11
REVIEWS Navigating The Labyrinth of Cardiac Regeneration Erin Lambers* and Tsutomu Kume* Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois Heart disease is the number one cause of morbidity and mortality in the world and is a major health and economic burden, costing the United States Health Care System more than $200 billion annually. A major cause of heart disease is the massive loss or dysfunction of cardiomyocytes caused by myocardial infarctions and hypertension. Due to the limited regenerative capacity of the heart, much research has focused on better understanding the process of differentiation toward cardiomyo- cytes. This review will highlight what is currently known about cardiac cell specification during mammalian development, areas of controversy, cellular sources of cardiomyocytes, and current and potential uses of stem cell derived cardiomyocytes for cardiac therapies. Developmental Dynamics 000:000–000, 2016. V C 2016 Wiley Periodicals, Inc. Key words: heart development; cardiomyogenesis; directed differentiation; direct lineage conversion; adult stem cells; reprogramming Submitted 6 November 2015; First Decision 26 January 2016; Accepted 10 February 2016; Published online 0 Month 2014 Health and Economic Burdens of Cardiovascular Disease Despite medical advances in modern medicine, heart failure caused by ischemic heart disease (IHD) is the leading cause of death worldwide, with myocardial infarction (MI) being the most common presentation (Finegold et al., 2013). In the United States, MI prevalence has reached a staggering 8.5 million cases (Lloyd- Jones et al., 2010). The Global Burdon of Disease Study estimated that 17.3 million deaths worldwide are attributed to cardiovascu- lar disease which is a 41% increase since 1990 (GBD 2013 Mor- tality and Causes of Death Collaborators, 2015). In addition to the high prevalence, patients with heart failure are faced with the poor prognosis of a 5-year mortality rate equaling nearly 50% (Stewart et al., 2001). From an economic perspective, there is an enormous financial burden associated with cardiovascular dis- ease costing the United States healthcare system approximately $204.4 billion annually (Mozaffarian et al., 2015). Currently, heart transplantation is an established therapy for heart failure, but it is inundated with major problems including limited donor hearts, chronic immunosuppressant therapy, and graft vasculop- athy (Stehlik et al., 2011). Therefore, to avoid these major compli- cations, there has been focused attention on new strategies to regenerate damaged heart tissue. An Overview of Heart Development Much of what the medical scientific community has learned about cardiac development and regeneration originates from studies of model organisms (Gould et al., 2013) such as Drosoph- ila (Seyres et al., 2012), zebrafish (Liu and Stainier, 2012), Xeno- pus (Kaltenbrun et al., 2011), chick (Martinsen, 2005), and mouse (de Boer et al., 2012). We have learned that cardiac development is governed by a complex circuitry involving signaling molecules (Fig. 1). During vertebrate embryogenesis the heart is the first organ to form (Moorman et al., 2003). During gastrulation, the process in which the three germ layers of an embryo are formed, a subset of epiblast cells moves as a sheet to the primitive streak and undergoes an epithelial-to-mesenchymal transition (EMT) (Gould et al., 2013). This sheet of epiblast cells then ingresses to transiently form the mesendoderm, a time-restricted rather than spatially defined intermediate germ layer, which later becomes the endoderm and the mesoderm (Gould et al., 2013). Studies in embryos and ESCs have shed light on this process (van Vliet et al., 2012). This process is controlled through families of sig- naling molecules that include wingless integrated (Wnt), fibroblast growth factor (FGF), and the transforming growth factor-beta (TGF- beta) superfamily. Nodal and bone morphogenetic protein-2 (BMP2) (Schlange et al., 2000), members of the TGF-beta family, work together with the Wnt/Beta-Catenin (Sumi et al., 2008) and FGF pathways (Willems and Leyns, 2008) by sending activating/inhibi- tory signals in a time and space dependent manner, patterning the embryo and allowing the successive formation of the mesendoderm, mesoderm, and cardiac mesoderm (Tada et al., 2005) (Fig. 1B). DEVELOPMENTAL DYNAMICS Grant sponsor: National Institutes of Health; Grant numbers: EY019484, HL126920; Grant sponsor: American Heart Association; Grant number: 15PRE25080006. *Correspondence to: Erin Lambers, Feinberg Cardiovascular Research Institute, Department of Medicine, Northwestern University School of Medicine, 303 E Chicago Avenue, Chicago, IL 60611. E-mail: e-lambers@ northwestern.edu and Tsutomu Kume, Ph.D., Feinberg Cardiovascular Research Institute, Department of Medicine, Northwestern University School of Medicine, 303 E Chicago Ave. Chicago IL 60611. E-mail: [email protected] Article is online at: http://onlinelibrary.wiley.com/doi/10.1002/dvdy. 24397/abstract V C 2016 Wiley Periodicals, Inc. DEVELOPMENTAL DYNAMICS 00:00–00, 2016 DOI: 10.1002/DVDY.24397 1

Navigating The Labyrinth of Cardiac Regeneration Running Short Title: Cardiac Regeneration Developmental Dynamics

Embed Size (px)

Citation preview

a

REVIEWS

Navigating The Labyrinth of Cardiac RegenerationErin Lambers* and Tsutomu Kume*

Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois

Heart disease is the number one cause of morbidity and mortality in the world and is a major health and economic burden,costing the United States Health Care System more than $200 billion annually. A major cause of heart disease is the massiveloss or dysfunction of cardiomyocytes caused by myocardial infarctions and hypertension. Due to the limited regenerativecapacity of the heart, much research has focused on better understanding the process of differentiation toward cardiomyo-cytes. This review will highlight what is currently known about cardiac cell specification during mammalian development,areas of controversy, cellular sources of cardiomyocytes, and current and potential uses of stem cell derived cardiomyocytesfor cardiac therapies. Developmental Dynamics 000:000–000, 2016. VC 2016 Wiley Periodicals, Inc.

Key words: heart development; cardiomyogenesis; directed differentiation; direct lineage conversion; adult stem cells;

reprogramming

Submitted 6 November 2015; First Decision 26 January 2016; Accepted 10 February 2016; Published online 0 Month 2014

Health and Economic Burdens ofCardiovascular Disease

Despite medical advances in modern medicine, heart failurecaused by ischemic heart disease (IHD) is the leading cause ofdeath worldwide, with myocardial infarction (MI) being the mostcommon presentation (Finegold et al., 2013). In the United States,MI prevalence has reached a staggering 8.5 million cases (Lloyd-Jones et al., 2010). The Global Burdon of Disease Study estimatedthat 17.3 million deaths worldwide are attributed to cardiovascu-lar disease which is a 41% increase since 1990 (GBD 2013 Mor-tality and Causes of Death Collaborators, 2015). In addition to thehigh prevalence, patients with heart failure are faced with thepoor prognosis of a 5-year mortality rate equaling nearly 50%(Stewart et al., 2001). From an economic perspective, there is anenormous financial burden associated with cardiovascular dis-ease costing the United States healthcare system approximately$204.4 billion annually (Mozaffarian et al., 2015). Currently,heart transplantation is an established therapy for heart failure,but it is inundated with major problems including limited donorhearts, chronic immunosuppressant therapy, and graft vasculop-athy (Stehlik et al., 2011). Therefore, to avoid these major compli-cations, there has been focused attention on new strategies toregenerate damaged heart tissue.

An Overview of Heart Development

Much of what the medical scientific community has learnedabout cardiac development and regeneration originates fromstudies of model organisms (Gould et al., 2013) such as Drosoph-ila (Seyres et al., 2012), zebrafish (Liu and Stainier, 2012), Xeno-pus (Kaltenbrun et al., 2011), chick (Martinsen, 2005), and mouse(de Boer et al., 2012). We have learned that cardiac developmentis governed by a complex circuitry involving signaling molecules(Fig. 1). During vertebrate embryogenesis the heart is the firstorgan to form (Moorman et al., 2003). During gastrulation, theprocess in which the three germ layers of an embryo are formed,a subset of epiblast cells moves as a sheet to the primitive streakand undergoes an epithelial-to-mesenchymal transition (EMT)(Gould et al., 2013). This sheet of epiblast cells then ingresses totransiently form the mesendoderm, a time-restricted rather thanspatially defined intermediate germ layer, which later becomesthe endoderm and the mesoderm (Gould et al., 2013).

Studies in embryos and ESCs have shed light on this process (vanVliet et al., 2012). This process is controlled through families of sig-naling molecules that include wingless integrated (Wnt), fibroblastgrowth factor (FGF), and the transforming growth factor-beta (TGF-beta) superfamily. Nodal and bone morphogenetic protein-2 (BMP2)(Schlange et al., 2000), members of the TGF-beta family, worktogether with the Wnt/Beta-Catenin (Sumi et al., 2008) and FGFpathways (Willems and Leyns, 2008) by sending activating/inhibi-tory signals in a time and space dependent manner, patterning theembryo and allowing the successive formation of the mesendoderm,mesoderm, and cardiac mesoderm (Tada et al., 2005) (Fig. 1B).

