Sie sind auf Seite 1von 16

C A R D I A C R E G E N E R AT I O N

REVIEWS
Therapeutic approaches for cardiac
regeneration and repair
Hisayuki Hashimoto1,2, Eric N. Olson1,2* and Rhonda Bassel-​Duby1,2
Abstract | Ischaemic heart disease is a leading cause of death worldwide. Injury to the heart is
followed by loss of the damaged cardiomyocytes, which are replaced with fibrotic scar tissue.
Depletion of cardiomyocytes results in decreased cardiac contraction, which leads to pathological
cardiac dilatation, additional cardiomyocyte loss, and mechanical dysfunction, culminating in
heart failure. This sequential reaction is defined as cardiac remodelling. Many therapies have
focused on preventing the progressive process of cardiac remodelling to heart failure. However,
after patients have developed end-​stage heart failure, intervention is limited to heart
transplantation. One of the main reasons for the dramatic injurious effect of cardiomyocyte loss is
that the adult human heart has minimal regenerative capacity. In the past 2 decades, several
strategies to repair the injured heart and improve heart function have been pursued, including
cellular and noncellular therapies. In this Review , we discuss current therapeutic approaches for
cardiac repair and regeneration, describing outcomes, limitations, and future prospects of
preclinical and clinical trials of heart regeneration. Substantial progress has been made towards
understanding the cellular and molecular mechanisms regulating heart regeneration, offering the
potential to control cardiac remodelling and redirect the adult heart to a regenerative state.

Left ventricular assist Ischaemic heart disease is the leading cause of death therapies are available for the advanced remodelled
devices worldwide, accounting for 9 million deaths per year1. heart at end-​stage heart failure. Mechanical support
(LVADs). Electromechanical Many of these patients not only undergo the acute therapies, such as left ventricular assist devices (LVADs)
devices to support circulation phase of myocardial infarction (MI) but also develop and cardiac resynchronization therapy , show benefi-
of a failing heart.
progressive heart failure derived from ventricular dys- cial outcomes in patients with end-​stage heart failure,
Cardiac resynchronization function caused by the ischaemic conditions, defined but heart transplantation remains the only current solu-
therapy as ischaemic cardiomyopathy. After MI, the damaged tion to renew the impaired heart8,9. However, pragmati­
Therapy that uses an myocardium is replaced by fibrotic scar tissue owing to cally, heart transplantation is not realistic as a standard
electromechanical device to
the minimal regenerative capacity of cardiomyocytes therapy because of the lack of donors worldwide and the
resynchronize ventricular
contraction in patients with in the adult human heart. The presence of scar tissue in surgical complexities10.
heart failure. the heart results in loss of pump function and circula- To protect the failing heart, scientists have recently
tory deficiency. Subsequently, the injured heart follows focused on approaches to promote heart regeneration.
a remodelling process that results in further fibrosis, Initial approaches, the so-​called first-​generation cell-​
1
Department of Molecular loss of myocardium, cardiac dysfunction, and dilatation, based therapies, involved transplanting noncardiac cells
Biology, Hamon Center for eventually leading to fatal heart failure2. because researchers could not obtain adequate num-
Regenerative Science and Treatment of ischaemic heart disease has focused on bers of functional cardiomyocytes to replace the lost
Medicine, University of Texas
Southwestern Medical Center,
protecting the heart from progression to heart failure3. myocardium11. Initial cell candidates included skeletal
Dallas, TX, USA. For example, revascularization by thrombolysis, cardiac myoblasts, which were expected to contribute to cardiac
2
Senator Paul D. Wellstone intervention, and bypass surgery serve to improve blood contraction, and bone marrow-​derived cells and mesen-
Muscular Dystrophy supply and can salvage the injured ischaemic myocardium. chymal stem cells, which showed cardiogenic potential
Cooperative Research Center, Pharmacological approaches that slow or reverse cardiac in vitro12–14. The next generation of cell-​based therapy
University of Texas remodelling, such as angiotensin-​converting enzyme used resident cardiac cells with stem cell-​like character-
Southwestern Medical Center,
Dallas, TX, USA.
(ACE) inhibitors, angiotensin receptor–neprilysin istics. These cardiac-​derived cells were expandable and
inhibitors, β-​blockers, and mineralocorticoid-​receptor demonstrated multipotency, differentiating into various
*e-​mail: eric.olson@
utsouthwestern.edu antagonists, have decreased heart failure mortality4–7. In cell types of the heart in vitro15. Another approach to
https://doi.org/10.1038/ contrast to these cardioprotective therapies that target heart regeneration involved the generation of functional
s41569-018-0036-6 the remodelling process in the failing heart, limited cardiomyocytes in vitro that then were transplanted

Nature Reviews | Cardiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
R e v ie w s

