You are on page 1of 9

Review

Human Induced Pluripotent Stem


CellDerived Cardiomyocytes
Insights Into Molecular, Cellular, and Functional Phenotypes
Ioannis Karakikes, Mohamed Ameen, Vittavat Termglinchan, Joseph C. Wu

Abstract: Disease models are essential for understanding cardiovascular disease pathogenesis and developing
new therapeutics. The human induced pluripotent stem cell (iPSC) technology has generated significant
enthusiasm for its potential application in basic and translational cardiac research. Patient-specific iPSC-derived
cardiomyocytes offer an attractive experimental platform to model cardiovascular diseases, study the earliest
stages of human development, accelerate predictive drug toxicology tests, and advance potential regenerative
therapies. Harnessing the power of iPSC-derived cardiomyocytes could eliminate confounding species-specific
and interpersonal variations and ultimately pave the way for the development of personalized medicine for
cardiovascular diseases. However, the predictive power of iPSC-derived cardiomyocytes as a valuable model is
contingent on comprehensive and rigorous molecular and functional characterization.(Circ Res. 2015;117:80-88.
DOI: 10.1161/CIRCRESAHA.117.305365.)
Key Words: cardiovascular diseases cardiovascular disease modeling induced pluripotent stem cells
myocytes, cardiac precision medicine

Induced Pluripotent Stem CellDerived models are particularly important for cardiovascular research
Cardiomyocytes: A New and Versatile Human because the physiology of animal models is different from hu-
In Vitro Cardiomyocyte Model man cardiomyocytes. Particularly, considerable differences
Recent advances in genomics and molecular medicine prom- exist between cardiomyocytes from small animal models and
ise to revolutionize human health by enabling more precise human cardiomyocytes, including beating rates, energetics,
prediction, prevention, and treatment of cardiovascular diseas- Ca2+ cycling, myofilament composition, expression of key ion
es on an individual level.1 This precision medicine approach channels, and cellular electrophysiology. These differences in
is based on the ability to diagnose and stratify patients into physiology are substantially less between humans and large
different treatment groups by correlating a patients genotype animal models such as nonhuman primates, pigs, and dogs.3
with their cellular phenotype, and uncover how the genetic The recent advent of the human induced pluripotent stem
differences among people could influence their responses to cell (iPSC) technology,4 and an increasingly refined capacity
therapies. However, realizing this potential requires the devel- to differentiate iPSCs into disease-relevant cell types such
opment of accurate disease models. Models that recapitulate as cardiomyocytes (iPSC-CMs),5 provides an unprecedent-
individual patients disease at the molecular and cellular level ed opportunity for the generation of human patient-specific
could lead to a better understanding of the disease progression cells for use in disease modeling, personalized drug screen-
and pathogenesis and ultimately enable the prediction of indi- ing, and regenerative approaches toward precision medi-
vidual patients responses to targeted treatments. cine.6,7 Implementation of this unique and clinically relevant
Disease models have been and will continue to be in- model system presents a significant advantage in cardiovas-
strumental in providing important insights into the molecu- cular research as it can circumvent complications in translat-
lar basis of cardiovascular development and disease.2 The ing data from models across different species and biological
knowledge gained from studying transgenic animals and characteristics.
transformed cell lines has already been successfully applied iPSC-CMs offer several advantages over current in vitro
to understand human cardiovascular disease. Nevertheless, models such as immortalized cell lines, human cadaveric tis-
this translation would be significantly strengthened by the sue, and primary cultures of nonhuman animal origin. First,
availability of patient-specific in vitro models. Human-based the derivation of iPSC-CMs is at most minimally invasive

Original received March 1, 2015; revision received May 20, 2015; accepted May 22, 2015. In April 2015, the average time from submission to first
decision for all original research papers submitted to Circulation Research was 13.84 days.
From the Stanford Cardiovascular Institute (I.K., M.A., V.T., J.C.W.), Department of Medicine, Division of Cardiovascular Medicine (I.K., V.T., J.C.W.),
and Institute of Stem Cell Biology and Regenerative Medicine (J.C.W.), Stanford University School of Medicine, CA.
Correspondence to Ioannis Karakikes, PhD, or Joseph C. Wu, MD, PhD, Stanford Cardiovascular Institute, Stanford University School of Medicine, 265
Campus Dr, Room G1120B, Stanford, CA 94305. E-mail ioannis1@stanford.edu or joewu@stanford.edu
2015 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.117.305365

