complex interdependence regulates heterotypic transcription factor distribution
2196, 2013. and coordinates cardiogenesis. Cell 164: 999 –1014, 2016.
150. Laslett LJ, Alagona P, Clark BA, Drozda JP, Saldivar F, Wilson SR, Poe C, Hart M. The 170. Ma D, Wei H, Lu J, Ho S, Zhang G, Sun X, Oh Y, Tan SH, Ng ML, Shim W, Wong P,
worldwide environment of cardiovascular disease: prevalence, diagnosis, therapy, Liew R. Generation of patient-specific induced pluripotent stem cell-derived cardio-
and policy issues a report from the American College of Cardiology. J Am Coll Cardiol myocytes as a cellular model of arrhythmogenic right ventricular cardiomyopathy. Eur
60: S1–S49, 2012. Heart J 34: 1122–1133, 2013.

151. Lee CH, Kim JH, Lee HJ, Jeon K, Lim H, Choi H, Lee ER, Park SH, Park JY, Hong S, Kim 171. Ma D, Wei H, Lu J, Huang D, Liu Z, Loh LJ, Islam O, Liew R, Shim W, Cook SA.
S, Cho SG. The generation of ips cells using non-viral magnetic nanoparticle based Characterization of a novel kcnq1 mutation for type 1 long qt syndrome and assess-
transfection. Biomaterials 32: 6683– 6691, 2011. ment of the therapeutic potential of a novel iks activator using patient-specific induced
pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther 6: 39, 2015.
152. Lee J, Sayed N, Hunter A, Au KF, Wong WH, Mocarski ES, Pera RR, Yakubov E,
Cooke JP. Activation of innate immunity is required for efficient nuclear reprogram- 172. Ma D, Wei H, Zhao Y, Lu J, Li G, Sahib NB, Tan TH, Wong KY, Shim W, Wong P, Cook
ming. Cell 151: 547–558, 2012. SA, Liew R. Modeling type 3 long qt syndrome with cardiomyocytes derived from
patient-specific induced pluripotent stem cells. Int J Cardiol 168: 5277–5286, 2013.
153. Lee YK, Lau YM, Ng KM, Lai WH, Ho SL, Tse HF, Siu CW, Ho PW. Efficient attenu-
ation of friedreich’s ataxia (frda) cardiomyopathy by modulation of iron homeostasis- 173. Ma Q, Lu AY. Pharmacogenetics, pharmacogenomics, and individualized medicine.
human induced pluripotent stem cell (hipsc) as a drug screening platform for frda. Int Pharmacol Rev 63: 437– 459, 2011.
J Cardiol 203: 964 –971, 2016.
174. Maddah M, Heidmann JD, Mandegar MA, Walker CD, Bolouki S, Conklin BR, Loewke
154. Lescroart F, Chabab S, Lin X, Rulands S, Paulissen C, Rodolosse A, Auer H, Achouri Y, KE. A non-invasive platform for functional characterization of stem-cell-derived car-
Dubois C, Bondue A, Simons BD, Blanpain C. Early lineage restriction in temporally diomyocytes with applications in cardiotoxicity testing. Stem Cell Reports 4: 621– 631,
distinct populations of mesp1 progenitors during mammalian heart development. Nat 2015.
Cell Biol 16: 829 – 840, 2014.
175. Malan D, Zhang M, Stallmeyer B, Muller J, Fleischmann BK, Schulze-Bahr E, Sasse P,
155. Li-Pook-Than J, Snyder M. Ipop goes the world: Integrated personalized omics pro- Greber B. Human ips cell model of type 3 long qt syndrome recapitulates drug-based
filing and the road toward improved health care. Chem Biol 20: 660 – 666, 2013. phenotype correction. Basic Res Cardiol 111: 14, 2016.

1122 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

176. Malik LH, Singh GD, Amsterdam EA. The epidemiology, clinical manifestations, and 197. Moretti A, Bellin M, Welling A, Jung CB, Lam JT, Bott-Flugel L, Dorn T, Goedel A,
management of chagas heart disease. Clin Cardiol 38: 565–569, 2014. Hohnke C, Hofmann F, Seyfarth M, Sinnecker D, Schomig A, Laugwitz KL. Patient-
specific induced pluripotent stem-cell models for long-qt syndrome. N Engl J Med 363:
177. Mandal PK, Rossi DJ. Reprogramming human fibroblasts to pluripotency using modi- 1397–1409, 2010.
fied mRNA. Nat Protoc 8: 568 –582, 2013.
198. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S,
178. Mardis ER. A decade’s perspective on DNA sequencing technology. Nature 470: Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Judd SE, Kissela BM, Lackland DT,
198 –203, 2011. Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Matchar DB, McGuire DK, Mohler ER,
3rd Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan
179. Maron BJ, Towbin JA, Thiene G, Antzelevitch C, Corrado D, Arnett D, Moss AJ,
L, Pandey DK, Reeves MJ, Rodriguez CJ, Sorlie PD, Stein J, Towfighi A, Turan TN,
Seidman CE, Young JB. Contemporary definitions and classification of the cardiomy- Virani SS, Willey JZ, Woo D, Yeh RW, Turner MB, American Heart Association. Heart
opathies: An american heart association scientific statement from the council on disease and stroke statistics–2015 update: a report from the American Heart Asso-
clinical cardiology, heart failure and transplantation committee; quality of care and ciation. Circulation 131: e29 –322, 2015.
outcomes research and functional genomics and translational biology interdisciplinary
working groups; and council on epidemiology and prevention. Circulation 113: 1807– 199. Mussolino C, Mlambo T, Cathomen T. Proven and novel strategies for efficient editing
1816, 2006. of the human genome. Curr Opin Pharmacol 24: 105–112, 2015.