DE

VE

LO

PM

EN

TA

L D

YN

AM

ICS

Grant sponsor: National Institutes of Health; Grant numbers:EY019484, HL126920; Grant sponsor: American Heart Association;Grant number: 15PRE25080006.*Correspondence to: Erin Lambers, Feinberg Cardiovascular ResearchInstitute, Department of Medicine, Northwestern University School ofMedicine, 303 E Chicago Avenue, Chicago, IL 60611. E-mail: [email protected] and Tsutomu Kume, Ph.D., Feinberg CardiovascularResearch Institute, Department of Medicine, Northwestern UniversitySchool of Medicine, 303 E Chicago Ave. Chicago IL 60611. E-mail:[email protected]

Article is online at: http://onlinelibrary.wiley.com/doi/10.1002/dvdy.24397/abstractVC 2016 Wiley Periodicals, Inc.

DEVELOPMENTAL DYNAMICS 00:00–00, 2016DOI: 10.1002/DVDY.24397

1

Cardiac progenitors can be first found in mouse embryo atembryonic day (E) 7.0 at the cardiac crescent stage (Vincent andBuckingham, 2010). The cardiac mesoderm is derived from atleast two mesodermal progenitor populations: The first heart field(FHF) is located in the anterior mesoderm forming the cardiaccrescent, and the second heart field (SHF) is located medial andanterior to the cardiac crescent (Buckingham et al., 2005) (Fig. 1).Genetic tracing, explant experiments, and fluorescent dye label-ing of cells using embryos have shown that FHF and SHF progen-itors transiently express a distinct set of gene expressionpatterns, which subsequently contribute to distinct regions of theadult heart (Meilhac et al., 2015). Nkx2.5, Gata4, and Mef2c aretranscription factors transiently expressed in both FHF and SHFprogenitors (Heikinheimo et al., 1994; Dodou et al., 2004; Zhanget al., 2014). While Tbx5 and Hcn4 are transiently and predomi-nantly expressed in FHF progenitors, Tbx1 and Isl1 are transi-

ently and predominantly expressed in SHF progenitors (Sp€ateret al., 2013, 2014; Xin et al., 2013; Rana et al., 2014).

Although genetic fate mapping of Isl1 expressing cells hasshown contribution to both heart fields (Ma et al., 2008), deletionof Isl1 in mouse models produce a phenotype that corresponds todefects of the SHF lineage and is considered a marker of the SHF(Cai et al., 2003) (Fig. 1C). Cardiac looping occurs at E9.0, duringwhich time cells undergo uneven growth and remodeling. FromE10 to E15, septation formation in the atria and ventricles formthe four chambers of the heart. Meanwhile, the myocardiumundergoes a process called compaction during which time loosenetworks of trabeculae, which are thought to allow adequateoxygenation and growth, diminish and cardiomyocytes formtighter connections and cell–cell junctions (Dellefave et al.,2009). Ultimately the FHF lineage predominantly gives rise to theleft ventricle and the SHF lineage predominantly contributes to

DE

VE

LO

PM

EN

TA

L D

YN

AM

ICS

Fig. 1. Comparison of embryonic stem cell (ESC) differentiation into cardiomyocytes and early heart development. A: Time course of the embry-onic stages of cardiac development in the mouse embryo. During gastrulation the cells of the inner cell mass (ICM) form three distinct germ layers:endoderm, ectoderm, and mesoderm. The heart is derived from the mesoderm. The first cardiac progenitors are found at the cardiac crescentstage forming the first heart field (FHF, red) and the second heart field (SHF, blue). Following looping, during which time cells undergo unevenmigrations and proliferation to form the heart tube, the FHF lineage predominantly give rise to the left ventricle and the SHF lineage predominantlycontributes to the right ventricle and the outflow tract (OFT) and portions of the atria. B: An analogous time course of pluripotent stem cell (PSC)differentiation into cardiomyocyte lineages recapitulating heart development. Key signaling pathways involved at different stages of cardiac devel-opment are shown: FHF (red), SHF (blue), atrial cardiomyocytes (orange), and ventricular cardiomyocytes (green). C: Key transcription factorsinvolved at different stages of cardiac development. Transcription factors transiently and predominantly expressed in FHF progenitors (red), SHFprogenitors (blue), and both progenitor populations (purple). Markers of lineage committed atrial cardiomyocytes (orange), ventricular cardiomyo-cytes (green), and both atrial and ventricular markers (gray).

2 LAMBERS AND KUME

the right ventricle, the outflow tract (OFT), and portions of theinflow tract and atria (Fig. 1A). A recent study published inNature exploited levels of tbx5 and pitx2 to control FHF and SHFcontributions to the zebrafish heart. They found that alterationsof the FHF and SHF contributions disrupt cardiac function andspeculate that FHF and SHF progenitors may give rise to cardio-myocytes with distinctive physiological properties (Mosimannet al., 2015).

Additionally, cells that form the coronary vessels and the car-diac fibrous matrix are derived from the epicardium and arerequired for proper cardiac function (Gittenberger-de Groot et al.,1998). The genesis of epicardial derived cells (EPDCs) throughEMT and lineage determination is also regulated by a complexnetwork of transcription factors, including Wt1, Tbx18, Nfatc1,and basic helix–loop–helix (bHLH) transcription factors (Kirsch-ner et al., 2006; von Gise et al., 2011; Combs et al., 2011;Acharya et al., 2012). Specifically, Tcf21, a bHLH factor, has beenshown to be a key regulator of cell lineage development in EPDCs(Braitsch et al., 2012; Acharya et al., 2012; Tandon et al., 2013).Additionally, a recent study demonstrated that cardiac regenera-tion in zebrafish is guided by the outflow tract and hedgehog sig-naling which may have implications for mobilizing epicardialcells for cardiac repair (J Wang et al., 2015). Detailed develop-mental stages and regulation of cardiogenesis is further reviewedin Xin et al. (2013).

Neonatal Regeneration

Because the neonatal mammalian heart can substantially regen-erate after injury through cardiomyocyte proliferation until post-natal day 7 (Porrello et al., 2011), it makes an excellent model tostudy important factors involved in mammalian regeneration.Meis1, for example, was shown to be a critical regulator of thecardiomyocyte cell cycle in neonatal mice. Meis1 deletion wassufficient to increase the postnatal proliferative window of cardi-omyocytes, reactivate entry into mitosis, with no harmful effecton cardiomyocyte function (Mahmoud et al., 2013). Others usedthe regenerating neonatal mouse heart to identify a regulator ofcell cycle entry, interleukin 13, in the regenerating mouse heart(O’Meara et al., 2015).

Neonatal models have also been useful in identifying otherprocesses aside from cardiomyocyte renewal, which contribute toregeneration. Recently, Mahmoud et al. demonstrated that phar-macological cholinergic nerve inhibition impairs neonatal miceheart regeneration and could be rescued by administration ofneuregulin1 and nerve growth factor (Mahmoud et al., 2015).Using a genetic approach to label neural crest lineages, Whiteet al. similarly demonstrated that sympathetic denervation with6-OHDA inhibits neonatal heart regeneration (White et al., 2015).

Adult Cardiac Regeneration and Areas ofControversy

If and how the adult heart regenerates after injury has beenwidely debated. Ventricular dilation and wall thinning are funda-mental structural aspects of heart failure due to cardiomyopathiesand ischemic heart disease (Sutton and Sharpe, 2000). Theremodeling of the heart’s architecture effects ventricular perform-ance and characterizes myocardial disease (Sutton and Sharpe,2000). Infarction causes massive loss of cardiomyocytes, which

are replaced by scar tissue (Sutton and Sharpe, 2000). Histori-cally, remodeling and plasticity of the heart was thought to bedue principally to cellular processes of cardiomyocyte hypertro-phy and death, with no existence of cardiomyocyte renewal (Zak,1973). However, in the past decade, studies have shown evidenceof significant cardiomyocyte turnover in the heart (Mollova et al.,2013).

Seminal studies that have contributed to the paradigm shiftinclude studies using retrospective [14C] birth dating of cardiomy-ocytes (Bergmann et al., 2009) and cardiomyocyte genetic fatemapping (Hsieh et al., 2007), showing that cardiomyocyte turn-over in the heart does exist at approximately 1% per year (Berg-mann et al., 2009). Additionally, many studies have reportedcardiomyocyte progenitor populations in the heart as sources ofregeneration such as side population (Pfister et al., 2008),Sca1þ(Matsuura et al., 2004), cardiosphere derived progenitorcells (Smith et al., 2007), and c-kitþ multipotent resident stemcells (Beltrami et al., 2003) also providing evidence for adult car-diac regeneration.c-kitþ cells, in particular, have been hotlydebated as a source of regeneration in the adult heart. C-kitþprogenitor cells were originally reported to be found in nicheswithin the heart (Urbanek et al., 2006). These niches were thoughtto be important for physiological turnover of cardiac cells, governthe migration and commitment of c-kitþ (Urbanek et al., 2006;Hosoda et al., 2009;), and significantly repair infarcted adulthearts after transplantation thereby attenuating left ventricularremodeling and improving left ventricular function (Beltramiet al., 2003; Dawn et al., 2005; Bolli et al., 2006; Bearzi et al.,2007; Rota et al., 2008; Fischer et al., 2009; Tang et al., 2010).However, many independent groups through much effort couldnot replicate the findings demonstrating that c-kitþ were impor-tant for significant physiological turnover of cardiac cells in theadult heart (Murry et al., 2004; Nygren et al., 2004; Jesty et al.,2012). Van Berlo et al. (2014) and Sultana et al. (2015) publishedtwo important studies concluding that c-kitþ cells are an irrele-vant source of cardiomyocytes in the heart. The results of thestudy by Van Berlo et al. revealed that it was possible for c-kitþcells to make new myocytes during development, with aging, andafter injury, but at an extremely low level (van Berlo et al., 2014).Similarly, with five independently derived lineage tracing mousemodels, Sultana et al. found that c-kitþ cells have the capacity togenerate less than 0.05% cardiomyocytes during development,aging, or after infarct and are instead a subpopulation of endo-thelial cells (Sultana et al., 2015).