80
Karakikes et al Current Status of iPSC-Derived Cardiomyocytes 81

applied to iPSCs derived from distinct sources of somatic


Nonstandard Abbreviations and Acronyms
cells and reprogramming methods.2628,34,35 However, the resul-
AP action potential tant iPSC-CM population is a heterogeneous pool of atrial-,
BTHS Barth syndrome ventricular-, and nodal-like cells. As native atrial, nodal, and
DCM dilated cardiomyopathy ventricular myocytes possess distinct molecular and func-
iPSC induced pluripotent stem cell tional properties, coaxing human iPSC differentiation toward
iPSC-CM induced pluripotent stem cellderived cardiomyocytes specific cardiomyocyte subtypes remains challenging for the
LQT long QT current methodologies. In this respect, recent reports suggest
SR sarcoplasmic reticulum that pluripotent stem cells could be directed either to atrial-
like or to ventricular-like cardiomyocytes by modulating the
retinoic acid36,37 and Wnt signaling pathways.38 However, the
elucidation of the molecular mechanism(s) underlying the car-
(typically via skin biopsy or blood draws) and can theoreti-
cally provide an unlimited supply of human cardiomyocytes. diomyocyte subtype specification would be essential to fur-
Second, iPSC-CMs can be functionally characterized in vitro ther refine the current differentiation protocols and improve
to model the complex cellular physiology of cardiomyocytes. our understanding of lineage-specific development.
Third, iPSC-CMs recapitulate the genome of a subject, allow-
ing for the assessment of genotypephenotype associations. Molecular Profiling
During the past few years, there has been considerable At the molecular level, the differentiation of iPSCs toward the
progress in the iPSC-CM technology and its contributions to cardiomyocyte lineage is orchestrated by the sequential ex-
cardiovascular research are already well recognized. For ex- pression of distinct sets of genes in specific stages in a pattern
ample, iPSC models have been recently used to describe car- consistent with normal cardiac development: mesoderm for-
diac channelopathies such as long-QT syndromes (LQT1,810 mation (T and MIXL1), cardiogenic mesoderm (MESP1, ISL1,
LQT2,1113 LQT3/Brugada syndrome,14 and LQT8/Timothy and KDR), cardiac-specific progenitors (NKX2.5, GATA4,
syndrome15), catecholaminergic polymorphic ventricular TBX5, MEF2C, and HAND1/2), and structural genes encod-
tachycardia (CPVT),1618 arrhythmogenic right ventricular ing for sarcomeric-related proteins of terminal differentiated
dysplasia (ARVD),19,20 familial hypertrophic cardiomyopathy cardiomyocytes (MYL2, MYL7, MYH6, and TNNT2).26,34 Gene
(HCM),21 and familial dilated cardiomyopathy (DCM).22 As expression analysis has revealed that normal iPSC-CMs and
the iPSC-CM technology continues to evolve, it will greatly disease-specific iPSC-CMs expressed all the major cardiac
facilitate the study of inherited and acquired cardiovascular ion channel genes found in the adult left ventricular cardiac
diseases, infectious diseases, cardiovascular development, tissue, including sodium channel (SCN5A), L-type calcium
drug discovery, toxicology screening, and personalized cell channels (CACNA1C and CACNA1D), and potassium chan-
therapy (Figure1). nels (KCNH2 and KCNQ1).39 Furthermore, iPSC-CMs ex-
In this review article, we will highlight the current state of press genes that encode critical components of the Ca2+
iPSC-CMs, focusing on their phenotype and function. We will cycling machinery such as inositol trisphosphate receptor
discuss the molecular phenotypes, electrophysiological and (IPTR3), ryanodine receptor 2 (RYR2), sarcoplasmic reticu-
calcium handling properties, and bioenergetics. We will also lum (SR) Ca2+-ATPase (SERCA2), calsequestrin 2 (CASQ2),
explore what the future may hold for their use in cardiovascu- calreticulin (CALR), junctophilin 2 (JPH2), phospholam-
lar research, pharmacology, and regenerative medicine toward ban (PLN), sodium calcium exchanger (NCX1), and triadin
precision medicine. (TRDN). However, their relative expression differs from that
of the human adult ventricular tissue.17,40,41 Mitochondrial
Generation of iPSC-CMs complexes IV and genes involved in cholesterol metabolism
Studies with different model organisms have demonstrated (PRKAG1 and PRKAG2) as well as genes that confer protec-
that signaling pathways, such as activin/nodal/transforming tion against apoptotic and oxidative stress processes (BCL2L1
growth factor-, Wnt, and bone morphogenetic protein, play and SOD1, respectively) are also expressed in iPSC-CMs.42
pivotal roles in establishing the cardiovascular system.2325 By Taken together, these studies demonstrated that the gene ex-
mimicking endogenous developmental signaling cues, direct- pression in iPSC-CMs closely mirrors the patterns observed in
ed differentiation methodologies for generating iPSC-CMs human cardiomyocytes (Figure2).
have been developed.2628 Several permutations of growth From a disease modeling perspective, the faithful expres-
factors and small molecules have recently been reported to sion of disease-associated alleles is a prerequisite for proper
improve reproducibility and efficiency of iPSC-CM differen- manifestation of the disease phenotypes in iPSC-CMs. For ex-
tiation protocols in both adherent and suspension cultures.5 ample, mutations in genes encoding sarcomeric components,
Further purification of CMs from a mixed population of iPSC- including TNNT2 and MYH7, have been implicated in the 2
derived cells can be accomplished by nongenetic methods, in- most common forms of inherited cardiomyopathies: HCM and
cluding cell-surface markers,29,30 mitochondria-specific dyes,31 DCM. Mutations in the genes encoding potassium (KCNQ1
fluorescent probes,32 and glucose deprivation.33 Although and KCNH2), sodium (SCN5A), and calcium (CACNA1C)
iPSC lines seem to respond differently to developmental sig- channels are the most common cause of the LQT syndromes,
nals because of the intrinsic differences in their genetic back- which is usually inherited in an autosomal dominant man-
ground, these differentiation protocols have been successfully ner. Recently, iPSC-CMs have been derived from patients
82Circulation ResearchJune 19, 2015

Figure 1. Current applications of patient-specific induced pluripotent stem cellderived cardiomyocyte (iPSC-CM) technology.
iPSC-CMs have been used for disease modeling of inherited cardiomyopathies and channelopathies, regenerative therapies, drug
discovery, and cardiotoxicity testing, as well as for studying metabolic abnormalities and cardiac development.

harboring deleterious mutation that recapitulated key disease progressive lengthening of 3-dimensional iPSC-CM tissues
aspects of HCM, DCM, and LQT syndromes.8,9,1115,21,22 Such that mimics heart growth during development further im-
models are currently being used to decipher the complex geno- proved cell alignment and increased sarcomeric ultrastructural
typephenotype relationships and to determine disease effects organization.47 Despite these advances, the contractile proper-
of specific genetic variants. ties of iPSC-CMs and engineered tissues remain at rudimen-
tary levels.4750
Ultrastructural Features
To maximize the potential applications of iPSC-CMs in car- Electrophysiological Phenotypes and Ion
diovascular medicine, it is essential for these cells to recapitu- Channel Function
late the ultrastructural properties of adult cardiomyocytes. It Comprehensive analyses of the electrophysiological proper-
has been reported that early stage iPSC-CMs are small mor- ties of iPSC-CMs have been reported. Based on the action
phologically and exhibit an immature ultrastructure closely potential (AP) phenotypes recorded in isolated cells, iPSC-
resembling that of fetal CMs (ie, absence of T-tubules and CMs consist of a heterogeneous population categorized as
underdeveloped contractile machinery).4345 However, on atrial-, nodal-, or ventricular-like.15,34,5153 However, cell cul-
prolonged culture, there were significant improvements in ture conditions and differentiation protocols may influence
myofibril alignment, density, and morphology showing adult- these AP properties, possibly undermining the correctness of
like appearance of Z-disks, A-bands, I-bands, and H-zones, this classification.54,55 Although the direct electrophysiologi-
although no clear M-bands or T-tubules were observed.44,45 cal comparison between iPSC-CMs and human adult CMs
Similarly, by combining bioengineering approaches with is challenging because of experimental discrepancies, tissue
electric stimulation, Nunes et al46 developed a platform called heterogeneity, and disease status, it is well documented that
biowire that enables the generation of iPSC-CMs with ultra- iPSC-CMs display mixed AP phenotypes characterized by
structural properties similar to those seen in the native CMs. The relatively positive maximum diastolic potential and slower
Karakikes et al Current Status of iPSC-Derived Cardiomyocytes 83