180. Masaki H, Ishikawa T, Takahashi S, Okumura M, Sakai N, Haga M, Kominami K, Migita 200. Mwinyi J, Johne A, Bauer S, Roots I, Gerloff T. Evidence for inverse effects of oatp-c
H, McDonald F, Shimada F, Sakurada K. Heterogeneity of pluripotent marker gene (slc21a6) 5 and 1b haplotypes on pravastatin kinetics. Clin Pharmacol Ther 75: 415–
expression in colonies generated in human ips cell induction culture. Stem Cell Res 1: 421, 2004.
105–115, 2007.
201. Nakagawa M, Koyanagi M, Tanabe K, Takahashi K, Ichisaka T, Aoi T, Okita K,
181. Matsa E, Burridge PW, Wu JC. Human stem cells for modeling heart disease and for Mochiduki Y, Takizawa N, Yamanaka S. Generation of induced pluripotent stem cells
drug discovery. Sci Transl Med 6: ps6, 2014. without myc from mouse and human fibroblasts. Nat Biotechnol 26: 101–106, 2008.

182. Matsa E, Denning C. In vitro uses of human pluripotent stem cell-derived cardiomy- 202. Narsinh KH, Jia F, Robbins RC, Kay MA, Longaker MT, Wu JC. Generation of adult
ocytes. J Cardiovasc Transl Res 5: 581–592, 2012. human induced pluripotent stem cells using nonviral minicircle DNA vectors. Nat
Protoc 6: 78 – 88, 2011.
183. Matsa E, Dixon JE, Medway C, Georgiou O, Patel MJ, Morgan K, Kemp PJ, Staniforth
A, Mellor I, Denning C. Allele-specific rna interference rescues the long-qt syndrome 203. Navarrete EG, Liang P, Lan F, Sanchez-Freire V, Simmons C, Gong T, Sharma A,
phenotype in human-induced pluripotency stem cell cardiomyocytes. Eur Heart J 5: Burridge PW, Patlolla B, Lee AS, Wu H, Beygui RE, Wu SM, Robbins RC, Bers DM, Wu
1078 –1087, 2014. JC. Screening drug-induced arrhythmia [corrected] using human induced pluripotent
stem cell-derived cardiomyocytes and low-impedance microelectrode arrays. Circu-
184. Matsa E, Rajamohan D, Dick E, Young L, Mellor I, Staniforth A, Denning C. Drug lation 128 Suppl 1: S3–13, 2013.
evaluation in cardiomyocytes derived from human induced pluripotent stem cells
carrying a long qt syndrome type 2 mutation. Eur Heart J 32: 952–962, 2011. 204. Nihongaki Y, Yamamoto S, Kawano F, Suzuki H, Sato M. Crispr-cas9-based photo-
activatable transcription system. Chem Biol 22: 169 –174, 2014.
185. May JE, Xu J, Morse HR, Avent ND, Donaldson C. Toxicity testing: the search for an
in vitro alternative to animal testing. Br J Biomed Sci 66: 160 –165, 2009. 205. Nimura K, Ura K, Shiratori H, Ikawa M, Okabe M, Schwartz RJ, Kaneda Y. A histone
h3 lysine 36 trimethyltransferase links nkx2-5 to wolf-hirschhorn syndrome. Nature
186. Mehta A, Chung YY, Ng A, Iskandar F, Atan S, Wei H, Dusting G, Sun W, Wong P, 460: 287–291, 2009.
Shim W. Pharmacological response of human cardiomyocytes derived from virus-free
induced pluripotent stem cells. Cardiovasc Res 91: 577–586, 2011. 206. Novak A, Barad L, Lorber A, Gherghiceanu M, Reiter I, Eisen B, Eldor L, Itskovitz-
Eldor J, Eldar M, Arad M, Binah O. Functional abnormalities in ipsc-derived cardio-
187. Mehta A, Sequiera GL, Ramachandra CJ, Sudibyo Y, Chung Y, Sheng J, Wong KY, Tan myocytes generated from cpvt1 and cpvt2 patients carrying ryanodine or calseques-
TH, Wong P, Liew R, Shim W. Re-trafficking of herg reverses long qt syndrome 2 trin mutations. J Cell Mol Med 19: 2006 –2018, 2014.
phenotype in human ips-derived cardiomyocytes. Cardiovasc Res 102: 497–506, 2014.
207. Novak A, Barad L, Zeevi-Levin N, Shick R, Shtrichman R, Lorber A, Itskovitz-Eldor J,
188. Mendjan S, Mascetti VL, Ortmann D, Ortiz M, Karjosukarso DW, Ng Y, Moreau T, Binah O. Cardiomyocytes generated from cpvtd307h patients are arrhythmogenic in
Pedersen RA. Nanog and cdx2 pattern distinct subtypes of human mesoderm during response to beta-adrenergic stimulation. J Cell Mol Med 16: 468 – 482, 2012.
exit from pluripotency. Cell Stem Cell 15: 310 –325, 2014.
208. Nunes SS, Miklas JW, Liu J, Aschar-Sobbi R, Xiao Y, Zhang B, Jiang J, Masse S, Gagliardi
189. Mercer AC, Gaj T, Fuller RP, Barbas CF. Chimeric tale recombinases with program- M, Hsieh A, Thavandiran N, Laflamme MA, Nanthakumar K, Gross GJ, Backx PH,
mable DNA sequence specificity. Nucleic Acids Res 40: 11163–11172, 2012. Keller G, Radisic M. Biowire: a platform for maturation of human pluripotent stem
cell-derived cardiomyocytes. Nat Methods 10: 781–787, 2013.
190. Merriman B, Ion Torrent R, Team D, Rothberg JM. Progress in ion torrent semicon-
ductor chip based sequencing. Electrophoresis 33: 3397–3417, 2012. 209. Okita K, Yamakawa T, Matsumura Y, Sato Y, Amano N, Watanabe A, Goshima N,
Yamanaka S. An efficient nonviral method to generate integration-free human-in-
191. Meyer T, Leisgen C, Gonser B, Gunther E. Qt-screen: high-throughput cardiac safety duced pluripotent stem cells from cord blood and peripheral blood cells. Stem Cells
pharmacology by extracellular electrophysiology on primary cardiac myocytes. Assay 31: 458 – 466, 2013.
Drug Dev Technol 2: 507–514, 2004.
210. Olivotto I, d’Amati G, Basso C, Van Rossum A, Patten M, Emdin M, Pinto Y, Tomberli
192. Milan DJ, MacRae CA. Animal models for arrhythmias. Cardiovasc Res 67: 426 – 437, B, Camici PG, Michels M. Defining phenotypes and disease progression in sarcomeric
2005. cardiomyopathies: Contemporary role of clinical investigations. Cardiovasc Res 105:
409 – 423, 2015.
193. Mioulane M, Foldes G, Ali NN, Schneider MD, Harding SE. Development of high
content imaging methods for cell death detection in human pluripotent stem cell- 212. Paige SL, Osugi T, Afanasiev OK, Pabon L, Reinecke H, Murry CE. Endogenous
derived cardiomyocytes. J Cardiovasc Transl Res 5: 593– 604, 2012. wnt/beta-catenin signaling is required for cardiac differentiation in human embryonic
stem cells. PLoS One 5: e11134, 2010.
194. Miyoshi N, Ishii H, Nagano H, Haraguchi N, Dewi DL, Kano Y, Nishikawa S, Tane-
mura M, Mimori K, Tanaka F, Saito T, Nishimura J, Takemasa I, Mizushima T, Ikeda M, 213. Park IH, Arora N, Huo H, Maherali N, Ahfeldt T, Shimamura A, Lensch MW, Cowan
Yamamoto H, Sekimoto M, Doki Y, Mori M. Reprogramming of mouse and human C, Hochedlinger K, Daley GQ. Disease-specific induced pluripotent stem cells. Cell
cells to pluripotency using mature microRNAs. Cell Stem Cell 8: 633– 638, 2011. 134: 877– 886, 2008.