Current and Preclinical Cellular Therapiesfor Cardiac Regeneration

Adult Progenitor Stem Cells

Due to the limited regenerative capacity of the heart, stem celltherapies have emerged as strategies to promote cardiac regenera-tion. Clinical trials have tested the safety and efficacy of severaldifferent adult progenitor stem cell populations for myocardialrepair. The cell types used in clinical trials include bone marrow-derived mononuclear cells (BMMNCs), bone marrow or peripheralblood derived endothelial progenitor cells (EPCs), adipose or bonemarrow derived mesenchymal stromal cells (MSCs), and cardiacderived cardiac progenitor cells (CPCs) (Table 1).

Approximately 2,000 patients have been treated withBMMNCs, representing the majority of patients receiving cell

DE

VE

LO

PM

EN

TA

L D

YN

AM

ICS

CARDIAC REGENERATION 3

DE

VE

LO

PM

EN

TA

L D

YN

AM

ICS

TA

BL

E1.

Cell

Th

era

py

So

urces

fro

mC

lin

ica

lT

ria

lsto

Pro

of

of

Co

ncep

tR

ese

arch

Cli

nic

al

con

dit

ion

Cli

nic

al

tria

ln

am

e/

pro

ofof

con

cep

t

Fact

ors

intr

odu

ced

Res

ult

Ref

eren

ces

Acu

tem

yoc

ard

ial

infa

rcti

onB

OO

ST

BM

MN

Cs

(2.5�

10

9)

LV

EF

6%

gre

ate

rat

6m

onth

sN

osi

gn

ifica

nt

dif

fere

nce

inL

VE

Fat

18

mon

ths

(Lu

nd

eet

al.

,2006;

Mey

eret

al.

,2006)

Ch

ron

icle

ftven

tric

ula

rd

ysf

un

ctio

nT

OP

CA

RE

-CH

DB

MM

NC

s(2�

10

8)

orE

PC

s(2�

10

7)

BM

MN

Cgro

up

had

2.9

%h

igh

erL

VE

Fth

an

EP

Cor

con

trol

gro

up

at

3m

onth

s(A

ssm

us

etal.

,2007)

Acu

tem

yoc

ard

ial

infa

rcti

onA

ST

AM

IB

MM

NC

s(7�

10

7)

No

sign

ifica

nt

dif

fere

nce

inL

VE

Fat

6m

onth

s:(C

lela

nd

etal.

,2007)

Acu

tem

yoc

ard

ial

infa

rcti

onR

EP

AIR

-AM

IB

MM

NC

s(2

.4�

10

8)

Gre

ate

rL

VE

Fin

crea

sein

cell

gro

up

(5.5

%)

vs

pla

cebo

gro

up

(3%

)at

4m

onth

s(S

ch€ ach

inger

etal.

,2006)

Lef

tven

tric

ula

rd

ysf

un

ctio

nC

CT

RN

(FO

CU

S)

BM

MN

Cs

(1.5�

10

8)

2.7

%d

iffe

ren

ceL

VE

Fin

the

cell

gro

up

vs

the

pla

cebo

gro

up

at

6m

onth

s(P

erin

etal.

,2012)

Lef

tven

tric

ula

rd

ysf

un

ctio

nC

CT

RN

(TIM

E)

BM

MN

Cs

(1�

10

8)

No

sign

ifica

nt

dif

fere

nce

inL

VE

Fat

6m

onth

s(T

raver

seet

al.

,2011

,2012)

Isch

emic

card

iom

yop

ath

yP

OS

EID

ON

All

ogen

icvs

Au

tolo

gou

sB

M-M

SC

s(2�

10

7–8)

No

sign

ifica

nt

chan

ges

inL

VE

Fin

eith

ergro

up

at

13

mon

ths

(Hare

etal.

,2012)

Isch

emic

card

iom

yop

ath

yC

-CU

RE

MS

Cs

(6-1

2�

10

8)

6.8

%in

crea

sein

LV

EF

com

pare

dto

con

trol

at

6m

onth

s(B

art

un

eket

al.

,2013)

Cor

onary

art

ery

byp

ass

pati

ents

SC

IPIO

CP

Cs

(5-1

0�

10

5)

12.3

un

itin

crea

sein

LV

EF

com

pare

dto

con

trol

at

1yea

r(B

olli

etal.

,2011

)

AM

IC

AD

AC

EU

SC

PC

s(6

-12�

10

9)

No

sign

ifica

nt

dif

fere

nce

inL

VE

Fat

6m

onth

sor

1yea

rbet

wee

nce

lltr

eatm

ent

gro

up

an

dst

an

dard

care

con

trol

gro

up

(Mak

kar

etal.

,2012;

Mall

iara

set

al.

,2014)

AM

IA

PO

LL

OA

dip

ose-

MS

Cs

(2�

10

7)

4%

gre

ate

rL

VE

Fin

cell

gro

up

(56.1

%)

vs

pla

cebo

(52.2

%)

at

6m

onth

s(H

outg

raaf

etal.

,2012)

Isch

emic

card

iom

yop

ath

yP

RE

CIS

EA

dip

ose-

MS

Cs

(.4-1

.2�

10

6)

cell

s/k

g

No

sign

ifica

nt

dif

fere

nce

sat

6,

12,

18m

onth

s(P

erin

etal.

,2014)

Sic

ksi

nu

ssy

nd

rom

e/

hea

rtblo

ckP

roof

ofco

nce

pt

Tbx18

Con

ver

sion

ofw

ork

ing

myoc

yte

sin

top

ace

mak

erce

lls

(Hu

etal.

,2014)

Myoc

ard

ial

infa

rcti

onP

roof

ofco

nce

pt

Gata

4,

Mef

2C

,T

bx5

Gen

erati

onof

wor

kin

gm

yoc

yte

sfr

omca

rdia

cfi

bro

bla

sts

(Qia

net

al.

,2013)

4 LAMBERS AND KUME

therapy compared with all other cell types combined. Meta-analysis of BMMNC clinical trials suggest a very modest improve-ment in heart function due to improved infarct size and improvedremodeling (Tian et al., 2014). Implanted cells survive only brieflyafter implantation and, therefore, the modest changes in left ven-tricular ejection fraction (LVEF) and remodeling are thought to bedue to paracrine effects, rather than by contribution of cells them-selves (Alfaro and Young, 2012). The most pronounced improve-ment in LVEF was seen in the recent SCIPIO RCT using c-kitþcells (Bolli et al., 2011). However, the trial has been under investi-gation for concerns about the integrity of data (The LancetEditors, 2014). Overall, the benefits of adult progenitor stem celltherapy have not produced the functional outcomes that wereonce anticipated. Thus, new avenues of generating cardiomyo-cytes for cellular transplantation therapies are being explored.

Directed Differentiation

Human embryonic stem cells (ESCs) were first derived in 1998(Thomson et al., 1998) and since then have been acclaimed as apromising source of cellular material to be used in the treatmentof many diseases such as heart failure. In 2006, Dr. Shinya Yama-naka discovered that forced expression of four transcription fac-tors (Oct4, Sox2, Klf4, and c-Myc) in mature fibroblasts wasenough to reprogram fully committed somatic cells back into apluripotent state (Takahashi and Yamanaka, 2006). This Nobelprize winning discovery generating induced pluripotent stemcells (iPSCs) has allowed stem cell researchers to bypass ethicalcontroversies surrounding the use of human ESCs to more effec-tively study methods to generate cells such as cardiomyocytes foruse in drug discovery (Sinnecker et al., 2014), modeling cardiacdiseases (Itzhaki et al., 2011), and cardiac regeneration (LiyingZhang et al., 2015).

The first efforts to differentiate human pluripotent stem cells(PSCs) into cardiomyocytes used basic methods of embryoid body(EB) formation in which the cells would differentiate spontane-ously into different cellular lineages, yielding a low percentage ofcardiomyocytes. Further studies have revealed that many of thesignaling pathways that control cardiac development in vivo arecapable of more efficiently directing the differentiation of PSCsinto cardiomyocytes in vitro. Induction and inhibition of Wnt-B-Catenin signaling pathways and induction of Activin/Nodal/TGFb, BMP, FGF, and noncanonical Wnt signaling pathways atvarious stages of cardiac differentiation can increase cardiomyo-cyte production up to 50% (Laflamme et al., 2007; Yang et al.,2008; Paige et al., 2010; Kattman et al., 2011; Noseda et al.,2011; Lian et al., 2012) (Fig. 2B).