Figure 2. Expression of key structural and functional genes in induced pluripotent stem cellderived cardiomyocytes (iPSC-CMs).
A, Schematic of the major structural and functional features of iPSC-CMs. In adult CMs, on membrane depolarization a small amount of Ca2+
influx induced by activation of voltage-dependent L-type Ca2+ channels (CACNAC1) triggers the release of Ca2+ from the sarcoplasmic reticulum
(SR) through the ryanodine receptors (ryanodine receptor 2 [RYR2]), termed Ca2+-induced Ca2+-release mechanism. The released Ca2+ ions diffuse
through the cytosolic space and bind to troponin C (TNNC1), resulting in the release of inhibition induced by troponin I (TNNI3), which activates the
sliding of thin and thick filaments, and lead to cardiac contraction. Recovery occurs as Ca2+ is extruded by the Na2+/Ca2+ exchanger (NCX1) and
returned to the SR by the sarco(endo)plasmic Ca2+-ATPase pumps on the nonjunctional region of the SR that are regulated by phospholamban
(PLN). This process is conserved in iPSC-CMs, but major differences exist, such as a nascent SR, the presence of an inositol 1,4,5-trisphosphate
releasable Ca2+ pool, and the complete absence of T-tubules. The genes encoding the major transmembrane ion channels involved in the generation
of action potential are also shown. B, Immunofluorescence staining of cardiac troponin T and -sarcomeric actinin in iPSC-CMs. C, Line-scan
images and spontaneous Ca2+ transients in iPSC-CMs. ACTC1 indicates actin, , cardiac muscle 1; CACNA1C, calcium channel, voltage-
dependent, L type, 1C subunit; IPTR3, inositol 1,4,5-trisphosphate receptor, type 3; KCNH2, potassium voltage-gated channel, subfamily H
(eag-related), member 2; KCNIP2, potassium channelinteracting protein 2; KCNJ2, potassium inwardly rectifying channel, subfamily J, member
2; KCNQ1, potassium voltage-gated channel, KQT-like subfamily, member 1; MYBPC3, myosin binding protein C; MYH6, myosin, heavy chain 6,
; MYH7, myosin, heavy chain 7, ; MYL2, myosin, light chain 2; MYL3, myosin, light chain 3; SCN5A, sodium channel, voltage-gated, type V, -
subunit; SERCA2, SR Ca2+-ATPase 2; TNNI3, troponin I type 3; TNNT2, troponin T type 2; TTN, titin; and TPM1, tropomyosin 1 ().
84Circulation ResearchJune 19, 2015

upstroke velocity when compared with the human native IK


counterparts.53 Moreover, recent studies suggest that the Three K+ currents (Ito, IKr, and IKs) have been recorded in
ventricular-like iPSC-CM subtype exhibits many key car- iPSC-CMs with maximum densities and activation proper-
diac electrophysiological properties analogous to those of ties comparable with the values reported for human cardiac
human CMs. The ventricular-like APs display properties of myocytes.8,53 By contrast, the density of IK1 is either absent
more mature human CMs, including a distinct plateau phase or significantly smaller than that reported for native ven-
(phase 2) after which repolarization accelerates (phase 3), tricular CMs. Importantly, IKr contributes to repolarization
with AP durations that are within the normal range of the of the cardiac AP, and block of IKr prolongs the ventricular
human electrocardiographic QT interval (Figure3A). AP, which is manifested by QT prolongation on the surface
Multiple ionic currents have been characterized in single ECG. Prolongation of the AP and associated increased QT
iPSC-CMs, including the sodium (INa), the L- and T-type calci- interval can lead to early afterdepolarizations on the cellu-
um (ICa,L and ICa,T), the hyperpolarization-activated pacemaker lar level and trigger the ventricular arrhythmia Torsades de
(If), the transient outward potassium (Ito), the inward rectifier pointes. Notably, by measuring AP duration and quantifying
potassium (IK1), and the rapid and slow activating components drug-induced arrhythmias, such as early afterdepolarizations
of the delayed rectifier potassium currents (IKr and IKs, respec- and delayed afterdepolarizations, drug-induced cardiotoxic-
ity profiles for healthy subjects, LQT syndrome, HCM, and
tively; Figure 3B). However, the functional properties of ad-
DCM patients were recapitulated at the single cell level us-
ditional currents, such as the ATP-sensitive K+ current (IK,ATP)
ing the iPSC-CM technology.39,57 Interestingly, the iPSC-CMs
and the Na+Ca+ exchange current (INCX), have not yet been re-
exhibited distinct responses to known cardiotoxic drugs when
ported in iPSC-CMs, whereas the atrial-selective ion current,
derived from healthy versus diseased individuals, suggesting
acetylcholine-activated K+ (IK,ACh), has recently been reported
that adverse drug responses could be accurately predicted in
in human embryonic stem cell-derived atrial-like CMs.36 We individual patients. This raises the prospect of proarrhythmic
briefly summarize some of these major channels below: drugs being readily identified early in a development program,
INa and those individuals at high risk for proarrhythmia could
iPSC-CMs have prominent Na+ currents with activation and be identified before deleterious drug exposures. The use of
inactivation gating characteristics that are analogous to those iPSC-CMs in screening for proarrhythmic potential of cur-
of native human ventricular CMs.14,53 rent and new drug entities in conjunction with in silico mod-
eling is a major focus of the Food and Drug Administration
ICa,L and ICa,T Comprehensive In Vitro Proarrhythmia Assay initiative.58
The activation and inactivation gating properties of ICa in iP- In summary, multiple voltage-gated ion channels are simi-
SC-CMs are similar to those obtained from human ventricular larly present in iPSC-CMs and adult CMs leading to charac-
myocytes.15,53 By contrast, although the ICa,T current has been teristic cardiac APs (Figure3B). But, significant differences
found in a subset of cells, its properties have not defined.56 do exist, such as reduced inward rectifier K+ currents and the
presence of prominent pacemaker currents. As a result, the
If iPSC-CMs exhibit spontaneous automaticity, which is not ob-
In ventricular-like iPSC-CMs, the presence of hyperpolar- served in healthy human ventricular myocytes. At the tissue
ization-activated If promotes phase 4 depolarization and thus level, these properties may be significantly altered when the
contribute to automaticity.53 cells are coupled in a functional syncytium and recent efforts