195. Moore GE. Cramming more components onto integrated circuits. Electronics 38: 214. Pattanayak V, Guilinger JP, Liu DR. Determining the specificities of talens, cas9, and
114 –117, 1965. other genome-editing enzymes. Methods Enzymol 546: 47–78, 2014.

196. Mordwinkin NM, Lee AS, Wu JC. Patient-specific stem cells and cardiovascular drug 215. Paull D, Sevilla A, Zhou H, Hahn AK, Kim H, Napolitano C, Tsankov A, Shang L,
discovery. JAMA 310: 2039 –2040, 2013. Krumholz K, Jagadeesan P, Woodard CM, Sun B, Vilboux T, Zimmer M, Forero E,

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1123


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

Moroziewicz DN, Martinez H, Malicdan MC, Weiss KA, Vensand LB, Dusenberry CR, 233a.Sayed N, Liu C, Wu JC. Translation of human-induced pluripotent stem cells: from
Polus H, Sy KT, Kahler DJ, Gahl WA, Solomon SL, Chang S, Meissner A, Eggan K, clinical trial in a dish to precision medicine. J Am Coll Cardiol 67: 2161–2176, 2016.
Noggle SA. Automated, high-throughput derivation, characterization and differentia-
tion of induced pluripotent stem cells. Nat Methods 12: 885– 892, 2015. 234. Schmid-Burgk JL, Schmidt T, Kaiser V, Honing K, Hornung V. A ligation-independent
cloning technique for high-throughput assembly of transcription activator-like effec-
216. Pereira GC, Silva AM, Diogo CV, Carvalho FS, Monteiro P, Oliveira PJ. Drug-induced tor genes. Nat Biotechnol 31: 76 – 81, 2013.
cardiac mitochondrial toxicity and protection: from doxorubicin to carvedilol. Curr
Pharm Des 17: 2113–2129, 2011. 235. Schroeder M, Niebruegge S, Werner A, Willbold E, Burg M, Ruediger M, Field LJ,
Lehmann J, Zweigerdt R. Differentiation and lineage selection of mouse embryonic
217. Phan D, Rasmussen TL, Nakagawa O, McAnally J, Gottlieb PD, Tucker PW, Richard- stem cells in a stirred bench scale bioreactor with automated process control. Bio-
son JA, Bassel-Duby R, Olson EN. Bop, a regulator of right ventricular heart develop- technol Bioeng 92: 920 –933, 2005.
ment, is a direct transcriptional target of mef2c in the developing heart. Development
132: 2669 –2678, 2005. 237. Sesti F, Abbott GW, Wei J, Murray KT, Saksena S, Schwartz PJ, Priori SG, Roden DM,
George AL Jr, Goldstein SA. A common polymorphism associated with antibiotic-
218. Pouton CW, Haynes JM. Embryonic stem cells as a source of models for drug discov- induced cardiac arrhythmia. Proc Natl Acad Sci USA 97: 10613–10618, 2000.
ery. Nat Rev Drug Discov 6: 605– 616, 2007.
238. Shanks N, Greek R, Greek J. Are animal models predictive for humans? PEHM 4: 2,
219. Pretorius RW, Gataric G, Swedlund SK, Miller JR. Reducing the risk of adverse drug 2009.
events in older adults. American Family Physician 87: 329 –334, 2013.
239. Sharma A, Marceau C, Hamaguchi R, Burridge PW, Rajarajan K, Churko JM, Wu H,
220. Price PS, Keenan RE, Swartout JC. Characterizing interspecies uncertainty using data Sallam KI, Matsa E, Sturzu AC, Che Y, Ebert A, Diecke S, Liang P, Red-Horse K,
from studies of anti-neoplastic agents in animals and humans. Toxicol Appl Pharmacol Carette JE, Wu SM, Wu JC. Human induced pluripotent stem cell-derived cardiomy-
233: 64 –70, 2008. ocytes as an in vitro model for coxsackievirus b3-induced myocarditis and antiviral
drug screening platform. Circ Res 115: 556 –566, 2014.
221. Puri MC, Nagy A. Concise review: embryonic stem cells versus induced pluripotent
stem cells: the game is on. Stem Cells 30: 10 – 4, 2012. 240. Shen B, Zhang W, Zhang J, Zhou J, Wang J, Chen L, Wang L, Hodgkins A, Iyer V, Huang
X, Skarnes WC. Efficient genome modification by crispr-cas9 nickase with minimal
222. Quail MA, Smith M, Coupland P, Otto TD, Harris SR, Connor TR, Bertoni A, Swerd- off-target effects. Nat Methods 11: 399 – 402, 2014.