Key points
week after birth when the mouse heart loses its regen-
erative capacity. Unsurprisingly, this regenerative time
• Preclinical outcomes of cardiac regenerative therapy approaches have not translated window correlates with the period after birth when
effectively to clinical trials. mammalian cardiomyocytes withdraw from the cell
• Transplantation of induced pluripotent stem cell-​derived cardiomyocytes for cardiac cycle27. Genetic lineage tracing showed that the major-
repair has encountered problems related to safety and low engraftment rates. ity of cardiomyocytes in the recovered area of hearts
• Cell-​free-based approaches for heart repair and regeneration involve from mice aged 1 day were derived from pre-​existing
cardioprotective secretory factors or direct reprogramming of resident cardiac cardiomyocytes22. These findings offer an opportu-
fibroblasts to cardiomyocyte-​like cells. nity to study the molecular mechanisms whereby the
• Endogenous cardiomyocyte proliferation can be evoked by modulating cell cycle mammalian heart transitions from a regenerative to a
regulators, the Hippo signalling pathway, and the cardiac microenvironment. non-regenerative organ.
• Genome editing can correct underlying mutations causing heart disease in animals Shifting the focus to humans, a case report of a
and offers a state-​of-the-​art therapeutic approach for cardiac repair. newborn baby demonstrating functional cardiac recov-
• The therapeutic potential of cardiac regeneration approaches can be improved by ery after MI suggests a degree of regenerative capacity in
optimizing the delivery method of the therapeutic factors. the human neonatal heart28. Furthermore, accumulating
evidence shows that cardiomyocyte turnover occurs in
the adult mammalian heart, including in humans29.
into the injured heart. Preclinical studies used pluri- However, the turnover of cardiomyocytes is obviously
potent stem cells, which can reliably differentiate into insufficient to restore the contractile function of an
functional cardiomyocytes in vitro16. injured human heart, which can lose up to 1 billion
Alternative cell-​free approaches for heart regener­ cardiomyocytes after an MI30. Therefore, establishing
ation have targeted cardiac resident cells. For example, cardiac regenerative therapies is an important step
a reprogramming approach focused on converting car- towards repairing the damaged heart in patients with
diac fibroblasts to a cardiomyocyte fate17,18. Inducing pro- heart disease.
liferation of the remaining endogenous cardiomyocytes
is another approach to repair the heart19. Additionally, Cell-​based therapies for cardiac repair
the fibrotic response after myocardial injury has Noncardiomyocytes. Noncardiac cells, which include
also been targeted to block cardiac remodelling 20. skeletal myoblasts, bone marrow-​derived cells, and
Deciphering and harnessing the molecular mechanisms mesenchymal stem cells (MSCs), have been the pri-
regulating the transient regenerative capacity of the neo- mary source for cell-​based therapies in heart failure.
natal mammalian heart might also provide insights into The first cells used were skeletal myoblasts, which were
regenerating the adult mammalian heart21,22. expected to remuscularize the injured heart and restore
Treating patients with ischaemic heart disease is the contractile function. In animal models, transplanted
the ultimate goal of therapeutic approaches for car- skeletal myoblasts survived and differentiated into a
diac regeneration11. Despite the enthusiasm and effort myogenic lineage, and the treatment improved the
invested in many clinical trials of heart repair and ejection fraction in both ischaemic and nonischaemic
regeneration, to date, no effective approaches are avail- cardiomyopathies31–34. Initial clinical trials showed pos-
able to regenerate the damaged human heart. With an itive effects, with transplanted skeletal myoblasts leading
eye on future clinical trials, we focus this Review on to improved heart function in patients with ischaemic
the advances in regenerative therapies that have clinical cardiomyopathy12,35. Unfortunately, long-​term follow-​
potential for the treatment of ischaemic heart disease. up studies did not show beneficial effects. Moreover,
The objective of this Review is to present a compre- adverse effects such as arrhythmogenesis occurred
hensive overview of therapeutic approaches for cardiac owing to the inability of skeletal myoblasts to integrate
regeneration and repair. However, we do not include electromechanically with surrounding cardiomyocytes36.
information on therapeutic approaches targeting the These poor outcomes ruled out the use of skeletal
inflammatory process and the epicardium for heart myoblasts in further clinical studies.
repair, as these topics are reviewed elsewhere in this The next cells used for cardiac regeneration therapy
Focus Issue23,24. were unselected bone marrow-​derived mononuclear
cells, which became widely tested in clinical studies for
Regenerative capacity of the heart heart disease37. This approach was supported by stud-
Cardiomyocytes are considered to be in a terminally ies demonstrating a beneficial contribution of bone
differentiated state. Nevertheless, cardiomyocyte cell marrow-​derived mononuclear cells to cardiac repair in
cycle activity and proliferative capacity differ in spe- animal models after acute MI38,39. Early clinical trials,
cies and life stages. Cardiomyocytes from certain such as the BOOST trial13 and the REPAIR-​AMI trial40,
animals, such as axolotls, frogs, newts, and zebrafish, showed that transplantation of bone marrow-​derived
retain lifelong capacity to proliferate25,26. By contrast, mononuclear cells conferred some beneficial effects
cardiomyocytes from adult mammals are perma- in patients with acute MI by improving the ejection
nently quiescent. However, upon injury, the heart of a fraction. Unfortunately, multiple clinical trials that
mouse aged 1 day can regenerate over a period of included larger patient populations and well-​randomized
Lineage tracing
A method to identify all
3 weeks, whereas the injured heart of a mouse aged and double-​blinded settings did not reproduce these
progeny originating from a ≥7 days cannot repopulate lost cardiomyocytes22 (Fig. 1). earlier results41–43. Much controversy remains, and no
single cell. These studies define a transitional period in the first definite conclusion has been reached on the potential

www.nature.com/nrcardio
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
C A R D I A C R E G E N ERREAvTI EI O
wNs