Purification methods yielding up to 98% purity have also beeninitiated isolating cardiomyocytes from other cell types based oncardiomyocyte specific traits. These include the expression of thesignal regulatory protein alpha (SIRPA) (Dubois et al., 2011), dif-ferences in cardiomyocyte glucose metabolism versus lactatemetabolism in noncardiomyocytes (Tohyama et al., 2013), andhigher content of mitochondria found in cardiomyocytes usingmitochondrial specific dyes (Hattori et al., 2010). A recent studyhas shown that human ESC-derived cardiomyocytes can bepropagated on clinical scale, transplanted, and integrate func-tionally into nonhuman primate recipient hearts (Chong et al.,2014). Although this demonstrates ESC-derived cardiomyocytesare getting closer to human clinical applications, safety concernsinvolving cardiac arrhythmias are still a major hurdle to over-

come. Regarding cell transplantation therapies, additional atten-tion will need to be placed on methods that can further directPSC differentiation into specific cardiomyocytes of the heart suchas ventricular cardiomyocytes, atrial cardiomyocytes, and cardiacconduction cells. These will allow tailored cell transplantationtherapies to repair specific regions of damaged heart tissue andlower risks of complications such as cardiac arrhythmias.

Direct Lineage Conversion

In 1987, a seminal discovery was made showing that ectopicexpression of a single skeletal muscle gene, MYOD1, could con-vert fibroblasts into skeletal muscle cells (Davis et al., 1987). Thiswas the first example of what we now call direct reprogrammingin which a terminally differentiated adult cell type can becomeanother adult cell type without ever passing through a pluripo-tent state. Inspired by this seminal discovery, a group recentlyintroduced a cocktail of three transcription factors known to reg-ulate normal cardiac development (Gata4, Mef2c, and Tbx5;together known as GMT) into mouse hearts with myocardialinfarction. They showed that these factors were able to reprogramcardiac fibroblasts, cells comprising scar tissue, into functionalcardiomyocytes capable of electrical coupling and showed areduction in scar tissue (Qian et al., 2013). It was demonstratedthat the introduction of Hand2 to the cocktail (GHMT) was ableincrease direct reprogramming efficiency from approximately 7%to 20% (Song et al., 2012).

To translate this direct reprogramming system from mouse tohuman, it was discovered that Myocd was additionally required(Nam et al., 2013). Recently, a group used polycistronic vectors toexpress different ratios of the GMT factors and found that, ifMef2c expression is increased compared with Gata4 and Tbx5,then reprogramming efficiency of fibroblasts into cardiomyocytesincreases three- to five-fold (L. Wang et al., 2015). This studydemonstrated that the stoichiometry of reprogramming factors iscritical for reprogramming efficiency and may explain why otherreports using the same factors demonstrate different reprogram-ming efficiencies (Muraoka and Ieda, 2015).

Epigenetic factors have also recently been found to be impor-tant for cardiac reprogramming. Four miRNAs, miR-1, miR-133,miR-208, and miR-499, were found to be sufficient to reprogramfibroblasts into cardiomyocytes in vitro and in vivo and this effi-ciency was enhanced by the inhibition of JAK1, janus kinase 1(Jayawardena et al., 2012) (Fig. 2B). Another epigenetic factor, anATP dependent chromatin remodeling factor, has also been iden-tified as an important regulator of the cardiac transcriptional pro-gram, BRG1-Associated factor 60c (BAF60c). The overexpressionBAF60c together with Gata4 and Tbx5 have been shown totrans-differentiate multipotent mesodermal cells into beating car-diomyocytes (Takeuchi and Bruneau, 2009).

Recently, there have been exciting advancements in the fieldof direct reprogramming demonstrating the ability to reprogramcardiomyocytes into specific subtypes of cardiac cells includingPurkinje cells and sinoatrial-like conduction cells (Fig. 2C). Acti-vation of Notch signaling has resulted in the conversion of cardi-omyocytes into Purkinje like cells (Rentschler et al., 2012). Forcedexpression of transcription factor Tbx3 or Tbx18 in cardiomyo-cytes was shown to induce pacemaker like cells in vitro and invivo (Bakker et al., 2012; Kapoor et al., 2013). In a follow-uplarge animal study, transcription factor Tbx18 was injected intothe hearts of pigs with complete heart block using minimally

DE

VE

LO

PM

EN

TA

L D

YN

AM

ICS

CARDIAC REGENERATION 5

invasive methods to successfully create new sinus nodes as analternative to pacemakers (Hu et al., 2014).

Short Perspectives

Clinical trials using bone marrow derived cells, mesenchymalstem cells and cardiac progenitor cells have demonstrated safety

of catheter based cell delivery, however, all with very modestimprovement in heart function. Overall, poor engraftment andlow survival have greatly limited the potential of adult progenitorcells to differentiate into cardiomyocytes to repair damaged myo-cardium. Thus, new avenues for cell transplantation are beingexplored. Insights that we have gained from the normal develop-ing embryonic heart, including key signaling molecules,

DE

VE

LO

PM

EN

TA

L D

YN

AM

ICS

Fig. 2. Cellular sources for cardiac transplantation. A: Adult progenitor stem cells that have been tested in human clinical trials. The major celltypes used for cardiac therapies include: (1) bone marrow mononuclear cells (BMMNCs, purple) and endothelial progenitor cells (EPCs, purple)derived from the bone marrow, (2) mesenchymal stem cells (MSCs, green) derived from adipose tissue, and (3) cardiac progenitor cells (CPCs,orange) derived from the heart. B: Pluripotent stem cell (PSC) derived cardiomyocytes have been generated from embryonic stem cells (ESCs)and induced-PSCs (iPSCs) (blue) induced from somatic cells (green). Differentiation efficiency into cardiac progenitors (red and blue) and cardio-myocytes (pink) depends on the introduction of signaling molecules involved during development in a finely tuned time and dose dependent man-ner. C: Generation of cardiomyocytes from direct lineage conversion bypasses the pluripotent state by reprogramming terminally differentiatedadult cells directly into another type of fully committed adult cell. This has been achieved in human fibroblasts (green) with the forced expressionof four developmental transcription factors: Gata4, Mef2c, Tbx5, and Hand2. It has also been accomplished in fibroblasts using four miRNAs mir-1, mir-133, mir-208, and mir499 along with the inhibition of Jak1 signaling. Direct lineage conversion has also been performed allowing the conver-sion of cardiomyocytes (pink) into Purkinje-like and pacemaker-like cells using Notch activation and Tbx18, respectively.

6 LAMBERS AND KUME

transcription factors, and epigenetic factors, have allowed thediscovery of innovative methods to direct the differentiation ofboth ESCs and iPSCs, pluripotent stem cells (PSCs), into cardio-myocytes and to reprogram adult cell types into cardiomyocytes.

There still exist many limitations and barriers associated withmethods of reprogrammed cardiomyocytes that need to be over-come before their use in clinical cellular transplantation therapiescan be realized. Although PSC and direct conversion derived car-diomyocytes beat spontaneously, express many cardiomyocytespecific genes, and recapitulate some electrophysiological fea-tures of adult cardiomyocytes, they still possess immature fea-tures of embryonic cardiomyocytes. These features include higherproliferation rates (Snir et al., 2003; McDevitt et al., 2005; Berg-mann et al., 2009), smaller and more rounded morphologies lack-ing t-tubules and bi-nucleation (Smolich, 1995; Lieu et al., 2009),and potassium currents considered to be responsible for arrhyth-mias (Abdul Kadir et al., 2009; Lahti et al., 2012). Although timein culture creates a more mature cardiomyocyte phenotype, it isclear more work needs to be done to drive the maturation of PSCderived cardiomyocytes (Robertson et al., 2013).

Additionally strategies used to create reprogrammed cardiomy-ocytes commonly use methods such as lentiviral delivery bywhich defined factors are randomly integrated into the genome,thereby creating genomic instability and increase risk of tumorformation. To bypass this shortcoming, new strategies going for-ward should be improved using nonintegrating methods such asepisomal delivery (Caxaria et al., 2016).

Directed differentiation and direct conversion methods of car-diomyocyte generation need to be further refined. The maincauses of death due to cardiovascular diseases include myocardialinfarction and heart failure, which primarily affect the ventriclesof the heart. Therefore, future strategies will need to bias differen-tiation and reprogramming strategies toward ventricular cardio-myocytes. Ventricular cardiomyocytes are electrically silent anddo not beat spontaneously, but only in response to electricalstimulation. Therefore, future studies will need to propagateand use ventricular cardiomyocytes to avoid transplantationcomplications such as arrhythmias caused by spontaneous beat-ing or inefficient coupling of endogenous and exogenouscardiomyocytes.

The key to understanding how to effectively repair the heartlies in elucidating the intricate pathways that construct the heartduring normal development. As we are overcoming barriers andobstacles in our path toward understanding cardiac development,the closer we are getting to the center labyrinth toward cardiacregeneration. Continued research in preclinical and clinical cellu-lar therapies is needed to fully realize our public health goals ofsignificantly decreasing the high cardiovascular mortality ratesthat plague the world.

AcknowledgmentsWe thank the members of the Kume Lab and of the Feinberg Cardi-ovascular Institute for their valuable discussion over many topicsof the review article. We also thank Dr. Margaret Buckingham ofthe Pasteur Institute for her intellectual contributions regardingcardiac lineage specification in FHF and SHF progenitor popula-tions. T.K. was funded by the NIH, and E.L. was funded by theAmerican Heart Association.

ReferencesAbdul Kadir SHS, Ali NN, Mioulane M, Brito-Martins M, Abu-

Hayyeh S, Foldes G, Moshkov AV, Williamson C, Harding SE,Gorelik J. 2009. Embryonic stem cell-derived cardiomyocytes asa model to study fetal arrhythmia related to maternal disease.J Cell Mol Med 13:3730–3741.