low HP, Gu Y. A tale of three next generation sequencing platforms: Comparison of
ion torrent, pacific biosciences and illumina miseq sequencers. BMC Genomics 13: 341, 241. Shirai M, Osugi T, Koga H, Kaji Y, Takimoto E, Komuro I, Hara J, Miwa T, Yamauchi-
2012. Takihara K, Takihara Y. The polycomb-group gene rae28 sustains nkx2.5/csx expres-
sion and is essential for cardiac morphogenesis. J Clin Invest 110: 177–184, 2002.
223. Quigley EM. Cisapride: what can we learn from the rise and fall of a prokinetic? J Dig
Dis 12: 147–156, 2011. 242. Sirenko O, Crittenden C, Callamaras N, Hesley J, Chen YW, Funes C, Rusyn I, Anson
B, Cromwell EF. Multiparameter in vitro assessment of compound effects on cardi-
224. Raha D, Hong M, Snyder M. Chip-seq: a method for global identification of regulatory omyocyte physiology using ipsc cells. J Biomol Screen 18: 39 –53, 2013.
elements in the genome. Curr Protoc Mol Biol 21: 1–14, 2010.
243. Siu CW, Lee YK, Ho JC, Lai WH, Chan YC, Ng KM, Wong LY, Au KW, Lau YM, Zhang
225. Rais Y, Zviran A, Geula S, Gafni O, Chomsky E, Viukov S, Mansour AA, Caspi I, J, Lay KW, Colman A, Tse HF. Modeling of lamin a/c mutation premature cardiac aging
Krupalnik V, Zerbib M, Maza I, Mor N, Baran D, Weinberger L, Jaitin DA, Lara-Astiaso using patient-specific induced pluripotent stem cells. Aging 4: 803– 822, 2012.
D, Blecher-Gonen R, Shipony Z, Mukamel Z, Hagai T, Gilad S, Amann-Zalcenstein D,
Tanay A, Amit I, Novershtern N, Hanna JH. Deterministic direct reprogramming of 244. Smith C, Gore A, Yan W, Abalde-Atristain L, Li Z, He C, Wang Y, Brodsky RA, Zhang
somatic cells to pluripotency. Nature 502: 65–70, 2013. K, Cheng L, Ye Z. Whole-genome sequencing analysis reveals high specificity of
crispr/cas9 and talen-based genome editing in human ipscs. Cell Stem Cell 15: 12–13,
226. Rajamohan D, Matsa E, Kalra S, Crutchley J, Patel A, George V, Denning C. Current 2014.
status of drug screening and disease modelling in human pluripotent stem cells. Bioes-
says 35: 281–298, 2013. 245. Smith JG, Celiz AD, Patel AK, Short RD, Alexander MR, Denning C. Scaling human
pluripotent stem cell expansion and differentiation: Are cell factories becoming a
227. Raval KK, Tao R, White BE, De Lange WJ, Koonce CH, Yu J, Kishnani PS, Thomson JA, reality? Regen Med 10: 925–930, 2015.
Mosher DF, Ralphe JC, Kamp TJ. Pompe disease results in a golgi-based glycosylation
deficit in human induced pluripotent stem cell-derived cardiomyocytes. J Biol Chem 246. Soldner F, Hockemeyer D, Beard C, Gao Q, Bell GW, Cook EG, Hargus G, Blak A,
290: 3121–3136, 2015. Cooper O, Mitalipova M, Isacson O, Jaenisch R. Parkinson’s disease patient-derived
induced pluripotent stem cells free of viral reprogramming factors. Cell 136: 964 –977,
228. Reuter Jason A, Spacek DV, Snyder MP. High-throughput sequencing technologies. 2009.
Mol Cell 58: 586 –597, 2015.
247. Solloway MJ, Harvey RP. Molecular pathways in myocardial development: A stem cell
229. Rhoads A, Au KF. Pacbio sequencing and its applications. Genomics Proteomics Bioin- perspective. Cardiovasc Res 58: 264 –277, 2003.
formatics 13: 278 –289, 2015.
248. Spears DA, Gollob MH. Genetics of inherited primary arrhythmia disorders. Appl Clin
230. Rossi D, Rasi S, Franceschetti S, Capello D, Castelli A, De Paoli L, Ramponi A, Chiap-
Genet 8: 215–233, 2015.
pella A, Pogliani EM, Vitolo U, Kwee I, Bertoni F, Conconi A, Gaidano G. Analysis of
the host pharmacogenetic background for prediction of outcome and toxicity in 249. Spies D, Ciaudo C. Dynamics in transcriptomics: advancements in rna-seq time
diffuse large b-cell lymphoma treated with r-chop21. Leukemia 23: 1118 –1126, 2009. course and downstream analysis. Comput Struct Biotechnol J 13: 469 – 477, 2015.