Figure 3. Comparison of action


potentials (APs) of ventricular-like
induced pluripotent stem cellderived
cardiomyocytes (iPSC-CMs) and adult
ventricular cardiomyocytes. A, Schematic
of ventricular APs. Phases 0 to 4 are the
rapid upstroke, early repolarization,
plateau, late repolarization, and diastole,
respectively. B, The ionic currents and
the genes that generate the currents with
schematics of the current trajectories
are shown above and below the APs.
For individual ion channel currents, the
voltage dependence of channel-gating
properties of ventricular-like iPSC-CMs is
remarkably analogous to adult ventricular
cardiomyocytes, but significant differences
also exist, such as reduced or absence of
the inward rectifier K+ currents (IK1) and the
presence of prominent pacemaker currents
that contribute to their automaticity. hiPSC
indicates human iPSC.
Karakikes et al Current Status of iPSC-Derived Cardiomyocytes 85

have been focused on using electric or mechanical stimulation pathways that may provide therapeutic relevance consistent
that seems to promote electrophysiological maturation.46-50 with the clinical type 2 diabetes mellitus subtype.61 However,
Future studies are clearly needed to characterize in detail the the genetic and the epigenetic basis underlying the observed
electrophysiological properties of iPSC-CMs at both single- cellular phenotypes and differential response to treatments
cell and tissue levels. were not examined. Similarly, by combining bioengineer-
ing and gene editing approaches, Wang et al62 modeled Barth
Excitation Contraction Coupling and Calcium syndrome (BTHS), an X-linked cardiac and skeletal mito-
Handling chondrial myopathy caused by mutation of the gene encod-
Myocardial contraction and relaxation are coordinated on a ing Tafazzin (TAZ). BTHS iPSC-CMs displayed an impaired
beat-to-beat basis by the orchestrated cycling of calcium from biogenesis of cardiolipin, a major phospholipid of the mito-
the cytoplasm, SR, and the sarcomere through excitation chondria. Subsequently, a novel mechanism was discovered
contraction coupling.59 Extensive characterization of sponta- showing that the TAZ deficiency in BTHS markedly increased
neous whole-cell [Ca2+]i transients in iPSC-CMs suggests the reactive oxygen species production, and that suppression of
presence of functional excitationcontraction coupling that reactive oxygen species reversed the BTHS cardiomyopathic
resembles the native myocardium.41 Specifically, it has been phenotype in iPSC-CMs.
demonstrated that Ca2+ influx via the depolarization-activated
L-type Ca2+ channels triggered a marked release of the SR Ca2+ Conclusions and Future Perspectives
stores via the Ca2+ sensitive ryanodine SR receptors, recapitu- The recent advent of iPSC-CM technology has enabled the
lating the Ca2+-induced Ca2+-release phenomenon in iPSC- modeling of human cardiovascular disease phenotypes, drug
CMs,41,46 a key mechanism underlying excitationcontraction screening, and the development of regenerative approaches,
coupling.59 Of note, iPSC-CM cultured on microgrooved sub- representing a technological breakthrough that could be trans-
strates displayed significantly improved Ca2+ cycling and more lated into revolutionary diagnostic and therapeutic modalities
organized SR Ca2+ release in response to caffeine, suggesting for individual patients. Patient-specific iPSC-CMs provide
that SR Ca2+ cycling properties can be influenced by culture a novel human-based experimental platform to recapitulate
conditions.40 In catecholaminergic polymorphic ventricular key features of human cardiomyocyte biology. To date, the
tachycardia, an inherited disease characterized by stress- detailed characterization of patient-specific iPSC-CMs has re-
induced ventricular arrhythmias, iPSC-CMbased modeling vealed that they share molecular, electrophysiological, meta-
also supports the presence of functional SR and ryanodine bolic, mechanical, and ultrastructural properties with primary
receptor Ca2+ transients.17 However, the Ca2+ handling kinet- human cardiomyocytes, but also exhibit diverse functional
ics in iPSC-CMs seem to be relatively slow and characterized characteristics resembling fetal rather than adult cardiomyo-
by a U-shape Ca2+ waveform, suggesting that iPSC-CMs have cytes. The structure and function of iPSC-CMs can be further
an immature Ca2+-induced Ca2+-release mechanism.60 Indeed, enhanced by prolonged culture and bioengineering approach-
iPSC-CMs exhibit a poorly developed SR and absence of es, but the factors and signaling pathways affecting maturation
T-tubules that likely affect their Ca2+ handling properties.43,44 are incompletely understood. Diverse epigenetic processes,
including long-noncoding RNA,63 microRNAs,64,65 chroma-
Metabolic Profile tin and histone proteins,66,67 and DNA methylation,68,69 have
Various studies have demonstrated that iPSC-CMs have a emerged as critical modulators of cardiac gene expression in
metabolic phenotype that resembles embryonic cardiomyo- development and disease. Hence, future studies using high-
cytes, which mostly rely on glycolysis for energy production throughput omic technologies63 will be essential to unravel
instead of lipid oxidation as seen in adult ventricular myo- the genetic and epigenetic mechanisms involved in shaping
cytes. Strategies to facilitate metabolic maturation have been the phenotype of iPSC-CMs. A better understanding of the
developed, including culturing iPSC-CMs with an adipogenic underlying mechanisms could potentially lead to the develop-
cocktail19 or glucose-free medium61 and tissue engineering ment of strategies to create cardiomyocytes with mature-like
approaches.62 These approaches were able to yield advanced phenotypes. The functional maturation is indeed a desirable
levels of metabolic phenotype maturation, as evidenced by phenotype, but it is important to acknowledge that a pheno-
significant increases of fatty acid -oxidation.19,61,62 type resembling the adult cardiomyocytes might not be attain-
By enhancing iPSC-CM metabolic maturation, iPSC-CMs able in in vitro cell culture conditions. It is also important to
have been used to recapitulate key features of mitochondrial recognize that the relatively immature phenotype of the iPSC-
disorders. In a recent study, Drawnel et al61 showed that expos- CMs is not necessarily a disadvantage for certain application.
ing normal iPSC-CMs in diabetic-like conditions (high glu- For example, immature cells could potentially be better suited
cose, endothelin-1, and cortisol) could induce an increment of for cell therapy applications, as they can become mature after
lipid accumulation, oxidative stress, and sarcomeric disarray, integration into the host myocardium, as recently observed for
phenocopying diabetic cardiomyopathy. Intriguingly, the iP- embryonic stem cellderived cardiomyocytes.70
SC-CMs derived from patients with type 2 diabetes mellitus, With the launching of the Precision on Medicine Initiative,1
a disease with complex and multifactorial pathogenesis, also the iPSC-based models of cardiovascular disease are well po-
recapitulated key pathophysiological phenotypes in vitro that sitioned to provide a powerful tool for studying genotype
corresponded to the clinical status of the original donor. A phe- phenotype association and for predicting individual patient
notypic drug screening in this model revealed molecules and responses to therapies. However, relating gene variations
86Circulation ResearchJune 19, 2015