Acharya A, Baek ST, Huang G, Eskiocak B, Goetsch S, Sung CY,Banfi S, Sauer MF, Olsen GS, Duffield JS, Olson EN, TallquistMD. 2012. The bHLH transcription factor Tcf21 is required forlineage-specific EMT of cardiac fibroblast progenitors. Develop-ment 139:2139–2149.

Alfaro MP, Young PP. 2012. Lessons from genetically altered mes-enchymal stem cells (MSCs): candidates for improved MSC-directed myocardial repair. Cell Transplant 21:1065–1074.

Assmus B, Fischer-Rasokat U, Honold J, Seeger FH, FichtlschererS, Tonn T, Seifried E, Sch€achinger V, Dimmeler S, Zeiher AM,TOPCARE-CHD Registry. 2007. Transcoronary transplantation offunctionally competent BMCs is associated with a decrease innatriuretic peptide serum levels and improved survival of patientswith chronic postinfarction heart failure: results of the TOPCARE-CHD Registry. Circ Res 100:1234–1241.

Bakker ML, Boink GJJ, Boukens BJ, Verkerk AO, van denBoogaard M, den Haan AD, Hoogaars WMH, Buermans HP, deBakker JMT, Seppen J, Tan HL, Moorman AFM, ’t Hoen PAC,Christoffels VM. 2012. T-box transcription factor TBX3 reprog-rammes mature cardiac myocytes into pacemaker-like cells. Car-diovasc Res 94:439–449.

Bartunek J, Behfar A, Dolatabadi D, Vanderheyden M, Ostojic M,Dens J, El Nakadi B, Banovic M, Beleslin B, Vrolix M, Legrand V,Vrints C, Vanoverschelde JL, Crespo-Diaz R, Homsy C, TenderaM, Waldman S, Wijns W, Terzic A. 2013. Cardiopoietic stem celltherapy in heart failure: the C-CURE (Cardiopoietic stem Celltherapy in heart failURE) multicenter randomized trial withlineage-specified biologics. J Am Coll Cardiol 61:2329–2338.

Bearzi C, Rota M, Hosoda T, Tillmanns J, Nascirnbene A, DeAngelis A, Yasuzawa-Amano S, Trofimova I, Siggins RW,LeCapitaine N, Cascapera S, Beltrami AP, D’Alessandro DA, ZiasE, Quaini F, Urbanek K, Michler RE, Bolli R, Kajstura J, Leri A,Anversa P. 2007. Human cardiac stem cells. Proc Natl Acad SciU S A 104:14068–14073.

Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S,Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J,Nadal-Ginard B, Anversa P. 2003. Adult cardiac stem cells aremultipotent and support myocardial regeneration. Cell 114:763–776.

Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnab�e-HeiderF, Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, JovingeS, Fris�en J. 2009. Evidence for cardiomyocyte renewal inhumans. Science 324:98–102.

Bolli R, Jneid H, Tang X-L, Dawn B, Rimoldi O, Mosna F, LoredoM, Gatti A, Kajstura J, Leri A, Bearzi C, Abdel-Latif A, Anversa P.2006. Intracoronary administration of cardiac stem cellsimproves cardiac function in pigs with old infarction. Circulation114:239–239.

Bolli R, Chugh AR, D’Amario D, Loughran JH, Stoddard MF, IkramS, Beache GM, Wagner SG, Leri A, Hosoda T, Sanada F, ElmoreJB, Goichberg P, Cappetta D, Solankhi NK, Fahsah I, RokoshDG, Slaughter MS, Kajstura J, Anversa P. 2011. Cardiac stemcells in patients with ischaemic cardiomyopathy (SCIPIO): initialresults of a randomised phase 1 trial. Lancet 378:1847–1857.

Braitsch CM, Combs MD, Quaggin SE, Yutzey KE. 2012. Pod1/Tcf21 is regulated by retinoic acid signaling and inhibits differen-tiation of epicardium-derived cells into smooth muscle in thedeveloping heart. Dev Biol 368:345–357.

Buckingham M, Meilhac S, Zaffran S. 2005. Building the mamma-lian heart from two sources of myocardial cells. Nat Rev Genet6:826–835.

Cai C-L, Liang X, Shi Y, Chu P-H, Pfaff SL, Chen J, Evans S. 2003.Isl1 identifies a cardiac progenitor population that proliferatesprior to differentiation and contributes a majority of cells to theheart. Dev Cell 5:877–889.

Caxaria S, Arthold S, Nathwani AC, Goh PA. 2016. Generation ofIntegration-Free Patient Specific iPS cells using episomal

DE

VE

LO

PM

EN

TA

L D

YN

AM

ICS

CARDIAC REGENERATION 7

plasmids under feeder free conditions. Methods Mol Biol 1353:355–366.

Chong JJH, Yang X, Don CW, Minami E, Liu Y-W, Weyers JJ,Mahoney WM, Van Biber B, Cook SM, Palpant NJ, Gantz JA,Fugate JA, Muskheli V, Gough GM, Vogel KW, Astley CA,Hotchkiss CE, Baldessari A, Pabon L, Reinecke H, Gill EA,Nelson V, Kiem H-P, Laflamme MA, Murry CE. 2014. Humanembryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature 510:273–277.

Cleland JGF, Coletta AP, Abdellah AT, Nasir M, Hobson N,Freemantle N, Clark AL. 2007. Clinical trials update from theAmerican Heart Association 2006: OAT, SALT 1 and 2, MAGIC,ABCD, PABA-CHF, IMPROVE-CHF, and percutaneous mitralannuloplasty. Eur J Heart Fail 9:92–97.

Combs MD, Braitsch CM, Lange AW, James JF, Yutzey KE. 2011.NFATC1 promotes epicardium-derived cell invasion into myocar-dium. Development 138:1747–1757.

Davis RL, Weintraub H, Lassar AB. 1987. Expression of a singletransfected cDNA converts fibroblasts to myoblasts. Cell 51:987–1000.

Dawn B, Stein AB, Urbanek K, Rota M, Whang B, Rastaldo R,Torella D, Tang XL, Rezazadeh A, Kajstura J, Leri A, Hunt G,Varma J, Prabhu SD, Anversa P, Bolli R. 2005. Cardiac stemcells delivered intravascularly traverse the vessel barrier, regener-ate infarcted myocardium, and improve cardiac function. ProcNatl Acad Sci U S A 102:3766–3771.

de Boer BA, van den Berg G, de Boer PAJ, Moorman AFM, RuijterJM. 2012. Growth of the developing mouse heart: an interactivequalitative and quantitative 3D atlas. Dev Biol 368:203–213.

Dellefave LM, Pytel P, Mewborn S, Mora B, Guris DL, Fedson S,Waggoner D, Moskowitz I, McNally EM. 2009. Sarcomere muta-tions in cardiomyopathy with left ventricular hypertrabeculation.Circ Cardiovasc Genet 2:442–449.

Dodou E, Verzi MP, Anderson JP, Xu S-M, Black BL. 2004. Mef2cis a direct transcriptional target of ISL1 and GATA factors in theanterior heart field during mouse embryonic development. Devel-opment 131:3931–3942.

Dubois NC, Craft AM, Sharma P, Elliott DA, Stanley EG, ElefantyAG, Gramolini A, Keller G. 2011. SIRPA is a specific cell-surfacemarker for isolating cardiomyocytes derived from human pluripo-tent stem cells. Nat Biotechnol 29:1011–1018.

Finegold JA, Asaria P, Francis DP. 2013. Mortality from ischaemicheart disease by country, region, and age: statistics from WorldHealth Organisation and United Nations. Int J Cardiol 168:934–945.

Fischer KM, Cottage CT, Wu W, Din S, Gude NA, Avitabile D,Quijada P, Collins BL, Fransioli J, Sussman MA. 2009. Enhance-ment of myocardial regeneration through genetic engineering ofcardiac progenitor cells expressing Pim-1 kinase. Circulation120:2077–2087.

GBD 2013 Mortality and Causes of Death Collaborators. 2015.Global, regional, and national age-sex specific all-cause andcause-specific mortality for 240 causes of death, 1990–2013: asystematic analysis for the Global Burden of Disease Study2013. Lancet 385:117–171.

Gittenberger-de Groot AC, Vrancken Peeters MP, Mentink MM,Gourdie RG, Poelmann RE. 1998. Epicardium-derived cells con-tribute a novel population to the myocardial wall and the atrio-ventricular cushions. Circ Res 82:1043–1052.

Gould RA, Aboulmouna LM, Varner JD, Butcher JT. 2013. Hierarch-ical approaches for systems modeling in cardiac development.Wiley Interdiscip Rev Syst Biol Med 5:289–305.

Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL,Zambrano JP, Suncion VY, Tracy M, Ghersin E, Johnston PV,Brinker JA, Breton E, Davis-Sproul J, Schulman IH, Byrnes J,Mendizabal AM, Lowery MH, Rouy D, Altman P, Wong Po Foo C,Ruiz P, Amador A, Da Silva J, McNiece IK, Heldman AW, GeorgeR, Lardo A. 2012. Comparison of allogeneic vs autologous bonemarrow–derived mesenchymal stem cells delivered by transen-docardial injection in patients with ischemic cardiomyopathy: thePOSEIDON randomized trial. JAMA 308:2369–2379.