231. Rouhani F, Kumasaka N, de Brito MC, Bradley A, Vallier L, Gaffney D. Genetic 250. Stadtfeld M, Hochedlinger K. Induced pluripotency: History, mechanisms, applica-
background drives transcriptional variation in human induced pluripotent stem cells. tions. Genes Dev 24: 2239 –2263, 2010.
PLoS Genet 10: e1004432, 2014.
251. Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K. Induced pluripotent stem
232. Sallam K, Li Y, Sager PT, Houser SR, Wu JC. Finding the rhythm of sudden cardiac cells generated without viral integration. Science 322: 945–949, 2008.
death: New opportunities using induced pluripotent stem cell-derived cardiomyo-
cytes. Circ Res 116: 1989 –2004, 2015. 252. Steiner D, Khaner H, Cohen M, Even-Ram S, Gil Y, Itsykson P, Turetsky T, Idelson M,
Aizenman E, Ram R, Berman-Zaken Y, Reubinoff B. Derivation, propagation and
232a.Sanchez-Freire V, Lee AS, Hu S, Abilez OJ, Liang P, Lan F, Huber BC, Ong SG, Hong controlled differentiation of human embryonic stem cells in suspension. Nat Biotech-
WX, Huang M, Wu JC. Effect of human donor cell source on differentiation and nol 28: 361–364, 2010.
function of cardiac induced pluripotent stem cells. Am J Cell Cardiol 64: 436 – 448,
2014. 253. Storchova Z, Kuffer C. The consequences of tetraploidy and aneuploidy. J Cell Sci 121:
3859 –3866, 2008.
233. Sato Y, Kobayashi H, Higuchi T, Shimada Y, Era T, Kimura S, Eto Y, Ida H, Ohashi T.
Disease modeling and lentiviral gene transfer in patient-specific induced pluripotent 254. Sun N, Yazawa M, Liu J, Han L, Sanchez-Freire V, Abilez OJ, Navarrete EG, Hu S,
stem cells from late-onset pompe disease patient. Mol Ther 2: 15023, 2015. Wang L, Lee A, Pavlovic A, Lin S, Chen R, Hajjar RJ, Snyder MP, Dolmetsch RE, Butte

1124 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
HUMAN INDUCED PLURIPOTENT STEM CELLS

MJ, Ashley EA, Longaker MT, Robbins RC, Wu JC. Patient-specific induced pluripo- 273. Tomlinson B, Hu M, Lee VW, Lui SS, Chu TT, Poon EW, Ko GT, Baum L, Tam LS, Li
tent stem cells as a model for familial dilated cardiomyopathy. Sci Transl Med 4: EK. Abcg2 polymorphism is associated with the low-density lipoprotein cholesterol
130ra47, 2012. response to rosuvastatin. Clin Pharmacol Ther 87: 558 –562, 2010.

255. Suzuki K, Yu C, Qu J, Li M, Yao X, Yuan T, Goebl A, Tang S, Ren R, Aizawa E, Zhang 274. Tsai SQ, Wyvekens N, Khayter C, Foden JA, Thapar V, Reyon D, Goodwin MJ, Aryee
F, Xu X, Rupa Soligalla D, Chen F, Kim J, Na Kim Y, Liao HK, Benner C, Concepcion MJ, Joung JK. Dimeric crispr rna-guided foki nucleases for highly specific genome
Esteban R, Jin Y, Liu GH, Li Y, Izpisua Belmonte JC. Targeted gene correction mini- editing. Nat Biotech 32: 569 –576, 2014.
mally impacts whole-genome mutational load in human-disease-specific induced plu-
ripotent stem cell clones. Cell Stem Cell 15: 31–36, 2014. 275. Tse HF, Ho JC, Choi SW, Lee YK, Butler AW, Ng KM, Siu CW, Simpson MA, Lai WH,
Chan YC, Au KW, Zhang J, Lay KW, Esteban MA, Nicholls JM, Colman A, Sham PC.
256. Swanton C, Nicke B, Schuett M, Eklund AC, Ng C, Li Q, Hardcastle T, Lee A, Roy R, Patient-specific induced-pluripotent stem cells-derived cardiomyocytes recapitulate
East P, Kschischo M, Endesfelder D, Wylie P, Kim SN, Chen JG, Howell M, Ried T, the pathogenic phenotypes of dilated cardiomyopathy due to a novel des mutation
Habermann JK, Auer G, Brenton JD, Szallasi Z, Downward J. Chromosomal instability identified by whole exome sequencing. Hum Mol Genet 22: 1395–1403, 2013.
determines taxane response. Proc Natl Acad Sci USA 106: 8671– 8676, 2009.
276. Tzatzalos E, Abilez OJ, Shukla P, Wu JC. Engineered heart tissues and induced pluri-
257. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. potent stem cells: macro- and microstructures for disease modeling, drug screening,
Induction of pluripotent stem cells from adult human fibroblasts by defined factors. and translational studies. Adv Drug Deliv Rev 15: 213–216, 2015.
Cell 131: 861– 872, 2007.
277. Uosaki H, Fukushima H, Takeuchi A, Matsuoka S, Nakatsuji N, Yamanaka S, Ya-
259. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic mashita JK. Efficient and scalable purification of cardiomyocytes from human embry-
and adult fibroblast cultures by defined factors. Cell 126: 663– 676, 2006. onic and induced pluripotent stem cells by vcam1 surface expression. PLoS One 6:
e23657, 2011.
260. Takaya T, Kawamura T, Morimoto T, Ono K, Kita T, Shimatsu A, Hasegawa K.
Identification of p300-targeted acetylated residues in gata4 during hypertrophic re- 278. Vaissiere T, Sawan C, Herceg Z. Epigenetic interplay between histone modifications
sponses in cardiac myocytes. J Biol Chem 283: 9828 –9835, 2008. and DNA methylation in gene silencing. Mutat Res 659: 40 – 48, 2008.

261. Tam PP, Loebel DA. Gene function in mouse embryogenesis: get set for gastrulation. 279. Van Dijk EL, Auger H, Jaszczyszyn Y, Thermes C. Ten years of next-generation
Nat Rev Genet 8: 368 –381, 2007. sequencing technology. Trends Genet 30: 418 – 426, 2014.