a Neonatal Coronary Myocardial Functional reports in animal studies suggest that MSCs have
mouse ligation infarction heart minimal to no cardiomyogenic potential51,52.
Regeneration Therefore, noncardiomyocyte-​based cell therapies
have not shown consistent positive results in the treat-
Ischaemic Suture Scar
area ment of heart disease. Nevertheless, these clinical trials
Ischaemic have effectively established a standard of operation
b Adult Coronary Myocardial heart failure in evaluating the safety and efficacy of cell-​b ased
mouse ligation infarction
regeneration therapies in heart failure.
Remodelling
Cardiac- ​ d erived cells. Studies cast doubt on the
cardiomyogenic potential of bone marrow-​d erived
mono­nuclear cells and MSCs; therefore, interest shifted
• Stem cell-derived cells • Exosomes
• Direct reprogramming • Growth factors to harvesting cardiac stem cells (CSCs) for repairing
• Small molecules • Cytokines the failing heart. CSCs are defined as resident heart
• Gradual hypoxia • microRNAs cells that show clonogenic, self-​renewal, and multi­
potent capacity. CSCs can differentiate into at least
c Human Coronary Myocardial ? Ischaemic three major cardiac cell types: cardiomyocytes, smooth
occlusion infarction heart failure muscle cells, and endothelial cells53. Therefore, CSCs
were expected to be an effective and potent cell source
Remodelling for replacing lost tissue in the damaged heart. Different
populations of CSCs have been defined by cell surface
markers such as mast/stem cell growth factor receptor
• ACE inhibitors
• ARNI KIT, stem cell antigen 1 (SCA1; also known as LY6A),
Thrombus • β-Blockers and insulin gene enhancer protein ISL1 (also known
• MR antagonists
• Thrombolysis as islet 1)15,54,55. Cardiospheres, a mixed population
• PCI • LVAD of CSCs (mainly KIT+ CSCs) that are isolated from
• CABG surgery • CRT
cell cultures of mouse and human heart explants, are
Fig. 1 | response and therapeutic approaches to myocardial injury. a | Response to another cardiac-​derived cell source for heart repair53.
myocardial injury differs between developmental stages in mice. Neonatal mice Indeed, preclinical trials showed some beneficial effects
(aged <1 week) are capable of regenerating the heart, with functional recovery after injury. of cardiac-​derived cell transplantation in animal mod-
This regenerative capacity is lost postnatally after the first week. b,c | In adult mice and els of MI56,57 (Table 1). However, clinical trials were
humans, the default response to myocardial injury is fibrosis, where the infarct necrotic initiated with limited understanding of how these cells
tissue is replaced with a fibrotic scar, causing loss of cardiac contractility. The damaged contribute to heart regeneration58.
adult heart enters a negative loop of cardiac remodelling that progresses to heart failure. Two clinical trials have been carried out using KIT+
In humans, the current goal of clinical therapies is either to salvage the ischaemic
CSCs and cardiosphere-​derived cells. The first was the
myocardium by early revascularization (light grey box) or to prevent cardiac remodelling
with drug therapy and electromechanical support (dark grey box). Accumulating evidence
SCIPIO trial59,60, in which KIT+ CSCs were used to treat
in preclinical studies demonstrates promising outcomes with therapeutic approaches patients with ischaemic cardiomyopathy. This study
aimed at heart regeneration (green box), although these new approaches have clinical showed a slight positive effect in patients treated with
translational problems. The dashed line indicates potential clinical therapeutic CSC therapies: left ventricular ejection fraction (LVEF)
approaches. ACE, angiotensin-​converting enzyme; ARNI, angiotensin receptor–neprilysin increased and infarct size decreased. However, because
inhibitor ; CRT, cardiac resynchronization therapy ; LVAD, left ventricular assist device; CSCs had shown low engraftment rates in preclinical
MR , mineralocorticoid receptor ; PCI, percutaneous coronary intervention. studies, other researchers questioned the capacity of
CSCs to form functional cardiomyocytes. Additionally,
conflicting findings on the cardiomyogenic potential
efficacy of cardiac regenerative therapies that are based of endogenous KIT+ CSCs were reported61,62. A study
on bone marrow-​derived mononuclear cells. based on lentiviral expression of Cre recombinase
MSCs are a subpopulation of stromal cells that can under the control of the Kit promoter, which has a
be isolated from various tissues, such as bone marrow pattern of expression restricted to KIT+ cells, showed
or adipose tissue, and in vitro studies have shown the that KIT+ CSCs were necessary and sufficient for heart
capacity of MSCs for self-​renewal and multipotent dif- regeneration after injury61. By contrast, another study
ferentiation into adipocytes, chondrocytes, hepatocytes, using a mouse model in which the tamoxifen-​inducible
osteoblasts, neurons, and skeletal muscle cells44. MSCs MerCreMer protein was targeted to the Kit locus, fol-
MerCreMer can also differentiate into cardiomyocytes in the presence lowed by cross-​breeding with the R26-GFP reporter
A fusion protein containing Cre of the DNA methyltransferase inhibitor 5-azacytidine or mouse line, showed a minimal contribution of KIT+
recombinase flanked at both
by co-​culture with primary cardiomyocytes45–47. Several cells to heart regeneration after injury62. In addition,
ends with a mutated murine
oestrogen receptor (Mer) preclinical studies showed that transplanting MSCs into a concern was expressed about the integrity of certain
ligand binding domain. the injured heart after MI led to improved cardiac func- data in the SCIPIO trial63. Despite the uncertainties of
MerCreMer generates an tion, although the mechanism was not understood48,49. the benefit of CSC-​based regeneration therapy, a sec-
inducible Cre recombinase However, in clinical trials such as the POSEIDON14 and ond clinical trial, the CADUCEUS trial64, was initiated,
activation system that can gain
access to the nuclear
MSC-​HF50 trials, transplantation of MSCs led to only in which a cardiosphere-​derived cell population was
compartment only with modest benefits for patients with ischaemic cardiomyo­ transplanted into patients after MI. The results from
exposure to tamoxifen. pathy. Moreover, during the past decade, inconsistent this clinical trial showed a reduction in infarct size

Nature Reviews | Cardiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
R e v ie w s

Table 1 | Cardiac regenerative therapies in preclinical studies


Therapeutic animal Cohort (n) disease delivery method Cardiac function Comments ref.
source model and model recovery versus
age control; assessment
method
Cardiac stem cells
Autologous • Pig • C = 11 • IHF • Intramyocardial • LVEF improvement LacZ+ CDCs detected in 56

CDCs (1 × 107) • Adult • T = 10 • MI • 4 weeks after MI (~7%) at 8 weeks the peri-​infarct zone with
• Ventriculography the use of cardiac marker
expression
Autologous • Pig • C = 10 • IHF • Intracoronary • LVEF improvement EGFP-​labelled CSC-​ 57

KIT+ CSCs • 8–10 weeks • T = 11 • IR 90 min • 3–4 months (8–10%) at 8 weeks derived cells detected
(5 × 105) after IR • Echocardiography in the infarct zone with
the use of cardiac marker
expression
Pluripotent stem cells
Human ESC-​ • Macaque • C = 2 • IHF • Intramyocardial • No significant • Proof-​of-principle 69