Hattori F, Chen H, Yamashita H, Tohyama S, Satoh Y-S, Yuasa S,Li W, Yamakawa H, Tanaka T, Onitsuka T, Shimoji K, Ohno Y,Egashira T, Kaneda R, Murata M, Hidaka K, Morisaki T, Sasaki

E, Suzuki T, Sano M, Makino S, Oikawa S, Fukuda K. 2010. Non-genetic method for purifying stem cell-derived cardiomyocytes.Nat Methods 7:61–66.

Heikinheimo M, Scandrett JM, Wilson DB, 1994. Localization oftranscription factor GATA-4 to regions of the mouse embryoinvolved in cardiac development. Dev Biol 164:361–373.

Hosoda T, D’Amario D, Cabral-da-Silva MC, Zheng H, Padin-Iruegas ME, Ogorek B, Ferreira-Martins J, Yasuzawa-Amano S,Amano K, Ide-Iwata N, Cheng W, Rota M, Urbanek K, KajsturaJ, Anversa P, Leri A. 2009. Clonality of mouse and human cardio-myogenesis in vivo. Proc Natl Acad Sci U S A 106:17169–17174.

Houtgraaf JH, den Dekker WK, van Dalen BM, Springeling T, deJong R, van Geuns RJ, Geleijnse ML, Fernandez-Aviles F, ZijlstaF, Serruys PW, Duckers HJ. 2012. First experience in humansusing adipose tissue–derived regenerative cells in the treatmentof patients with ST-segment elevation myocardial infarction.J Am Coll Cardiol 59:539–540.

Hsieh PCH, Segers VFM, Davis ME, MacGillivray C, Gannon J,Molkentin JD, Robbins J, Lee RT. 2007. Evidence from a geneticfate-mapping study that stem cells refresh adult mammalian car-diomyocytes after injury. Nat Med 13:970–974.

Hu Y-F, Dawkins JF, Cho HC, Marb�an E, Cingolani E. 2014. Biologi-cal pacemaker created by minimally invasive somatic reprogram-ming in pigs with complete heart block. Sci Transl Med 6:245ra94.

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. 2011. Modelling the long QT syndrome with inducedpluripotent stem cells. Nature 471:225–229.

Jayawardena TM, Egemnazarov B, Finch EA, Zhang L, Payne JA,Pandya K, Zhang Z, Rosenberg P, Mirotsou M, Dzau VJ. 2012.MicroRNA-mediated in vitro and in vivo direct reprogramming ofcardiac fibroblasts to cardiomyocytes. Circ Res 110:1465–1473.

Jesty SA, Steffey MA, Lee FK, Breitbach M, Hesse M, Reining S,Lee JC, Doran RM, Nikitin AY, Fleischmann BK, Kotlikoff MI.2012. c-kitþ precursors support postinfarction myogenesis in theneonatal, but not adult, heart. Proc Natl Acad Sci U S A 109:13380–13385.

Kaltenbrun E, Tandon P, Amin NM, Waldron L, Showell C, ConlonFL. 2011. Xenopus: an emerging model for studying congenitalheart disease. Birth Defect Res A Clin Mol Teratol 91:495–510.

Kapoor N, Liang W, Marb�an E, Cho HC. 2013. Direct conversion ofquiescent cardiomyocytes to pacemaker cells by expression ofTbx18. Nat Biotechnol 31:54–62.

Kattman SJ, Witty AD, Gagliardi M, Dubois NC, Niapour M, HottaA, Ellis J, Keller G. 2011. Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac differentiation ofmouse and human pluripotent stem cell lines. Cell Stem Cell 8:228–240.

Kirschner KM, Wagner N, Wagner K-D, Wellmann S, Scholz H.2006. The Wilms tumor suppressor Wt1 promotes cell adhesionthrough transcriptional activation of the alpha4integrin gene.J Biol Chem 281:31930–31939.

Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA,Dupras SK, Reinecke H, Xu C, Hassanipour M, Police S,O’Sullivan C, Collins L, Chen Y, Minami E, Gill EA, Ueno S, YuanC, Gold J, Murry CE. 2007. Cardiomyocytes derived from humanembryonic stem cells in pro-survival factors enhance function ofinfarcted rat hearts. Nat Biotechnol 25:1015–1024.

Lahti AL, Kujala VJ, Chapman H, Koivisto A-P, Pekkanen-MattilaM, Kerkel€a E, Hyttinen J, Kontula K, Swan H, Conklin BR,Yamanaka S, Silvennoinen O, Aalto-Set€al€a K. 2012. Model forlong QT syndrome type 2 using human iPS cells demonstratesarrhythmogenic characteristics in cell culture. Dis Model Mech 5:220–230.

Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, RavalKK, Zhang J, Kamp TJ, Palecek SP. 2012. Robust cardiomyo-cyte differentiation from human pluripotent stem cells via tempo-ral modulation of canonical Wnt signaling. Proc Natl Acad Sci US A 109:E1848–E1857.

Lieu DK, Liu J, Siu C-W, McNerney GP, Tse H-F, Abu-Khalil A,Huser T, Li RA. 2009. Absence of transverse tubules contributesto non-uniform Ca(2þ) wavefronts in mouse and human

DE

VE

LO

PM

EN

TA

L D

YN

AM

ICS

8 LAMBERS AND KUME

embryonic stem cell-derived cardiomyocytes. Stem Cells Dev18:1493–1500.

Liu J, Stainier DYR. 2012. Zebrafish in the study of early cardiacdevelopment. Circ Res 110:870–874.

Lloyd-Jones D, Adams RJ, Brown TM, Carnethon M, Dai S, deSimone G, Ferguson TB, Ford E, Furie K, Gillespie C, Go A,Greenlund K, Haase N, Hailpern S, Ho PM, Howard V, Kissela B,Kittner S, Lackland D, Lisabeth L, Marelli A, McDermott MM,Meigs J, Mozaffarian D, Mussolino M, Nichol G, Roger VL,Rosamond W, Sacco R, Sorlie P, Stafford R, Thom T,Wasserthiel-Smoller S, Wong ND, Wylie-Rosett J, AmericanHeart Association Statistics Committee and Stroke StatisticsSubcommittee. 2010. Executive summary: heart disease andstroke statistics--2010 update: a report from the American HeartAssociation. Circulation 121:948–954.

Lunde K, Solheim S, Aakhus S, Arnesen H, Abdelnoor M, EgelandT, Endresen K, Ilebekk A, Mangschau A, Fjeld JG, Smith HJ,Taraldsrud E, Grøgaard HK, Bjørnerheim R, Brekke M, M€uller C,Hopp E, Ragnarsson A, Brinchmann JE, Forfang K. 2006. Intra-coronary injection of mononuclear bone marrow cells in acutemyocardial infarction. N Engl J Med 355:1199–1209.

Ma Q, Zhou B, Pu WT. 2008. Reassessment of Isl1 and Nkx2-5cardiac fate maps using a Gata4-based reporter of Cre activity.Dev Biol 323:98–104.

Mahmoud AI, Kocabas F, Muralidhar SA, Kimura W, Koura AS,Thet S, Porrello ER, Sadek HA. 2013. Meis1 regulates postnatalcardiomyocyte cell cycle arrest. Nature 497:249–253.

Mahmoud AI, O’Meara CC, Gemberling M, Zhao L, Bryant DM,Zheng R, Gannon JB, Cai L, Choi W-Y, Egnaczyk GF, Burns CE,Burns CG, Macrae CA, Poss KD, Lee RT. 2015. Nerves regulatecardiomyocyte proliferation and heart regeneration. Dev Cell 34:387–399.

Makkar RR, Smith RR, Cheng K, Malliaras K, Thomson LEJ,Berman D, Czer LSC, Marb�an L, Mendizabal A, Johnston PV,Russell SD, Schuleri KH, Lardo AC, Gerstenblith G, Marb�an E.2012. Intracoronary cardiosphere-derived cells for heart regener-ation after myocardial infarction (CADUCEUS): a prospective,randomised phase 1 trial. Lancet 379:895–904.

Malliaras K, Makkar RR, Smith RR, Cheng K, Wu E, Bonow RO,Marb�an L, Mendizabal A, Cingolani E, Johnston PV, GerstenblithG, Schuleri KH, Lardo AC, Marb�an E. 2014. Intracoronarycardiosphere-derived cells after myocardial infarction. J Am CollCardiol 63:110–122.

Martinsen BJ. 2005. Reference guide to the stages of chick heartembryology. Dev Dyn 233:1217–1237.

Matsuura K, Nagai T, Nishigaki N, Oyama T, Nishi J, Wada H, SanoM, Toko H, Akazawa H, Sato T, Nakaya H, Kasanuki H, KomuroI. 2004. Adult cardiac Sca-1-positive cells differentiate into beat-ing cardiomyocytes. J Biol Chem 279:11384–11391.

McDevitt TC, Laflamme MA, Murry CE. 2005. Proliferation of cardi-omyocytes derived from human embryonic stem cells is medi-ated via the IGF/PI 3-kinase/Akt signaling pathway. J Mol CellCardiol 39:865–873.

Meilhac SM, Lescroart F, Blanpain C, Buckingham ME. 2015. Car-diac cell lineages that form the heart. Cold Spring Harb PerspectMed 5:a026344.