262. Tanaka A, Yuasa S, Mearini G, Egashira T, Seki T, Kodaira M, Kusumoto D, Kuroda Y, 280. Van Hoof D, Dormeyer W, Braam SR, Passier R, Monshouwer-Kloots J, Ward-van
Oostwaard D, Heck AJ, Krijgsveld J, Mummery CL. Identification of cell surface
Okata S, Suzuki T, Inohara T, Arimura T, Makino S, Kimura K, Kimura A, Furukawa T,
proteins for antibody-based selection of human embryonic stem cell-derived cardio-
Carrier L, Node K, Fukuda K. Endothelin-1 induces myofibrillar disarray and contrac-
myocytes. J Proteome Res 9: 1610 –1618, 2010.
tile vector variability in hypertrophic cardiomyopathy-induced pluripotent stem cell-
derived cardiomyocytes. J Am Heart Assoc 3: e001263, 2014. 281. Veres A, Gosis BS, Ding Q, Collins R, Ragavendran A, Brand H, Erdin S, Talkowski ME,
Musunuru K. Low incidence of off-target mutations in individual crispr-cas9 and talen
263. Tanwar V, Bylund JB, Hu J, Yan J, Walthall JM, Mukherjee A, Heaton WH, Wang WD,
targeted human stem cell clones detected by whole-genome sequencing. Cell Stem
Potet F, Rai M, Kupershmidt S, Knapik EW, Hatzopoulos AK. Gremlin 2 promotes
Cell 15: 27–30, 2014.
differentiation of embryonic stem cells to atrial fate by activation of the jnk signaling
pathway. Stem Cells 32: 1774 –1788, 2014. 282. Vicini P, Fields O, Lai E, Litwack ED, Martin AM, Morgan TM, Pacanowski MA,
Papaluca M, Perez OD, Ringel MS, Robson M, Sakul H, Vockley J, Zaks T, Dolsten M,
264. Terrenoire C, Wang K, Tung KW, Chung WK, Pass RH, Lu JT, Jean JC, Omari A,
Sogaard M. Precision medicine in the age of big data: The present and future role of
Sampson KJ, Kotton DN, Keller G, Kass RS. Induced pluripotent stem cells used to
large scale unbiased sequencing in drug discovery and development. Clin Pharmacol
reveal drug actions in a long qt syndrome family with complex genetics. J Gen Physiol
Ther 99: 198 –207, 2016.
141: 61–72, 2013.
283. Visscher H, Ross CJ, Rassekh SR, Barhdadi A, Dube MP, Al-Saloos H, Sandor GS,
265. Tewey KM, Chen GL, Nelson EM, Liu LF. Intercalative antitumor drugs interfere with
Caron HN, van Dalen EC, Kremer LC, van der Pal HJ, Brown AM, Rogers PC, Phillips
the breakage-reunion reaction of mammalian DNA topoisomerase II. J Biol Chem 259:
MS, Rieder MJ, Carleton BC, Hayden MR, Canadian Pharmacogenomics Network for
9182–9187, 1984. Drug Safety. Pharmacogenomic prediction of anthracycline-induced cardiotoxicity in
children. J Clin Oncol 30: 1422–1428, 2012.
266. Thavandiran N, Dubois N, Mikryukov A, Masse S, Beca B, Simmons CA, Deshpande
VS, McGarry JP, Chen CS, Nanthakumar K, Keller GM, Radisic M, Zandstra PW. 284. Visscher H, Ross CJD, Rassekh SR, Sandor GSS, Caron HN, van Dalen EC, Kremer
Design and formulation of functional pluripotent stem cell-derived cardiac microtis- LC, van der Pal HJ, Rogers PC, Rieder MJ, Carleton BC, Hayden MR. Validation of
sues. Proc Natl Acad Sci USA 110: E4698 – 4707, 2013. variants in slc28a3 and ugt1a6 as genetic markers predictive of anthracycline-induced
cardiotoxicity in children. Pediatr Blood Cancer 60: 1375–1381, 2013.
267. Thomas RJ, Anderson D, Chandra A, Smith NM, Young LE, Williams D, Denning C.
Automated, scalable culture of human embryonic stem cells in feeder-free conditions. 285. Volkova M, Russell R 3rd. Anthracycline cardiotoxicity: Prevalence, pathogenesis and
Biotechnol Bioeng 102: 1636 –1644, 2009. treatment. Curr Cardiol Rev 7: 214 –220, 2011.
268. Thompson JF, Steinmann KE. Single molecule sequencing with a heliscope genetic 286. Wall JD, Tang LF, Zerbe B, Kvale MN, Kwok PY, Schaefer C, Risch N. Estimating
analysis system. Curr Protoc Mol Biol 7: 10, 2010. genotype error rates from high-coverage next-generation sequence data. Genome Res
24: 1734 –1739, 2014.
269. Thompson R, Drew CJ, Thomas RH. Next generation sequencing in the clinical
domain: clinical advantages, practical, and ethical challenges. Adv Protein Chem Struct 287. Wandelt S, Bux M, Leser U. Trends in genome compression. Curr Bioinformatics 9:
Biol 89: 27– 63, 2012. 315–326, 2014.

270. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, 288. Wandelt S, Rheinländer A, Bux M, Thalheim L, Haldemann B, Leser U. Data manage-
Jones JM. Embryonic stem cell lines derived from human blastocysts. Science 282: ment challenges in next generation sequencing. Datenbank-Spektrum 12: 161–171,
1145–1147, 1998. 2012.

271. Thorn CF, Oshiro C, Marsh S, Hernandez-Boussard T, McLeod H, Klein TE, Altman 289. Wang G, McCain ML, Yang L, He A, Pasqualini FS, Agarwal A, Yuan H, Jiang D, Zhang
RB. Doxorubicin pathways: pharmacodynamics and adverse effects. Pharmacogenet D, Zangi L, Geva J, Roberts AE, Ma Q, Ding J, Chen J, Wang DZ, Li K, Wang J, Wanders
Genomics 21: 440 – 446, 2011. RJ, Kulik W, Vaz FM, Laflamme MA, Murry CE, Chien KR, Kelley RI, Church GM,
Parker KK, Pu WT. Modeling the mitochondrial cardiomyopathy of barth syndrome
272. Tohyama S, Hattori F, Sano M, Hishiki T, Nagahata Y, Matsuura T, Hashimoto H, with induced pluripotent stem cell and heart-on-chip technologies. Nat Med 20:
Suzuki T, Yamashita H, Satoh Y, Egashira T, Seki T, Muraoka N, Yamakawa H, Ohgino 616 – 623, 2014.
Y, Tanaka T, Yoichi M, Yuasa S, Murata M, Suematsu M, Fukuda K. Distinct metabolic
flow enables large-scale purification of mouse and human pluripotent stem cell-de- 290. Wang Y, Liang P, Lan F, Wu H, Lisowski L, Gu M, Hu S, Kay MA, Urnov FD, Shinnawi
rived cardiomyocytes. Cell Stem Cell 12: 127–137, 2013. R, Gold JD, Gepstein L, Wu JC. Genome editing of isogenic human induced pluripo-

Physiol Rev • VOL 96 • JULY 2016 • www.prv.org 1125


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.
MATSA ET AL.

tent stem cells recapitulates long qt phenotype for drug testing. J Am Coll Cardiol 64: progenitor cells develop from a kdr⫹ embryonic-stem-cell-derived population. Na-
451– 459, 2014. ture 453: 524 –528, 2008.