among individuals to clinical phenotypes may not be straight- Disclosures


forward. Ultimately, we will need to evaluate and validate the J.C. Wu is a cofounder of Stem Cell Theranostics. The other authors
iPSC-CM technology using larger numbers of patient-specific report no conflicts.
cell lines. To this end, the availability of well-characterized,
disease-relevant iPSC biobanks will be indispensable to as- References
sess their predictive power.71 The recent initiatives by the 1. Collins FS, Varmus H. A new initiative on precision medicine. N Engl J
Med. 2015;372:793795. doi: 10.1056/NEJMp1500523.
National Institutes of Health and the California Institute of 2. Houser SR, Margulies KB, Murphy AM, Spinale FG, Francis GS, Prabhu
Regenerative Medicine to establish state-of-the-art human SD, Rockman HA, Kass DA, Molkentin JD, Sussman MA, Koch WJ,
iPSC biorepositories will address the increasing need for Koch W; American Heart Association Council on Basic Cardiovascular
Sciences, Council on Clinical Cardiology, and Council on Functional
quality-controlled, disease-relevant, and research-grade iPSC
Genomics and Translational Biology. Animal models of heart failure:
lines. Eventually, these efforts will provide researchers across a scientific statement from the American Heart Association. Circ Res.
academia and industry with access to high-quality iPSC lines 2012;111:131150. doi: 10.1161/RES.0b013e3182582523.
from diverse genetic backgrounds and cardiovascular diseases 3. Dixon JA, Spinale FG. Large animal models of heart failure: a critical
link in the translation of basic science to clinical practice. Circ Heart Fail.
to conduct prospective studies to establish causal associations 2009;2:262271. doi: 10.1161/CIRCHEARTFAILURE.108.814459.
between genetic variations and drug response. 4. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K,
New technologies, including next-generation sequencing72 Yamanaka S. Induction of pluripotent stem cells from adult human fi-
broblasts by defined factors. Cell. 2007;131:861872. doi: 10.1016/j.
and nuclease-mediated genome editing,73 are rapidly advanc- cell.2007.11.019.
ing the application of the iPSC-CM technology toward future 5. Burridge PW, Keller G, Gold JD, Wu JC. Production of de novo cardiomy-
precision medicine approaches. Targeted gene editing using ocytes: human pluripotent stem cell differentiation and direct reprogram-
ming. Cell Stem Cell. 2012;10:1628. doi: 10.1016/j.stem.2011.12.013.
site-specific nucleases, such as zinc finger nucleases (ZNF),
6. Matsa E, Burridge PW, Wu JC. Human stem cells for modeling heart dis-
transcription activatorlike effector nucleases (TALENs), and ease and for drug discovery. Sci Transl Med. 2014;6:239ps6. doi: 10.1126/
the clustered regularly interspaced short palindromic repeat/ scitranslmed.3008921.
clustered regularly interspaced short palindromic repeatas- 7. Mordwinkin NM, Lee AS, Wu JC. Patient-specific stem cells and car-
diovascular drug discovery. JAMA. 2013;310:20392040. doi: 10.1001/
sociated protein 9 systems (CRISPR/Cas9), is a powerful tool jama.2013.282409.
that allows for reverse genetics, genome engineering, and tar- 8. Moretti A, Bellin M, Welling A, Jung CB, Lam JT, Bott-Flgel L, Dorn
geted transgene integration experiments to be performed in a T, Goedel A, Hhnke C, Hofmann F, Seyfarth M, Sinnecker D, Schmig
A, Laugwitz KL. Patient-specific induced pluripotent stem-cell models for
precise and predictable manner. Indeed, genes have been add- long-QT syndrome. N Engl J Med. 2010;363:13971409. doi: 10.1056/
ed into specific loci and gene mutations have been introduced NEJMoa0908679.
or used to correct disease-causing mutations in iPSCs-based 9. Egashira T, Yuasa S, Suzuki T, et al. Disease characterization using LQTS-
cardiovascular disease models in vitro. The most common specific induced pluripotent stem cells. Cardiovasc Res. 2012;95:419
429. doi: 10.1093/cvr/cvs206.
application to date has been to correct mutations that cause 10. Wang Y, Liang P, Lan F, Wu H, Lisowski L, Gu M, Hu S, Kay MA, Urnov
monogenic cardiomyopathies, including DCM74 and BTHS,62 FD, Shinnawi R, Gold JD, Gepstein L, Wu JC. Genome editing of isogenic
and LQT syndrome.75 These studies have provided a proof human induced pluripotent stem cells recapitulates long QT phenotype
for drug testing. J Am Coll Cardiol. 2014;64:451459. doi: 10.1016/j.
of principle that the observed phenotypes are caused by spe- jacc.2014.04.057.
cific mutations, suggesting that this approach could be used 11. Lahti AL, Kujala VJ, Chapman H, Koivisto AP, Pekkanen-Mattila M,
to uncover the underlying pathological disease mechanism. It Kerkel E, Hyttinen J, Kontula K, Swan H, Conklin BR, Yamanaka S,
Silvennoinen O, Aalto-Setl K. Model for long QT syndrome type 2 us-
should be noted, however, that gene editing with engineered
ing human iPS cells demonstrates arrhythmogenic characteristics in cell
nucleases is problematic. Significant challenges remain, in- culture. Dis Model Mech. 2012;5:220230. doi: 10.1242/dmm.008409.
cluding specificity and off-target effects, efficiency, selection 12. Matsa E, Rajamohan D, Dick E, Young L, Mellor I, Staniforth A, Denning
of targeted sites, and delivery methods.76 C. Drug evaluation in cardiomyocytes derived from human induced plu-
ripotent stem cells carrying a long QT syndrome type 2 mutation. Eur
Overall, the iPSC-CMs present a new and rapidly devel- Heart J. 2011;32:952962. doi: 10.1093/eurheartj/ehr073.
oping technology with exciting applications, and with further 13. Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O, Winterstern A,
refinements it could pave the way for the development of per- Feldman O, Gepstein A, Arbel G, Hammerman H, Boulos M, Gepstein
L. Modelling the long QT syndrome with induced pluripotent stem cells.
sonalized medicine for cardiovascular diseases. Nature. 2011;471:225229. doi: 10.1038/nature09747.
14. Davis RP, Casini S, van den Berg CW, Hoekstra M, Remme CA, Dambrot
Acknowledgments C, Salvatori D, Oostwaard DW, Wilde AA, Bezzina CR, Verkerk AO,
Freund C, Mummery CL. Cardiomyocytes derived from pluripotent stem
We gratefully acknowledge Joseph Gold, Ian Chen, and Blake Wu
cells recapitulate electrophysiological characteristics of an overlap syn-
for critical reading, and Varachaya Khwanjaipanich for preparing the
drome of cardiac sodium channel disease. Circulation. 2012;125:3079
illustrations. Because of space limitations, we are unable to include 3091. doi: 10.1161/CIRCULATIONAHA.111.066092.
all of the important citation relevant to this subject; we apologize to 15. Yazawa M, Hsueh B, Jia X, Pasca AM, Bernstein JA, Hallmayer J,