Meyer GP, Wollert KC, Lotz J, Steffens J, Lippolt P, Fichtner S,Hecker H, Schaefer A, Arseniev L, Hertenstein B, Ganser A,Drexler H. 2006. Intracoronary bone marrow cell transfer aftermyocardial infarction: eighteen months’ follow-up data from therandomized, controlled BOOST (BOne marrOw transfer toenhance ST-elevation infarct regeneration) trial. Circulation 113:1287–1294.

Mollova M, Bersell K, Walsh S, Savla J, Das LT, Park S-Y,Silberstein LE, Dos Remedios CG, Graham D, Colan S, K€uhn B.2013. Cardiomyocyte proliferation contributes to heart growth inyoung humans. Proc Natl Acad Sci U S A 110:1446–1451.

Moorman A, Webb S, Brown NA, Lamers W, Anderson RH. 2003.Development of the heart: (1) formation of the cardiac chambersand arterial trunks. Heart 89:806–814.

Mosimann C, Pan�akov�a D, Werdich AA, Musso G, Burger A,Lawson KL, Carr LA, Nevis KR, Sabeh MK, Zhou Y, DavidsonAJ, DiBiase A, Burns CE, Burns CG, Macrae CA, Zon LI. 2015.

Chamber identity programs drive early functional partitioning ofthe heart. Nat Commun 6:8146.

Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ,Cushman M, de Ferranti S, Despr�es J-P, Fullerton HJ, HowardVJ, Huffman MD, Judd SE, Kissela BM, Lackland DT, LichtmanJH, Lisabeth LD, Liu S, Mackey RH, Matchar DB, McGuire DK,Mohler ER, Moy CS, Muntner P, Mussolino ME, Nasir K, NeumarRW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ,Rodriguez CJ, Sorlie PD, Stein J, Towfighi A, Turan TN, ViraniSS, Willey JZ, Woo D, Yeh RW, Turner MB, American HeartAssociation Statistics Committee and Stroke Statistics Subcom-mittee. 2015. Heart disease and stroke statistics--2015 update: areport from the American Heart Association. Circulation 131:e29–e322.

Muraoka N, Ieda M. 2015. Stoichiometry of transcription factors iscritical for cardiac reprogramming. Circ Res 116:216–218.

Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO,Rubart M, Pasumarthi KBS, Virag JI, Bartelmez SH, Poppa V,Bradford G, Dowell JD, Williams DA, Field LJ. 2004. Haemato-poietic stem cells do not transdifferentiate into cardiac myocytesin myocardial infarcts. Nature 428:664–668.

Nam Y-J, Song K, Luo X, Daniel E, Lambeth K, West K, Hill JA,DiMaio JM, Baker LA, Bassel-Duby R, Olson EN. 2013. Reprog-ramming of human fibroblasts toward a cardiac fate. Proc NatlAcad Sci U S A 110:5588–5593.

Noseda M, Peterkin T, Sim~oes FC, Patient R, Schneider MD. 2011.Cardiopoietic factors: extracellular signals for cardiac lineagecommitment. Circ Res 108:129–152.

Nygren JM, Jovinge S, Breitbach M, S€aw�en P, R€oll W, Hescheler J,Taneera J, Fleischmann BK, Jacobsen SEW. 2004. Bonemarrow-derived hematopoietic cells generate cardiomyocytes ata low frequency through cell fusion, but not transdifferentiation.Nat Med 10:494–501.

O’Meara CC, Wamstad JA, Gladstone RA, Fomovsky GM, ButtyVL, Shrikumar A, Gannon JB, Boyer LA, Lee RT. 2015. Transcrip-tional reversion of cardiac myocyte fate during mammalian car-diac regeneration. Circ Res 116:804–815.

Paige SL, Osugi T, Afanasiev OK, Pabon L, Reinecke H, Murry CE.2010. Endogenous Wnt/beta-catenin signaling is required for car-diac differentiation in human embryonic stem cells. PLoS One 5:e11134.

Perin EC, Sanz-Ruiz R, S�anchez PL, Lasso J, P�erez-Cano R,Alonso-Farto JC, P�erez-David E, Fern�andez-Santos ME, SerruysPW, Duckers HJ, Kastrup J, Chamuleau S, Zheng Y, Silva GV,Willerson JT, Fern�andez-Avil�es F. 2014. Adipose-derived regener-ative cells in patients with ischemic cardiomyopathy: The PRE-CISE Trial. Am Heart J 168:88–95.e2.

Perin EC, Willerson JT, Pepine CJ, Henry TD, Ellis SG, Zhao DXM,Silva GV, Lai D, Thomas JD, Kronenberg MW, Martin AD,Anderson RD, Traverse JH, Penn MS, Anwaruddin S,Hatzopoulos AK, Gee AP, Taylor DA, Cogle CR, Smith D,Westbrook L, Chen J, Handberg E, Olson RE, Geither C,Bowman S, Francescon J, Baraniuk S, Piller LB, Simpson LM,Loghin C, Aguilar D, Richman S, Zierold C, Bettencourt J, SayreSL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M,Moy�e LA, Simari RD, Cardiovascular Cell Therapy Research Net-work (CCTRN). 2012. Effect of transendocardial delivery of auto-logous bone marrow mononuclear cells on functional capacity,left ventricular function, and perfusion in chronic heart failure: theFOCUS-CCTRN trial. JAMA 307:1717–1726.

Pfister O, Oikonomopoulos A, Sereti K-I, Sohn RL, Cullen D, FineGC, Mouquet F, Westerman K, Liao R. 2008. Role of the ATP-binding cassette transporter Abcg2 in the phenotype and func-tion of cardiac side population cells. Circ Res 103:825–835.

Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA,Olson EN, Sadek HA. 2011. Transient regenerative potential ofthe neonatal mouse heart. Science 331:1078–1080.

Qian L, Huang Y, Spencer CI, Foley A, Vedantham V, Liu L,Conway SJ, Fu J.D, Srivastava D. 2013. In vivo reprogrammingof murine cardiac fibroblasts into induced cardiomyocytes.Nature 485:593–598.

Rana MS, Th�eveniau-Ruissy M, De Bono C, Mesbah K, Francou A,Rammah M, Dom�ınguez JN, Roux M, Laforest B, Anderson RH,Mohun T, Zaffran S, Christoffels VM, Kelly RG. 2014. Tbx1

DE

VE

LO

PM

EN

TA

L D

YN

AM

ICS

CARDIAC REGENERATION 9

coordinates addition of posterior second heart field progenitorcells to the arterial and venous poles of the heart. Circ Res 115:790–799.

Rentschler S, Yen AH, Lu J, Petrenko NB, Lu MM, Manderfield LJ,Patel VV, Fishman GI, Epstein JA. 2012. Myocardial Notch sig-naling reprograms cardiomyocytes to a conduction-like pheno-type. Circulation 126:1058–1066.

Robertson C, Tran DD, George SC. 2013. Concise review: matura-tion phases of human pluripotent stem cell-derived cardiomyo-cytes. Stem Cells 31:829–837.

Rota M, Padin-Iruegas ME, Misao Y, De Angelis A, Maestroni S,Ferreira-Martins J, Fiumana E, Rastaldo R, Arcarese ML, MitchellTS, Boni A, Bolli R, Urbanek K, Hosoda T, Anversa P, Leri A,Kajstura J. 2008. Local activation or implantation of cardiac pro-genitor cells rescues scarred infarcted myocardium improvingcardiac function. Circ Res 103:107–116.

Sch€achinger V, Erbs S, Els€asser A, Haberbosch W, Hambrecht R,H€olschermann H, Yu J, Corti R, Mathey DG, Hamm CW,S€uselbeck T, Assmus B, Tonn T, Dimmeler S, Zeiher AM,REPAIR-AMI Investigators. 2006. Intracoronary bone marrow-derived progenitor cells in acute myocardial infarction. N Engl JMed 355:1210–1221.

Schlange T, Andr�ee B, Arnold HH, Brand T. 2000. BMP2 is requiredfor early heart development during a distinct time period. MechDev 91:259–270.

Seyres D, R€oder L, Perrin L. 2012. Genes and networks regulatingcardiac development and function in flies: genetic and functionalgenomic approaches. Brief Funct Genomics 11:366–374.

Sinnecker D, Laugwitz K-L, Moretti A. 2014. Induced pluripotentstem cell-derived cardiomyocytes for drug development and tox-icity testing. Pharmacol Ther 143:246–252.

Smith RR, Barile L, Cho HC, Leppo MK, Hare JM, Messina E,Giacomello A, Abraham MR, Marb�an E. 2007. Regenerativepotential of cardiosphere-derived cells expanded from percuta-neous endomyocardial biopsy specimens. Circulation 115:896–908.

Smolich JJ. 1995. Ultrastructural and functional features of thedeveloping mammalian heart: a brief overview. Reprod Fertil Dev7:451–461.

Snir M, Kehat I, Gepstein A, Coleman R, Itskovitz-Eldor J, Livne E,Gepstein L. 2003. Assessment of the ultrastructural and prolifer-ative properties of human embryonic stem cell-derived cardio-myocytes. Am J Physiol Heart Circ Physiol 285:H2355–H2363.

Song K, Nam Y-J, Luo X, Qi X, Tan W, Huang GN, Acharya A,Smith CL, Tallquist MD, Neilson EG, Hill JA, Bassel-Duby R,Olson EN. 2012. Heart repair by reprogramming non-myocyteswith cardiac transcription factors. Nature 485:599–604.