CMs (~1 × 109) • 5–14 years • T = 4 • IR 90 min • 14 days after MI change in LVEF study
• Echocardiography • GFP-​labelled human
ESC-​CMs detected in
the peri-​infarct zone
• Electromechanical
integration with host
myocardium
• All animals receiving
cell transplants had
ventricular arrhythmias
Allogeneic • Macaque • C = 5 • IHF • Intramyocardial • LVEF improvement • GFP-​labelled iPSC-​CMs 74

iPSC-​CMs • 4–5 years • T = 5 • MI • 14 days after MI (~10%) at 12 weeks detected in the peri-​
(4 × 108) • CT imaging infarct zone
• Electromechanical
integration with host
myocardium
• All animals receiving
cell transplants had
ventricular arrhythmias
Secretory factors
• NRG1-MPs • Pig • C = 6 • IHF • Intramyocardial • LVFS improvement • Increase in 91

• FGF1-MPs • 12–24 • T (NRG1-MPs) = 5 • IR 120 min • 1 week after MI (~9%) at 3 months vascularization
months • T (FGF1-MPs) = 6 • Echocardiography with NRG1-MP and
FGF1-MP treatment
• Reduced fibrosis with
NRG1-MP treatment
• miR-199a-3p • Mouse • C = 13 • IHF • Intramyocardial • LVEF improvement Increased numbers in 97

• miR-590-3p • 2 months • T (miR-199a-3p) = 20 • MI • After coronary (10–20%) at 8 weeks the peri-​infarct zone of
• T (miR-590-3p) = 13 ligation (with • Echocardiography CMs positive for the DNA
cationic lipid synthesis marker EdU
formulations)
Human • Pig • C = 6 • IHF • Intramyocardial • LVEF improvement • Reduced scar size 109

CDC-​derived • Adult • T = 6 • MI • 4 weeks after MI (~5%) at 1 month • Increased numbers in
exosomes • MRI the peri-​infarct zone
(~16.5 × 1011) of CMs positive for the
cell cycle active phase
marker Ki67
Direct cardiac reprogramming
Retroviral GMT • Mouse • C = 9 • IHF • Intramyocardial • LVEF improvement • Reduced scar size 135

• 2 months • T = 9 • MI • After coronary (~10%) at 12 weeks • Fibroblast lineage-​
ligation • MRI traced cells (Fsp1-Cre or
Postn-​Cre) expressing
cardiac markers in the
peri-​infarct zone
Retroviral • Mouse • C = 9 • IHF • Intramyocardial • LVEF improvement • Reduced scar size 18

GHMT • 8–10 weeks • T = 10 • MI • After coronary (~25%) at 12 weeks • Fibroblast lineage-​
ligation • MRI traced cells (Tcf21-Cre)
expressing cardiac
markers in the
peri-infarct zone

www.nature.com/nrcardio
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
C A R D I A C R E G E N ERREAvTI EI O
wNs

Table 1 (cont.) | Cardiac regenerative therapies in preclinical studies


Therapeutic animal Cohort (n) disease delivery method Cardiac function Comments ref.
source model and model recovery versus
age control; assessment
method
Stimulation of endogenous cardiac repair
Agrin • Mouse • C = 7 • IHF • Intramyocardial • LVEF improvement • Reduced scar size 166

• 12 weeks • T = 8 • MI • After coronary (~10%) at 5 weeks • Increased numbers in
ligation • Echocardiography the peri-​infarct zone
of CMs positive for the
proliferation markers
Ki67 , BrdU, and AURKB
Systemic • Mouse • C = 9 • IHF • Gradual hypoxia • LVEF improvement • Reduced scar size 168

exposure to • 2 months • T = 9 • MI induction (~20%) at ~6 weeks • Increased numbers
hypoxia reaching 7% O2 • Echocardiography of BrdU+ CMs, PHH3+
level for 2 weeks CMs, and AURKB+ CMs,
• 1 week after MI mainly in the MI remote
zone
AURKB, aurora kinase B; BrdU, 5-bromodeoxyuridine; C, control; CDC, cardiosphere-​derived cell; CM, cardiomyocyte; CSC, cardiac stem cell; EdU, 5-ethynyl-2′-
deoxyuridine; EGFP, enhanced green fluorescent protein; ESC, embryonic stem cell; FGF1, fibroblast growth factor 1; GFP, green fluorescent protein; GMT, Gata4,
Mef2c, and Tbx5; GHMT, Gata4, Hand2, Mef2c, and Tbx5; IHF, ischaemic heart failure; iPSC, induced pluripotent stem cell; IR , ischaemia–reperfusion; KIT, mast/stem
cell growth factor receptor KIT; LVEF, left ventricular ejection fraction; LVFS, left ventricular fractional shortening; MI, myocardial infarction; MP, microparticle;
NRG1, neuregulin 1; PHH3, phosphohistone H3; T, treatment.