those investigators whose work was omitted here. Dolmetsch RE. Using induced pluripotent stem cells to investigate car-
diac phenotypes in Timothy syndrome. Nature. 2011;471:230234. doi:
Sources of Funding 10.1038/nature09855.
16. Fatima A, Xu G, Shao K, et al. In vitro modeling of ryanodine receptor
This work was support from the National Institutes of Health (NIH) 2 dysfunction using human induced pluripotent stem cells. Cell Physiol
R01 HL113006, NIH R01 HL123968, NIH R24 HL117756, and Biochem. 2011;28:579592. doi: 10.1159/000335753.
California Institute of Regenerative Medicine IT1-06596 and DR2A- 17. Jung CB, Moretti A, Mederos y Schnitzler M, et al. Dantrolene rescues
05394 (J.C. Wu), NIH K99 HL104002, AHA 15BGIA22730027 and arrhythmogenic RYR2 defect in a patient-specific stem cell model of cat-
Stanford CVI Seed Grant (I. Karakikes), and Prince Mahidol Award echolaminergic polymorphic ventricular tachycardia. EMBO Mol Med.
Foundation, Thailand (V. Termglinchan). 2012;4:180191. doi: 10.1002/emmm.201100194.
Karakikes et al Current Status of iPSC-Derived Cardiomyocytes 87

18. Novak A, Barad L, Zeevi-Levin N, Shick R, Shtrichman R, Lorber A, 37. Zhang Q, Jiang J, Han P, et al. Direct differentiation of atrial and ventricu-
Itskovitz-Eldor J, Binah O. Cardiomyocytes generated from CPVTD307H lar myocytes from human embryonic stem cells by alternating retinoid
patients are arrhythmogenic in response to -adrenergic stimulation. J Cell signals. Cell Res. 2011;21:579587. doi: 10.1038/cr.2010.163.
Mol Med. 2012;16:468482. doi: 10.1111/j.1582-4934.2011.01476.x. 38. Karakikes I, Senyei GD, Hansen J, Kong CW, Azeloglu EU, Stillitano F,
19. Kim C, Wong J, Wen J, Wang S, Wang C, Spiering S, Kan NG, Forcales S, Lieu DK, Wang J, Ren L, Hulot JS, Iyengar R, Li RA, Hajjar RJ. Small
Puri PL, Leone TC, Marine JE, Calkins H, Kelly DP, Judge DP, Chen HS. molecule-mediated directed differentiation of human embryonic stem cells
Studying arrhythmogenic right ventricular dysplasia with patient-specific toward ventricular cardiomyocytes. Stem Cells Transl Med. 2014;3:1831.
iPSCs. Nature. 2013;494:105110. doi: 10.1038/nature11799. doi: 10.5966/sctm.2013-0110.
20. Asimaki A, Kapoor S, Plovie E, Karin Arndt A, Adams E, Liu Z, James 39. Liang P, Lan F, Lee AS, Gong T, Sanchez-Freire V, Wang Y, Diecke
CA, Judge DP, Calkins H, Churko J, Wu JC, MacRae CA, Klber AG, S, Sallam K, Knowles JW, Wang PJ, Nguyen PK, Bers DM, Robbins
Saffitz JE. Identification of a new modulator of the intercalated disc in RC, Wu JC. Drug screening using a library of human induced pluripo-
a zebrafish model of arrhythmogenic cardiomyopathy. Sci Transl Med. tent stem cell-derived cardiomyocytes reveals disease-specific patterns
2014;6:240ra74. doi: 10.1126/scitranslmed.3008008. of cardiotoxicity. Circulation. 2013;127:16771691. doi: 10.1161/
21. Lan F, Lee AS, Liang P, et al. Abnormal calcium handling properties un- CIRCULATIONAHA.113.001883.
derlie familial hypertrophic cardiomyopathy pathology in patient-specific 40. Rao C, Prodromakis T, Kolker L, Chaudhry UA, Trantidou T, Sridhar A,
induced pluripotent stem cells. Cell Stem Cell. 2013;12:101113. doi: Weekes C, Camelliti P, Harding SE, Darzi A, Yacoub MH, Athanasiou
10.1016/j.stem.2012.10.010. T, Terracciano CM. The effect of microgrooved culture substrates on
22. Sun N, Yazawa M, Liu J, et al. Patient-specific induced pluripotent stem calcium cycling of cardiac myocytes derived from human induced plu-
cells as a model for familial dilated cardiomyopathy. Sci Transl Med. ripotent stem cells. Biomaterials. 2013;34:23992411. doi: 10.1016/j.
2012;4:130ra47. doi: 10.1126/scitranslmed.3003552. biomaterials.2012.11.055.
23. Evans SM, Yelon D, Conlon FL, Kirby ML. Myocardial lin-
41. Itzhaki I, Rapoport S, Huber I, Mizrahi I, Zwi-Dantsis L, Arbel G, Schiller
eage development. Circ Res. 2010;107:14281444. doi: 10.1161/ J, Gepstein L. Calcium handling in human induced pluripotent stem cell
CIRCRESAHA.110.227405. derived cardiomyocytes. PLoS One. 2011;6:e18037. doi: 10.1371/journal.
24. Olson EN. Gene regulatory networks in the evolution and development of pone.0018037.
the heart. Science. 2006;313:19221927. doi: 10.1126/science.1132292. 42. Rana P, Anson B, Engle S, Will Y. Characterization of human-induced plu-
25. Noseda M, Peterkin T, Simes FC, Patient R, Schneider MD. Cardiopoietic ripotent stem cell-derived cardiomyocytes: bioenergetics and utilization
factors: extracellular signals for cardiac lineage commitment. Circ Res. in safety screening. Toxicol Sci. 2012;130:117131. doi: 10.1093/toxsci/
2011;108:129152. doi: 10.1161/CIRCRESAHA.110.223792. kfs233.
26. Kattman SJ, Witty AD, Gagliardi M, Dubois NC, Niapour M, Hotta A, 43. Gherghiceanu M, Barad L, Novak A, Reiter I, Itskovitz-Eldor J, Binah
Ellis J, Keller G. Stage-specific optimization of activin/nodal and BMP O, Popescu LM. Cardiomyocytes derived from human embryonic and in-
signaling promotes cardiac differentiation of mouse and human plu- duced pluripotent stem cells: comparative ultrastructure. J Cell Mol Med.
ripotent stem cell lines. Cell Stem Cell. 2011;8:228240. doi: 10.1016/j. 2011;15:25392551. doi: 10.1111/j.1582-4934.2011.01417.x.
stem.2010.12.008. 44. Lundy SD, Zhu WZ, Regnier M, Laflamme MA. Structural and functional
27. Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, Raval KK, maturation of cardiomyocytes derived from human pluripotent stem cells.
Zhang J, Kamp TJ, Palecek SP. Robust cardiomyocyte differentiation Stem Cells Dev. 2013;22:19912002. doi: 10.1089/scd.2012.0490.
from human pluripotent stem cells via temporal modulation of canonical 45. Kamakura T, Makiyama T, Sasaki K, Yoshida Y, Wuriyanghai Y, Chen J,
Wnt signaling. Proc Natl Acad Sci U S A. 2012;109:E1848E1857. doi: Hattori T, Ohno S, Kita T, Horie M, Yamanaka S, Kimura T. Ultrastructural
10.1073/pnas.1200250109. maturation of human-induced pluripotent stem cell-derived cardiomyo-
28. Zhang J, Klos M, Wilson GF, et al. Extracellular matrix promotes highly cytes in a long-term culture. Circ J. 2013;77:13071314.
efficient cardiac differentiation of human pluripotent stem cells: the ma- 46. Nunes SS, Miklas JW, Liu J, et al. Biowire: a platform for maturation
trix sandwich method. Circ Res. 2012;111:11251136. doi: 10.1161/ of human pluripotent stem cell-derived cardiomyocytes. Nat Methods.
CIRCRESAHA.112.273144. 2013;10:781787. doi: 10.1038/nmeth.2524.
29. Dubois NC, Craft AM, Sharma P, Elliott DA, Stanley EG, Elefanty AG, 47. Kensah G, Roa Lara A, Dahlmann J, et al. Murine and human pluripotent
Gramolini A, Keller G. SIRPA is a specific cell-surface marker for iso- stem cell-derived cardiac bodies form contractile myocardial tissue in vi-
lating cardiomyocytes derived from human pluripotent stem cells. Nat tro. Eur Heart J. 2013;34:11341146. doi: 10.1093/eurheartj/ehs349.
Biotechnol. 2011;29:10111018. doi: 10.1038/nbt.2005. 48. McCain ML, Agarwal A, Nesmith HW, Nesmith AP, Parker KK.