Sp€ater D, Abramczuk MK, Buac K, Zangi L, Stachel MW, Clarke J,Sahara M, Ludwig A, Chien KR. 2013. A HCN4þ cardiomyogenicprogenitor derived from the first heart field and human pluripo-tent stem cells. Nat Cell Biol 15:1098–1106.

Sp€ater D, Hansson EM, Zangi L, Chien KR. 2014. How to make acardiomyocyte. Development 141:4418–4431.

Stehlik J, Edwards LB, Kucheryavaya AY, Benden C, Christie JD,Dobbels F, Kirk R, Rahmel AO, Hertz MI. 2011. The registry ofthe international society for heart and lung transplantation:twenty-eighth adult heart transplant report--2011. J Heart LungTransplant 30:1078–1094.

Stewart S, MacIntyre K, Hole DJ, Capewell S, McMurray JJ. 2001.More “malignant” than cancer? Five-year survival following a firstadmission for heart failure. Eur J Heart Fail 3:315–322.

Sultana N, Zhang L, Yan J, Chen J, Cai W, Razzaque S, Jeong D,Sheng W, Bu L, Xu M, Huang G-Y, Hajjar RJ, Zhou B, Moon A,Cai CL. 2015. Resident c-kit(þ) cells in the heart are not cardiacstem cells. Nat Commun 6:8701.

Sumi T, Tsuneyoshi N, Nakatsuji N, Suemori H. 2008. Defining earlylineage specification of human embryonic stem cells by theorchestrated balance of canonical Wnt/beta-catenin, Activin/Nodal and BMP signaling. Development 135:2969–2979.

Sutton MG, Sharpe N. 2000. Left ventricular remodeling after myo-cardial infarction: pathophysiology and therapy. Circulation 101:2981–2988.

Tada S, Era T, Furusawa C, Sakurai H, Nishikawa S, Kinoshita M,Nakao K, Chiba T, Nishikawa S-I. 2005. Characterization of mes-

endoderm: a diverging point of the definitive endoderm andmesoderm in embryonic stem cell differentiation culture. Devel-opment 132:4363–4374.

Takahashi K, Yamanaka S. 2006. Induction of pluripotent stem cellsfrom mouse embryonic and adult fibroblast cultures by definedfactors. Cell 126:663–676.

Takeuchi JK, Bruneau BG. 2009. Directed transdifferentiation ofmouse mesoderm to heart tissue by defined factors. Nature 459:708–711.

Tandon P, Miteva YV, Kuchenbrod LM, Cristea IM, Conlon FL.2013. Tcf21 regulates the specification and maturation of proepi-cardial cells. Development 140:2409–2421.

Tang X-L, Rokosh G, Sanganalmath SK, Yuan F, Sato H, Mu J, DaiS, Li C, Chen N, Peng Y, Dawn B, Hunt G, Leri A, Kajstura J,Tiwari S, Shirk G, Anversa P, Bolli R. 2010. Intracoronary admin-istration of cardiac progenitor cells alleviates left ventricular dys-function in rats with a 30-day-old infarction. Circulation 121:293–U235.

The Lancet Editors. 2014. Expression of concern: the SCIPIO trial.Lancet 383:1279.

Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, SwiergielJJ, Marshall VS, Jones JM, 1998. Embryonic stem cell linesderived from human blastocysts. Science 282:1145–1147.

Tian T, Chen B, Xiao Y, Yang K, Zhou X. 2014. Intramyocardialautologous bone marrow cell transplantation for ischemic heartdisease: a systematic review and meta-analysis of randomizedcontrolled trials. Atherosclerosis 233:485–492.

Tohyama S, Hattori F, Sano M, Hishiki T, Nagahata Y, Matsuura T,Hashimoto H, Suzuki T, Yamashita H, Satoh Y, Egashira T, SekiT, Muraoka N, Yamakawa H, Ohgino Y, Tanaka T, Yoichi M,Yuasa S, Murata M, Suematsu M, Fukuda K. 2013. Distinct met-abolic flow enables large-scale purification of mouse and humanpluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 12:127–137.

Traverse JH, Henry TD, Ellis SG, Pepine CJ, Willerson JT, ZhaoDXM, Forder JR, Byrne BJ, Hatzopoulos AK, Penn MS, PerinEC, Baran KW, Chambers J, Lambert C, Raveendran G, SimonDI, Vaughan DE, Simpson LM, Gee AP, Taylor DA, Cogle CR,Thomas JD, Silva GV, Jorgenson BC, Olson RE, Bowman S,Francescon J, Geither C, Handberg E, Smith DX, Baraniuk S,Piller LB, Loghin C, Aguilar D, Richman S, Zierold C, BettencourtJ, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF,Kwak M, Moy�e LA, Simari RD, Cardiovascular Cell TherapyResearch Network. 2011. Effect of intracoronary delivery of auto-logous bone marrow mononuclear cells 2 to 3 weeks followingacute myocardial infarction on left ventricular function: the Late-TIME randomized trial. JAMA 306:2110–2119.

Traverse JH, Henry TD, Pepine CJ, Willerson JT, Zhao DXM, EllisSG, Forder JR, Anderson RD, Hatzopoulos AK, Penn MS, PerinEC, Chambers J, Baran KW, Raveendran G, Lambert C, LermanA, Simon DI, Vaughan DE, Lai D, Gee AP, Taylor DA, Cogle CR,Thomas JD, Olson RE, Bowman S, Francescon J, Geither C,Handberg E, Kappenman C, Westbrook L, Piller LB, SimpsonLM, Baraniuk S, Loghin C, Aguilar D, Richman S, Zierold C,Spoon DB, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI,Gordon DJ, Ebert RF, Kwak M, Moy�e LA, Simari RD, Cardiovas-cular Cell Therapy Research Network (CCTRN). 2012. Effect ofthe use and timing of bone marrow mononuclear cell delivery onleft ventricular function after acute myocardial infarction: theTIME randomized trial. JAMA 308:2380–2389.

Urbanek K, Cesselli D, Rota M, Nascimbene A, De Angelis A,Hosoda T, Bearzi C, Boni A, Bolli R, Kajstura J, Anversa P, LeriA. 2006. Stem cell niches in the adult mouse heart. Proc NatlAcad Sci U S A 103:9226–9231.

van Berlo JH, Kanisicak O, Maillet M, Vagnozzi RJ, Karch J, Lin S-CJ, Middleton RC, Marb�an E, Molkentin JD. 2014. c-kitþ cellsminimally contribute cardiomyocytes to the heart. Nature 509:337–341.

van Vliet P, Wu SM, Zaffran S, Puc�eat M. 2012. Early cardiacdevelopment: a view from stem cells to embryos. CardiovascRes 96:352–362.

Vincent SD, Buckingham ME. 2010. How to make a heart: the ori-gin and regulation of cardiac progenitor cells. Curr Top Dev Biol90:1–41.

DE

VE

LO

PM

EN

TA

L D

YN

AM

ICS

10 LAMBERS AND KUME

Von Gise A, Zhou B, Honor LB, Ma Q, Petryk A, Pu WT. 2011.WT1 regulates epicardial epithelial to mesenchymal transitionthrough b-catenin and retinoic acid signaling pathways. Dev Biol356:421–431.

Wang J, Cao J, Dickson AL, Poss KD. 2015. Epicardial regenera-tion is guided by cardiac outflow tract and Hedgehog signalling.Nature 522:226–230.

Wang L, Liu Z, Yin C, Asfour H, Chen O, Li Y, Bursac N, Liu J,Qian L. 2015. Stoichiometry of Gata4, Mef2c, and Tbx5 influen-ces the efficiency and quality of induced cardiac myocytereprogramming. Circ Res 116:237–244.

White IA, Gordon J, Balkan W, Hare JM. 2015. Sympathetic Rein-nervation Is Required for Mammalian Cardiac Regeneration. CircRes 117:990–994.

Willems E, Leyns L. 2008. Patterning of mouse embryonic stemcell-derived pan-mesoderm by Activin A/Nodal and Bmp4 signal-ing requires Fibroblast Growth Factor activity. Differentiation 76:745–759.

Xin M, Olson EN, Bassel-Duby R. 2013. Mending broken hearts:cardiac development as a basis foradult heart regeneration andrepair. Nature Publishing Group 14:529–541.

Yang L, Soonpaa MH, Adler ED, Roepke TK, Kattman SJ, KennedyM, Henckaerts E, Bonham K, Abbott GW, Linden RM, Field LJ,Keller GM. 2008. Human cardiovascular progenitor cells developfrom a KDRþ embryonic-stem-cell-derived population. Nature453:524–528.

Zak R. 1973. Cell proliferation during cardiac growth. Am J Cardiol31:211–219.

Zhang Liying, Guo J, Zhang P, Xiong Q, Wu SC, Xia L, Roy SS,Tolar J, O’Connell TD, Kyba M, Liao K, Zhang J. 2015. Derivationand high engraftment of patient-specific cardiomyocyte sheetusing induced pluripotent stem cells generated from adult car-diac fibroblast. Circ Heart Fail 8:156–166.

Zhang L, Nomura-Kitabayashi A, Sultana N, Cai W, Cai X, Moon AM,Cai C-L. 2014. Mesodermal Nkx2.5 is necessary and sufficient forearly second heart field development. Dev Biol 390:68–79.

DE

VE

LO

PM

EN

TA

L D

YN

AM

ICS

CARDIAC REGENERATION 11