and an increase in viable myocardium, as shown by ethical opposition to using human embryonic cells
cardiac MRI at 6 months and 12 months after therapy. even for therapeutic use. Therefore, only one clinical
However, in the CADUCEUS trial, patients receiving the trial using ESC-​derived cardiomyocytes is ongoing70.
cell therapy did not show any improvement in left ven- Fortuitously, Yamanaka and colleagues solved the
tricular global function, indicating that, under current ethical issues of using ESCs when they reported that
conditions, transplanting cardiosphere-​derived cells is mouse and human fibroblasts could be reprogrammed
not an effective therapy to repair the heart64. Given the to an ESC-​like pluripotent state, called induced pluri-
limited clinical data reported to date, additional studies potent stem cells (iPSCs), by forced expression of four
are needed to determine the efficacy of CSCs in clinical genes encoding transcription factors: OCT3/OCT4 (also
regenerative therapy. known as POU5F1), SOX2, KLF4, and MYC (referred
to as OSKM factors)71,72. Not only did Yamanaka win
Pluripotent stem cells. A major deficiency of the early the 2012 Nobel Prize in Physiology or Medicine for
clinical trials was that the transplanted stem cells had this discovery, but iPSCs also offered a new cell-​based
a limited capacity to differentiate into cardiomyocytes. approach for heart repair, enabling autologous or alloge-
Therefore, scientists were challenged to generate neic transplantation and avoiding the ethical concerns
functional cardiomyocytes efficiently in vitro to ena- associated with ESCs. However, because autologous
ble transplantation of de novo cardiomyocytes to the transplantation is considered unrealistic owing to the
injured heart. The first cell source studied was embry- vast amount of work and high cost associated with this
onic stem cells (ESCs) derived from the inner cell approach, preclinical studies are predominantly per-
mass at the blastocyst stage of early embryos. ESCs are formed by transplantation of allogeneic or xenogeneic
clonogenic, self-​renewing, and pluripotent, thereby cells with administration of immunosuppressants to
having the capacity to differentiate into all cell types avoid rejection of the transplanted cells. Although an
of the three germ layers (endoderm, ectoderm, and early study showed that transplantation of human iPSC-​
mesoderm) 65. Because ESCs are easily expandable derived cardiomyocytes attenuated cardiac remodelling
and can differentiate into cardiomyocytes in vitro, and improved LVEF in an immunosuppressed porcine
these cells offer the opportunity to obtain sufficient MI model, most of the transplanted human iPSC-​
amounts of cardiomyocytes for transplantation. derived cardiomyocytes did not show long-​t erm
Transplantation of ESC-​derived cardiomyocytes into survival in the injured heart73. By contrast, in a 2016
the injured heart in animal models of MI improved study, transplanted allogeneic iPSC-​derived cardiomyo­
cardiac function despite low engraftment rates 66,67. cytes survived up to 12 weeks after treatment in the
Remarkably, transplanted ESC-​derived cardiomyocytes infarcted heart of a macaque, with evidence of improved
electromechanically coupled with resident cardiomyo­ cardiac function74 (Table 1). A notable finding of this
cytes in animal models, which was not the case with study is that the iPSC-​derived cardiomyocytes showed
skeletal myoblasts. Nevertheless, complications asso- long-​term survival in the immunosuppressed macaque
ciated with the ESC-​derived cardiomyocyte therapy, heart without any tumour formation. However, as
such as arrhythmias, were detected in some species68,69 predicted, the animals with cell transplants had a sub-
(Table 1) . Moreover, using ESCs poses major issues stantial incidence of ventricular tachycardia, which
for clinical application, such as risk of tumorigenesis could be due to the immature state of the transplanted
and immune rejection. In addition, there is also broad iPSC-​derived cardiomyocytes.

Nature Reviews | Cardiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
R e v ie w s

)TQYVJHCEVQTU OKETQ40#U 'ZQUQOGU


r8')( r04) rOK4CR rOK4s rOK4 r%&%U
r()( r()( rOK4R rOK4 rOK4C r%2%U
rOK4s

+UEJCGOKE
%QTQPCT[ #PIKQIGPGUKU %CTFKQO[QE[VGRTQNKHGTCVKQP JGCTVHCKNWTG
QEENWUKQP

4GOQFGNNKPI

415
+UEJCGOKE #RQRVQUKU 1ZKFCVKXGUVTGUU
CTGC 5ECT
(KDTQUKU

r()( r%&%U
r04) ()( OK4 OK4C r%2%U /5%U
)TQYVJHCEVQTU OKETQ40#U 'ZQUQOGU

Fig. 2 | Contributions of secretory factors to cardiac repair and regeneration. A scheme of cardioprotective
effects (green boxes) or cardiac remodelling effects (red boxes) by representative secretory factors (growth factors,
microRNAs, and exosomes) is shown. Various secretory factors promote angiogenesis or cardiomyocyte proliferation,
thereby promoting cardiac repair. Other secretory factors elicit cardioprotective effects by attenuating cardiac
remodelling through inhibition of fibrosis, cardiomyocyte apoptosis, and oxidative stress. The dashed arrow indicates
incompletely understood mechanisms. CDC, cardiosphere-​derived cell; CPC, cardiac progenitor cell; FGF, fibroblast
growth factor ; MSC, mesenchymal stem cell; NRG1, neuregulin 1; ROS, reactive oxygen species; VEGF, vascular
endothelial growth factor.

Pluripotent stem cell-​b ased approaches prob- (miRNAs), and exosomes secreted by the stem cells
ably share similar problems with other cell-​b ased were responsible and potentially crucial for cardiac repair
approaches: inconsistencies in the reported engraft- and regeneration81.
ment rate and the risk of arrhythmia. Additionally,
risk of tumorigenesis remains an underlying con- Growth factors to promote cardiac repair. Growth
cern of pluripotent stem cell-​based therapies and has factors are signalling molecules that contribute to
been detected in immunodeficient mouse models75,76. multiple cellular processes. The growth factor neu-
However, improvements have been made in generating regulin 1 (NRG1) and its receptors, receptor tyrosine-​
mature iPSC-​derived cardiomyocytes with high purity protein kinase ERBB2 and ERBB4, have a critical role
and sufficient numbers, and these improvements are in trabeculation and endocardial cushion formation
expected to enhance the cell retention rate by increas- during heart development82. Activation of the NRG1–
ing the number of transplanted cells and to reduce the ERBB2/ERBB4 signalling pathway in the injured adult
risk of tumorigenesis and arrhythmia associated with mouse heart induces cardiomyocyte proliferation and
undifferentiated immature cells77,78. In addition, pro- improves cardiac function83,84 (Fig. 2). In a clinical trial
viding scaffolds for iPSC-​derived cardiomyocytes, such in patients with heart failure, systemic delivery of
as hydrogels or cell sheets, has been shown to improve human recombinant NRG1 prevented cardiac remod-
therapeutic outcomes79,80. Regardless of the advance- elling, as shown by decreases in end-​diastolic volume
ment in transplantation methods, before translating and end-​systolic volume compared with placebo85.
these therapies to clinical trials, studies in large-​animal However, the proliferative effect of NRG1 in cardio-
models are warranted to assess the efficacy and safety of myocytes in vivo has been challenged on the basis of
iPSC-​derived cardiomyocyte-​based therapy. findings in a mouse model of MI where NRG1 treat-
ment did not increase cardiomyocyte DNA synthesis86.
Secretory factors for cardiac repair Therefore, additional studies are required to explain
In cell-​based therapy, transplanted cells were expected to and confirm this beneficial outcome.
Hydrogels
Colloid gels composed of a restore cardiac function by engrafting and differentiating Other growth factors that showed beneficial effects
network of hydrophilic polymer into functional cardiomyocytes in vivo. Although this in preclinical studies did not realize this promise in
chains. cell-​based approach did provide modest cardioprotec- clinical trials. For instance, administration of vascu-
tive benefits, paradoxically, the transplanted stem cells lar endothelial growth factor A (VEGFA) — a well-​
Paracrine effects
The effects on a cell that are
rarely differentiated into cardiomyocytes. To rational- known pro-​angiogenic factor — improved regional
induced by secreted factors ize the beneficial effects observed, researchers reasoned coronary flow and restored cardiac function in an ani-
from another cell. that paracrine effects from growth factors, microRNAs mal model of MI87, but this benefit was not observed