291. Wang Z. An integrated chip for the high-throughput synthesis of transcription activa- 308. Yang L, Yang JL, Byrne S, Pan J, Church GM. Crispr/cas9-directed genome editing of
tor-like effectors. Angew Chem Int Ed Engl 51: 8505– 8508, 2012. cultured cells. Curr Protoc Mol Biol 107: 31 1 1 1 17524 –31, 2014.

292. Wang Z, Oron E, Nelson B, Razis S, Ivanova N. Distinct lineage specification roles for 309. Yang X, Pabon L, Murry CE. Engineering adolescence: maturation of human pluripo-
nanog, oct4, and sox2 in human embryonic stem cells. Cell Stem Cell 10: 440 –554, tent stem cell-derived cardiomyocytes. Circ Res 114: 511–523, 2014.
2012.
310. Yasuno T, Osafune K, Sakurai H, Asaka I, Tanaka A, Yamaguchi S, Yamada K, Hitomi
293. Warren L, Manos PD, Ahfeldt T, Loh YH, Li H, Lau F, Ebina W, Mandal PK, Smith ZD, H, Arai S, Kurose Y, Higaki Y, Sudo M, Ando S, Nakashima H, Saito T, Kaneoka H.
Meissner A, Daley GQ, Brack AS, Collins JJ, Cowan C, Schlaeger TM, Rossi DJ. Highly Functional analysis of ipsc-derived myocytes from a patient with carnitine palmitoyl-
efficient reprogramming to pluripotency and directed differentiation of human cells transferase II deficiency. Biochem Biophys Res Commun 448: 175–181, 2014.
with synthetic modified mRNA. Cell Stem Cell 7: 618 – 630, 2010.
311. Yazawa M, Hsueh B, Jia X, Pasca AM, Bernstein JA, Hallmayer J, Dolmetsch RE. Using
294. Warren L, Ni Y, Wang J, Guo X. Feeder-free derivation of human induced pluripotent induced pluripotent stem cells to investigate cardiac phenotypes in timothy syn-
stem cells with messenger RNA. Sci Rep 2: 657, 2012. drome. Nature 471: 230 –234, 2011.

312. Yazawa M, Hsueh B, Jia X, Pasca AM, Bernstein JA, Hallmayer J, Dolmetsch RE. Using
295. Watanabe Y, Zaffran S, Kuroiwa A, Higuchi H, Ogura T, Harvey RP, Kelly RG, Buck-
induced pluripotent stem cells to investigate cardiac phenotypes in timothy syn-
ingham M. Fibroblast growth factor 10 gene regulation in the second heart field by
drome. Nature 471: 230 –234, 2011.
tbx1, nkx2-5, and islet1 reveals a genetic switch for down-regulation in the myocar-
dium. Proc Natl Acad Sci USA 109: 18273–18280, 2012. 313. Yip VL, Pirmohamed M. Expanding role of pharmacogenomics in the management of
cardiovascular disorders. Am J Cardiovasc Drugs 13: 151–162, 2013.
296. Weng Z, Kong CW, Ren L, Karakikes I, Geng L, He J, Chow MZ, Mok CF, Keung W,
Chow H, Leung AY, Hajjar RJ, Li RA, Chan CW. A simple, cost-effective but highly 314. Yokoo N, Baba S, Kaichi S, Niwa A, Mima T, Doi H, Yamanaka S, Nakahata T, Heike
efficient system for deriving ventricular cardiomyocytes from human pluripotent stem T. The effects of cardioactive drugs on cardiomyocytes derived from human induced
cells. Stem Cells Dev 23: 1704 –1716, 2014. pluripotent stem cells. Biochem Biophys Res Commun 387: 482– 488, 2009.

297. Wicks EC, Elliott PM. Genetics and metabolic cardiomyopathies. Herz 37: 598 – 610, 315. Yoshioka N, Gros E, Li HR, Kumar S, Deacon DC, Maron C, Muotri AR, Chi NC, Fu
2012. XD, Yu BD, Dowdy SF. Efficient generation of human ipscs by a synthetic self-
replicative RNA. Cell Stem Cell 13: -2462–54, 2013.
298. Willmann CA, Hemeda H, Pieper LA, Lenz M, Qin J, Joussen S, Sontag S, Wanek P,
Denecke B, Schuler HM, Zenke M, Wagner W. To clone or not to clone? Induced 316. Yu J, Chau KF, Vodyanik MA, Jiang J, Jiang Y. Efficient feeder-free episomal repro-
pluripotent stem cells can be generated in bulk culture. PLoS One 8: e65324, 2014. gramming with small molecules. PLoS One 6: e17557, 2011.