30. Uosaki H, Fukushima H, Takeuchi A, Matsuoka S, Nakatsuji N, Yamanaka Micromolded gelatin hydrogels for extended culture of engineered
S, Yamashita JK. Efficient and scalable purification of cardiomyocytes from cardiac tissues. Biomaterials. 2014;35:54625471. doi: 10.1016/j.
human embryonic and induced pluripotent stem cells by VCAM1 surface biomaterials.2014.03.052.
expression. PLoS One. 2011;6:e23657. doi: 10.1371/journal.pone.0023657. 49. Tulloch NL, Muskheli V, Razumova MV, Korte FS, Regnier M, Hauch
31. Hattori F, Chen H, Yamashita H, et al. Nongenetic method for purifying KD, Pabon L, Reinecke H, Murry CE. Growth of engineered human
stem cell-derived cardiomyocytes. Nat Methods. 2010;7:6166. myocardium with mechanical loading and vascular coculture. Circ Res.
32. Ban K, Wile B, Kim S, Park HJ, Byun J, Cho KW, Saafir T, Song MK, 2011;109:4759. doi: 10.1161/CIRCRESAHA.110.237206.
Yu SP, Wagner M, Bao G, Yoon YS. Purification of cardiomyocytes from 50. Hirt MN, Boeddinghaus J, Mitchell A, Schaaf S, Brnchen C, Mller C,
differentiating pluripotent stem cells using molecular beacons that target Schulz H, Hubner N, Stenzig J, Stoehr A, Neuber C, Eder A, Luther PK,
cardiomyocyte-specific mRNA. Circulation. 2013;128:18971909. doi: Hansen A, Eschenhagen T. Functional improvement and maturation of rat
10.1161/CIRCULATIONAHA.113.004228. and human engineered heart tissue by chronic electrical stimulation. J Mol
33. Tohyama S, Hattori F, Sano M, et al. Distinct metabolic flow enables Cell Cardiol. 2014;74:151161. doi: 10.1016/j.yjmcc.2014.05.009.
large-scale purification of mouse and human pluripotent stem cell-de- 51. Zhang J, Wilson GF, Soerens AG, Koonce CH, Yu J, Palecek SP, Thomson
rived cardiomyocytes. Cell Stem Cell. 2013;12:127137. doi: 10.1016/j. JA, Kamp TJ. Functional cardiomyocytes derived from human induced
stem.2012.09.013. pluripotent stem cells. Circ Res. 2009;104:e30e41. doi: 10.1161/
34. Burridge PW, Matsa E, Shukla P, Lin ZC, Churko JM, Ebert AD, Lan F, CIRCRESAHA.108.192237.
Diecke S, Huber B, Mordwinkin NM, Plews JR, Abilez OJ, Cui B, Gold 52. Zwi L, Caspi O, Arbel G, Huber I, Gepstein A, Park IH, Gepstein

JD, Wu JC. Chemically defined generation of human cardiomyocytes. Nat L. Cardiomyocyte differentiation of human induced pluripo-
Methods. 2014;11:855860. doi: 10.1038/nmeth.2999. tent stem cells. Circulation. 2009;120:15131523. doi: 10.1161/
35. Sanchez-Freire V, Lee AS, Hu S, Abilez OJ, Liang P, Lan F, Huber BC, CIRCULATIONAHA.109.868885.
Ong SG, Hong WX, Huang M, Wu JC. Effect of human donor cell source 53. Ma J, Guo L, Fiene SJ, Anson BD, Thomson JA, Kamp TJ, Kolaja KL,
on differentiation and function of cardiac induced pluripotent stem cells. J Swanson BJ, January CT. High purity human-induced pluripotent stem
Am Coll Cardiol. 2014;64:436448. doi: 10.1016/j.jacc.2014.04.056. cell-derived cardiomyocytes: electrophysiological properties of ac-
36. Devalla HD, Schwach V, Ford JW, Milnes JT, El-Haou S, Jackson C, tion potentials and ionic currents. Am J Physiol Heart Circ Physiol.
Gkatzis K, Elliott DA, Chuva de Sousa Lopes SM, Mummery CL, 2011;301:H2006H2017. doi: 10.1152/ajpheart.00694.2011.
Verkerk AO, Passier R. Atrial-like cardiomyocytes from human pluripo- 54. Du DT, Hellen N, Kane C, Terracciano CM. Action potential morphol-
tent stem cells are a robust preclinical model for assessing atrial-selec- ogy of human induced pluripotent stem cell-derived cardiomyocytes does
tive pharmacology. EMBO Mol Med. 2015;7:394410. doi: 10.15252/ not predict cardiac chamber specificity and is dependent on cell density.
emmm.201404757. Biophys J. 2015;108:14. doi: 10.1016/j.bpj.2014.11.008.
88Circulation ResearchJune 19, 2015