www.nature.com/nrcardio
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
C A R D I A C R E G E N ERREAvTI EI O
wNs

in clinical trials88. Cardiac-​specific overexpression in been reported to contribute to cardiac repair in vivo in
mice of the gene encoding another pro-​angiogenic animal studies98–101. Therefore, the positive outcomes
factor, fibroblast growth factor 2 (FGF2), decreased in preclinical studies demonstrate the therapeutic
infarct size and improved cardiac function after MI potential of miRNAs.
compared with wild-​type mice89. Furthermore, FGF2 However, lessons from clinical trials of regenera-
depletion exacerbated cardiac dysfunction after ischae- tive therapies based on growth factors caution us to
mia–reperfusion injury89. Despite these encouraging consider a major challenge for the clinical translation
early results, treatment with FGF2 provided only mini- of miRNA-​b ased approaches: how can miRNAs be
mal cardioprotective effects in patients with ischaemic efficiently delivered to the target site? Similar to the
heart disease90. studies on growth factors, providing a scaffold seems to
One possible explanation for these inconsistent be beneficial for continuous local delivery of miRNAs.
findings is the inadequate amount of therapeutic factor As an example, miR-302 is reported to promote cardio­
available at the target sites. This low availability could myocyte proliferation by inhibiting Hippo signalling
be due to inefficient delivery of the therapeutic growth components99. Use of a hydrogel scaffold to deliver
factors and/or to a short half-​life of the growth factors. To miR-302 allowed the sustained, gradual release of the
enhance exposure of the injured heart to the therapeutic miRNA in mouse hearts, and a single injection of
growth factors, scaffolds of biomaterials that enable sus- the hydrogel–miR-302 complex into the mouse heart
tained release of the factors to the target sites have been after MI led to continuous cardiomyocyte proliferation
used and have shown improved cardioprotective effects. and improved cardiac function102.
For instance, in a pig MI model, loading NRG1 and
fibroblast growth factor 1 (FGF1) into microparticles Exosomes in cardiac repair. Exosomes provide a
provided sustained local release of the therapeutic fac- plausible therapeutic approach to cardiac repair.
tors, which improved left ventricular function associated Exosomes are small extracellular vesicles (30–100 nm
with increased angiogenesis and reduced ventricular diameter) that are produced by cells and are charac-
remodelling91 (Table 1). Additionally, improved gene-​ terized by the presence of specific surface markers
based therapeutics have also led to increased efficiency such as CD9, CD63, and CD81 (ref.103). Exosomes are
of delivery and expression of therapeutic factors, such released from cells by fusion of intracellular multive-
as synthetic modified RNA, a method that has been used sicular bodies with the plasma membrane103. Exosomes
to express human VEGFA in the mouse heart after MI92. are secreted by many cell types, including stem cells,
Treatment with this modified RNA led to an increase and contain various cargos such as RNAs, lipids,
in VEGFA expression, induced vascular regeneration, or proteins. Exosomes can function as a vehicle for
and improved cardiac function and long-​term survival intercellular communication by carrying cell-​specific
compared with the use of DNA vectors92. mRNAs and miRNAs, and accumulating evidence
supports a role for exosomes in cell–cell communica-
microRNAs in cardiac repair. miRNAs have been tion among cardiac cells104,105. For instance, after MI in
implicated in paracrine signalling. miRNAs are highly mice, administration of murine cardiosphere-​derived
conserved, single-​strand, small non-​c oding RNAs exosomes enriched in miR-451 inhibited cardiomyo-
that regulate gene expression post-​transcriptionally by cyte apoptosis106 (Fig. 2). Administration of extracellular
annealing with complementary sequences of mRNAs93. vesicles, predominantly comprising exosomes, derived
Individual miRNAs interfere with multiple target from human CSCs also attenuated cardiac remodelling
mRNAs, thereby controlling a variety of biological and improved cardiac function in preclinical studies of
processes, including heart development and disease MI107–109 (Table 1). Delivery of MSC-​derived exosomes
(reviewed elsewhere94,95). For example, miR-15 family into the mouse heart reduced oxidative stress and
members have been reported to control postnatal mitotic promoted survival of cardiomyocytes after ischaemia–
arrest in the mouse heart96. Cardiac over­expression of reperfusion injury, thereby reducing infarct size and
miR-195, a miR-15 family member, causes premature improving cardiac function110.
cardiomyocyte cell cycle arrest96. Moreover, inhibition In addition to these cardioprotective properties,
of miR-15 family members with synthetic modified the capacity of exosomes to transport therapeutic fac-
RNAs induced cardiomyocyte proliferation in vivo and tors supports the prospect of exosomes as a biological
improved cardiac function in the ischaemic adult mouse vehicle for clinical application. Interestingly, fusing
heart96 (Fig. 2). the neuron-​specific rabies viral glycoprotein peptide
Other studies have identified miRNAs that acti- (RVG) with an exosomal membrane protein enabled
vate DNA replication in cardiomyocytes. An unbiased targeted exosomal delivery of small interfering RNAs
screen using a library of 875 human miRNAs detec­ to knock down the expression of specific genes in the
ted multiple miRNAs that promote neonatal rat brain 111. A cardiac-​t argeted exosome would be an
cardiomyocyte proliferation97. Among these miRNAs, attractive delivery vehicle for cardioprotective factors
cardiac overexpression of miR-199a or miR-590 such as miRNAs or ligands because the use of exosomes
in vivo induced proliferation of postnatal and adult could diminish the risk of activating the immune
Synthetic modified RNA cardiomyocytes and improved cardiac function and response associated with viral delivery systems 112.
Chemically synthesized RNA
with changes to the chemical
decreased fibrosis in the infarcted heart of adult mice Understanding the molecular mechanisms of exoso-
composition that alter function (Table 1). Other miRNAs such as the miR-17-92 cluster, mal biology will advance the therapeutic potential of
or stability of the RNA. miR-214, miR-302-367, and the miR-222 cluster have exosomes in cardiac regenerative therapy.