298a.Wilson KD, Shen P, Fung E, Karakikes I, Zhang A, InanlooRahatloo K, Odegaard J, 317. Yu J, Hu K, Smuga-Otto K, Tian S, Stewart R, Slukvin, II, Thomson JA. Human induced pluri-
Sallam K, Davis RW, Lui GK, Ashley EA, Scharfe C, Wu JC. A rapid, high-quality, potent stem cells free of vector and transgene sequences. Science 324: 797–801, 2009.
cost-effective, comprehensive and expandable targeted next generation sequencing
assay for inherited heart diseases. Circ Res 117: 603– 611, 2015. 318. Yu J, Hu K, Smuga-Otto K, Tian S, Stewart R, Slukvin II, Thomson JA. Human induced pluri-
potent stem cells free of vector and transgene sequences. Science 324: 797–801, 2009.
299. Wilson KD, Wu JC. Induced pluripotent stem cells. JAMA 313: 1613–1614, 2015.
319. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J,
300. Wojnowski L, Kulle B, Schirmer M, Schluter G, Schmidt A, Rosenberger A, Vonhof S, Jonsdottir GA, Ruotti V, Stewart R, Slukvin II, Thomson JA. Induced pluripotent stem
Bickeboller H, Toliat MR, Suk EK, Tzvetkov M, Kruger A, Seifert S, Kloess M, Hahn H, cell lines derived from human somatic cells. Science 318: 1917–1920,
Loeffler M, Nurnberg P, Pfreundschuh M, Trumper L, Brockmoller J, Hasenfuss G. 2007.
Nad(p)h oxidase and multidrug resistance protein genetic polymorphisms are associ-
320. Zeevi-Levin N, Itskovitz-Eldor J, Binah O. Cardiomyocytes derived from human plu-
ated with doxorubicin-induced cardiotoxicity. Circulation 112: 3754 –3762, 2005.
ripotent stem cells for drug screening. Pharmacol Ther 134: 180 –188, 2012.
300a.World Health Organization. World Health Statistics Report 2008. Geneva, Switzerland:
321. Zhang J, Klos M, Wilson GF, Herman AM, Lian X, Raval KK, Barron MR, Hou L,
WHO, 2008.
Soerens AG, Yu J, Palecek SP, Lyons GE, Thomson JA, Herron TJ, Jalife J, Kamp TJ.
Extracellular matrix promotes highly efficient cardiac differentiation of human
301. Wu H, Lee J, Vincent LG, Wang Q, Gu M, Lan F, Churko JM, Sallam KI, Matsa E,
pluripotent stem cells: the matrix sandwich method. Circ Res 111: 1125–1136,
Sharma A, Gold JD, Engler AJ, Xiang YK, Bers DM, Wu JC. Epigenetic regulation of
2012.
phosphodiesterases 2a and 3a underlies compromised beta-adrenergic signaling in an
ipsc model of dilated cardiomyopathy. Cell Stem Cell 17: 89 –100, 2015. 322. Zhang XH, Haviland S, Wei H, Saric T, Fatima A, Hescheler J, Cleemann L, Morad M.
Ca2⫹ signaling in human induced pluripotent stem cell-derived cardiomyocytes (ips-
302. Wu X, Scott DA, Kriz AJ, Chiu AC, Hsu PD, Dadon DB, Cheng AW, Trevino AE,
cm) from normal and catecholaminergic polymorphic ventricular tachycardia (cpvt)-
Konermann S, Chen S, Jaenisch R, Zhang F, Sharp PA. Genome-wide binding of the
afflicted subjects. Cell Calcium 54: 57–70, 2013.
crispr endonuclease cas9 in mammalian cells. Nat Biotechnol 32: 670 – 676, 2014.
323. Zhi D, Irvin MR, Gu CC, Stoddard AJ, Lorier R, Matter A, Rao DC, Srinivasasai-
303. Wu Y, Gao T, Wang X, Hu Y, Hu X, Hu Z, Pang J, Li Z, Xue J, Feng M, Wu L, Liang D.
nagendra V, Tiwari HK, Turner A, Broeckel U, Arnett DK. Whole-exome sequencing
Tale nickase mediates high efficient targeted transgene integration at the human and an ipsc-derived cardiomyocyte model provides a powerful platform for gene
multi-copy ribosomal DNA locus. Biochem Biophys Res Commun 446: 261–266, 2014. discovery in left ventricular hypertrophy. Front Genet 3: 92, 2012.
304. Wyles SP, Li X, Hrstka SC, Reyes S, Oommen S, Beraldi R, Edwards J, Terzic A, Olson 324. Zhou H, Wu S, Joo JY, Zhu S, Han DW, Lin T, Trauger S, Bien G, Yao S, Zhu Y, Siuzdak
TM, Nelson TJ. Modeling structural and functional deficiencies of rbm20 familial G, Scholer HR, Duan L, Ding S. Generation of induced pluripotent stem cells using
dilated cardiomyopathy using human induced pluripotent stem cells. Hum Mol Genet recombinant proteins. Cell Stem Cell 4: 381–384, 2009.
25: 254 –265, 2016.
325. Zhou W, Freed CR. Adenoviral gene delivery can reprogram human fibroblasts to
305. Xu C, Police S, Rao N, Carpenter MK. Characterization and enrichment of cardio- induced pluripotent stem cells. Stem Cells 27: 2667–2674, 2009.
myocytes derived from human embryonic stem cells. Circ Res 91: 501–508, 2002.
326. Zhu WZ, Xie Y, Moyes KW, Gold JD, Askari B, Laflamme MA. Neuregulin/erbb
306. Yang H, Wang H, Shivalila CS, Cheng AW, Shi L, Jaenisch R. One-step generation of signaling regulates cardiac subtype specification in differentiating human embryonic
mice carrying reporter and conditional alleles by crispr/cas-mediated genome engi- stem cells. Circ Res 107: 776 –786, 2010.
neering. Cell 154: 1370 –1379, 2013.
327. Zunder ER, Lujan E, Goltsev Y, Wernig M, Nolan GP. A continuous molecular road-
307. Yang L, Soonpaa MH, Adler ED, Roepke TK, Kattman SJ, Kennedy M, Henckaerts E, map to ipsc reprogramming through progression analysis of single-cell mass cytom-
Bonham K, Abbott GW, Linden RM, Field LJ, Keller GM. Human cardiovascular etry. Cell Stem Cell 16: 323–337, 2015.

1126 Physiol Rev • VOL 96 • JULY 2016 • www.prv.org


Downloaded from www.physiology.org/journal/physrev (031.223.142.132) on November 16, 2019.

Vous aimerez peut-être aussi