55. Priori SG, Napolitano C, Di Pasquale E, Condorelli G. Induced pluripotent 66. Zhang CL, McKinsey TA, Chang S, Antos CL, Hill JA, Olson EN. Class II
stem cell-derived cardiomyocytes in studies of inherited arrhythmias. J histone deacetylases act as signal-responsive repressors of cardiac hyper-
Clin Invest. 2013;123:8491. doi: 10.1172/JCI62838. trophy. Cell. 2002;110:479488.
56. Ivashchenko CY, Pipes GC, Lozinskaya IM, Lin Z, Xiaoping X, Needle S, 67. Anand P, Brown JD, Lin CY, Qi J, Zhang R, Artero PC, Alaiti MA, Bullard
Grygielko ET, Hu E, Toomey JR, Lepore JJ, Willette RN. Human-induced J, Alazem K, Margulies KB, Cappola TP, Lemieux M, Plutzky J, Bradner
pluripotent stem cell-derived cardiomyocytes exhibit temporal changes in JE, Haldar SM. BET bromodomains mediate transcriptional pause release
phenotype. Am J Physiol Heart Circ Physiol. 2013;305:H913H922. doi: in heart failure. Cell. 2013;154:569582. doi: 10.1016/j.cell.2013.07.013.
10.1152/ajpheart.00819.2012. 68. Gilsbach R, Preissl S, Grning BA, Schnick T, Burger L, Benes V, Wrch A,
57. Navarrete EG, Liang P, Lan F, et al. Screening drug-induced arrhyth- Bnisch U, Gnther S, Backofen R, Fleischmann BK, Schbeler D, Hein L.
mia [corrected] using human induced pluripotent stem cell-derived car- Dynamic DNA methylation orchestrates cardiomyocyte development, matu-
diomyocytes and low-impedance microelectrode arrays. Circulation. ration and disease. Nat Commun. 2014;5:5288. doi: 10.1038/ncomms6288.
2013;128:S313. doi: 10.1161/CIRCULATIONAHA.112.000570. 69. Sim CB, Ziemann M, Kaspi A, Harikrishnan KN, Ooi J, Khurana I, Chang
58. Sager PT, Gintant G, Turner JR, Pettit S, Stockbridge N. Rechanneling L, Hudson JE, El-Osta A, Porrello ER. Dynamic changes in the cardiac
the cardiac proarrhythmia safety paradigm: a meeting report from the methylome during postnatal development. FASEB J. 2015;29:13291343.
Cardiac Safety Research Consortium. Am Heart J. 2014;167:292300. doi: 10.1096/fj.14-264093.
doi: 10.1016/j.ahj.2013.11.004. 70. Chong JJ, Yang X, Don CW, et al. Human embryonic-stem-cell-de-

59. Bers DM. Cardiac excitation-contraction coupling. Nature. 2002;415:198 rived cardiomyocytes regenerate non-human primate hearts. Nature.
205. doi: 10.1038/415198a. 2014;510:273277. doi: 10.1038/nature13233.
60. Lee YK, Ng KM, Lai WH, Chan YC, Lau YM, Lian Q, Tse HF, Siu 71. Marx V. Stem cells: Disease models that show and tell. Nature Methods.
CW. Calcium homeostasis in human induced pluripotent stem cell- 2015;12:111114
derived cardiomyocytes. Stem Cell Rev. 2011;7:976986. doi: 10.1007/ 72. Churko JM, Mantalas GL, Snyder MP, Wu JC. Overview of high

s12015-011-9273-3. throughput sequencing technologies to elucidate molecular pathways in
61. Drawnel FM, Boccardo S, Prummer M, et al. Disease modeling and cardiovascular diseases. Circ Res. 2013;112:16131623. doi: 10.1161/
phenotypic drug screening for diabetic cardiomyopathy using human in- CIRCRESAHA.113.300939.
duced pluripotent stem cells. Cell Rep. 2014;9:810821. doi: 10.1016/j. 73. Kim H, Kim JS. A guide to genome engineering with programmable nu-
celrep.2014.09.055. cleases. Nat Rev Genet. 2014;15:321334. doi: 10.1038/nrg3686.
62. Wang G, McCain ML, Yang L, et al. Modeling the mitochondrial cardio- 74. Karakikes I, Stillitano F, Nonnenmacher M, et al. Correction of human
myopathy of Barth syndrome with induced pluripotent stem cell and heart- phospholamban R14del mutation associated with cardiomyopathy using
on-chip technologies. Nat Med. 2014;20:616623. doi: 10.1038/nm.3545. targeted nucleases and combination therapy. Nat Commun. 2015;6:6955.
63. Han P, Li W, Lin CH, et al. A long noncoding RNA protects the heart doi: 10.1038/ncomms7955.
from pathological hypertrophy. Nature. 2014;514:102106. doi: 10.1038/ 75. Bellin M, Casini S, Davis RP, DAniello C, Haas J, Ward-van Oostwaard
nature13596. D, Tertoolen LG, Jung CB, Elliott DA, Welling A, Laugwitz KL, Moretti
64. Matkovich SJ. MicroRNAs in the Stressed Heart: Sorting the Signal from A, Mummery CL. Isogenic human pluripotent stem cell pairs reveal the
the Noise. Cells. 2014;3:778801. doi: 10.3390/cells3030778. role of a KCNH2 mutation in long-QT syndrome. EMBO J. 2013;32:3161
65. van Rooij E, Marshall WS, Olson EN. Toward microRNA-based therapeu- 3175. doi: 10.1038/emboj.2013.240.
tics for heart disease: the sense in antisense. Circ Res. 2008;103:919928. 76. Cox DB, Platt RJ, Zhang F. Therapeutic genome editing: prospects and
doi: 10.1161/CIRCRESAHA.108.183426. challenges. Nat Med. 2015;21:121131. doi: 10.1038/nm.3793.

You might also like