Nature Reviews | Cardiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
R e v ie w s

Direct reprogramming for heart repair approach to that for establishing iPSCs has been taken,
Direct reprogramming of mouse fibroblasts into where multiple transcription factors related to cardiac
cardiomyocytes in vitro. Two major issues arising development are combined and overexpressed in fibro-
from the use of iPSCs for heart repair and regeneration blasts. Forced expression of a combination of three
are the oncogenic potential of the remaining undifferen- genes encoding cardiac transcription factors — Gata4,
tiated cells and the low engraftment rates of transplanted Mef2c, and Tbx5 (a cocktail referred to as GMT) — or
cells. An approach that could bypass these challenges of GMT factors plus Hand2 (referred to as GHMT)
is to convert resident cardiac cells directly into de novo successfully reprogrammed murine fibroblasts into
cardiomyocytes. Fibroblasts constitute a large cell popu­ induced cardiomyocyte-​like cells in vitro, and these cells
lation of the heart, and considering that cardiac injury expressed major cardiac genes and had cardiomyocyte
such as MI is followed by a fibrotic response, transdif- characteristics (such as sarcomere structure, sponta-
ferentiating cardiac fibroblasts to cardiomyocytes is an neous intracellular calcium oscillations, and beating
attractive possibility for repairing the heart and dimin- contractions) without going through a cardiac precur-
ishing cardiac fibrosis113. To achieve this goal, a similar sor state17,18 (Fig. 3). Remarkably, these reprogramming

C+PXKVTQFKTGEVTGRTQITCOOKPI r,#-
/6)0$ r906 %CTFKCE
RTQIGPKVQTU
(KDTQDNCUVU

K%/U
r)/6*#0& rOK4 r$/+ r6)(β r,#-
r#-6 rOK4 r26$ r41%- r906
r<0( rOK4 r016%*
rOK4
+PȱCOOCVKQP

%CTFKQO[QE[VGU
D+PXKVTQFKTGEVTGRTQITCOOKPI r/'52
r'65 %CTFKCE
RTQIGPKVQTU
K%/U
(KDTQDNCUVU

r)/6*#0& r'544γ rOK4 % r6)(β


r/'52 r<(2/ rOK4 r906
r/;1%&

rOK4
E+PXKXQFKTGEVTGRTQITCOOKPI rOK4
rOK4 r6)(β
%QTQPCT[ r)/6*#0& rOK4 r906 *GCTVTGRCKT
NKICVKQP 5WVWTG

+UEJCGOKECTGC

Fig. 3 | direct reprogramming of fibroblasts into cardiomyocytes. a | Forced expression of cardiac transcription factors
or myogenic microRNAs directly reprogrammes mouse fibroblasts to induced cardiomyocyte-​like cells (iCMs) or cardiac
progenitors. As iCMs are functionally and structurally different from endogenous cardiomyocytes, studies have aimed to
improve the efficiency and quality of reprogrammed iCMs by adding factors (green box) or blocking the transdifferentia-
tion barriers (red box) in vitro. b | In humans, the cardiac reprogramming factors are different from the factors used in
mouse cells in vitro. However, inhibition of transforming growth factor-​β (TGFβ) and WNT signalling pathways enhances
reprogramming in both mouse and human cells. c | The reprogramming cocktails determined in vitro can reprogramme
resident cardiac fibroblasts in mice in vivo. The dashed arrows indicate differentiation potential. 9C, CHIR99021, A83-01,
BIX01294, AS8351, SC1, Y27632, OAC2, SU16F, and JNJ10198409; AKT1, RAC-​α serine/threonine-​protein kinase; BMI1,
Polycomb complex protein BMI1; ESRRγ, oestrogen-​related receptor-​γ; GMT, transcription factor GATA4, myocyte-​specific
enhancer factor 2C (MEF2C), and T-​box transcription factor TBX5; HAND2, heart and neural crest derivatives-​expressed
protein 2; JAK , Janus kinase; MESP1, mesoderm posterior protein 1; MTGNB, MESP1, TBX5, GATA4, homeobox protein
NKX2-5, and BRG1-associated factor 60C (BAF60C; also known as SMARCD3); MYOCD, myocardin; PTB, polypyrimidine
tract-​binding protein 1; ROCK , RHO-​associated protein kinase 1.

www.nature.com/nrcardio
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
C A R D I A C R E G E N ERREAvTI EI O
wNs

cocktails are sufficient to suppress fibrotic signalling and zinc-​finger protein ZFPM2, or even miR-1 and/or
and convert fibroblasts towards a cardiac fate. However, miR-133, to successfully induce the conversion of
several hurdles need to be addressed before the clinical human fibroblasts towards a cardiac fate128–130 (Fig. 3).
application of this approach, including inadequate Human reprogrammed cardiomyocyte-​like cells showed
reprogramming efficiency, lack of understanding of a similar gene expression profile to cardiomyocytes and
the molecular mechanisms, and the heterogeneous exhibited sarcomere structure and spontaneous calcium
population of induced cardiomyocyte-​like cells114,115. transients128–130. Moreover, transdifferentiation of human
Many approaches have been implemented to fibroblasts to cardiomyocyte-​like cells has also been
improve cardiac reprogramming efficiency, includ- achieved with a virus-​free method consisting of a pluri-
ing the addition of miRNAs and growth factors to potent cocktail with a combination of nine chemicals,
the reprogramming cocktails and the modification of with up to 97% of the reprogrammed cells spontaneously
endogenous signalling pathways such as RAC-​α serine/ beating131. These chemically treated human fibroblasts
threonine-​protein kinase (AKT1), transforming growth were transplanted into an immunodeficient mouse
factor-​β (TGFβ), WNT, and Notch signalling116–122. Using heart after MI, where the transplanted cells were even-
an unbiased screen of human transcription factors, the tually reprogrammed to cardiomyocyte-​like cells that
zinc-​finger protein ZNF281 was identified as an inducer expressed cardiac genes and had a well-​organized sar-
of cardiac reprogramming123. Addition of ZNF281 to the comere structure. However, there was no evidence that
reprogramming cocktail suppressed the expression of the chemically reprogrammed cardiomyocyte-​like cells
genes associated with the inflammatory response and were directly converted from fibroblasts without passing
modulated cardiac gene expression by interacting with through a progenitor state.
the transcription factor GATA4 (ref.123). In other studies, Direct cardiac reprogramming in human cells is
suppressing the expression of genes encoding certain fac- more challenging than in mouse cells, given the low
tors such as the Polycomb complex protein BMI1 and the reprogramming efficiency and the longer time needed
splicing factor polypyrimidine tract-​binding protein 1 for human cells to exhibit cardiomyocyte characteristics.
(PTB) enhanced cardiac reprogramming, suggest- This increased difficulty is consistent with other cellular
ing that these factors act as barriers to cardiac repro­ reprogramming strategies132. For example, human iPSC
gramming124,125. However, reaching a consensus protocol reprogramming efficiency is less than that observed
for in vitro direct reprogramming of fibroblasts into for mouse iPSCs and requires a longer time to repro-
cardiomyocytes is difficult because in these studies gramme, even though the reprogramming cocktail
the cardiomyocyte markers, source of fibroblasts, and the contains the same factors for both cell types. Similarly,
time of analysis differ. Therefore, a precise comparison neuronal cell reprogramming with human cells requires
of the reprogramming efficiency between the different additional factors and longer duration than neuronal cell
protocols should be performed. reprogramming with mouse cells133. The difficulty in
Another strategy to reprogramme fibroblasts into reprogramming human cells could be attributed to the
cardiomyocytes has been the partial reprogramming of difference in the epigenetic landscape between mouse
the cells into cardiac progenitor cells, bypassing a pluri- and human fibroblasts, suggesting additional epigenetic
potent state. Forced expression of a combination of five barriers for reprogramming human cells134. Another
genes encoding early cardiac factors — Mesp1, Gata4, feature to consider is that humans have a longer devel-
Tbx5, Nkx2-5, and Baf60c (also known as Smarcd3) — opmental time than mice, potentially contributing to the
reprogrammed murine fibroblasts into an expandable species differences in cell reprogramming. Interestingly,
multipotent cardiac progenitor cell population126. These despite the requirement of different factors for direct
induced cardiac progenitor-​like cells were transplanted cardiac reprogramming between species, crucial endo­
into murine hearts after MI, where the cells differenti- genous signalling pathways such as the TGFβ1 and WNT
ated into cardiomyocytes, endothelial cells, and smooth signalling pathways contribute similarly to cardiac
muscle cells, and improved survival. However, the draw- reprogramming in both mouse and human cells120.
backs of transplanting cells diminish the advantages of
direct reprogramming. Direct reprogramming of fibroblasts into cardiomyo-
cytes in vivo. Direct reprogramming in vivo has been
Direct reprogramming of human cells in vitro. To trans- reported in mice with the use of GMT and GHMT
late reprogramming approaches to the clinic, efforts reprogramming cocktails with retroviral delivery,
have focused on performing direct cardiac reprogram- which exclusively infects proliferating cells such as acti-
ming in human cells. Previous studies reported that vated cardiac fibroblasts after myocardial injury18,135,136
expression of a combination of the transcription factors (Fig.  3; Table  1) . Lineage tracing of fibroblasts with
protein c-​ETS2 (ETS2) and mesoderm posterior pro- the use of Fsp1 (also known as S100a4)–Cre and
tein 1 (MESP1) converted human dermal fibroblasts Myh6–MerCreMer mice demonstrated that these
into cardiac progenitors that expressed early cardiac newly generated cardiomyocyte-​like cells were derived
markers such as ISL1 and homeobox protein NKX2-5, from fibroblasts and not from cell fusion with endo­
which are not detected with direct reprogramming of genous cardiomyocytes. Interestingly, these in vivo-​
mouse fibroblasts127. Interestingly, for direct reprogram- generated cardiomyocyte-​like cells demonstrated similar
ming of human cells, GMT or GHMT reprogramming characteristics to endogenous ventricular cardiomyo­
cocktails require additional factors such as myocar- cytes, such as their cardiac gene expression profile,
din, MESP1, oestrogen-​related receptor-​γ (ESRRγ), sarcomere structure, contractility, calcium transients,

Nature Reviews | Cardiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
R e v ie w s

and even pre-​existing cardiomyocytes. This possibility


(KDTQDNCUVU raises concerns because direct cardiac reprogram-
#PIKQVGPUKP
ming has been honed using fibroblasts; therefore, in
4GNCZKP other cell lineages, the reprogramming cocktail might
r'PFQVJGNKCNEGNNU
%J[OCUG 6)(β r'RKECTFKWO induce incomplete reprogramming that could elicit
+UEJCGOKE
r$QPGOCTTQYEGNNU unfavourable effects such as arrhythmias.
! r2GTKXCUEWNCTEGNNU The positive outcomes observed in murine direct
JGCTVHCKNWTG /[QȰDTQDNCUVU
cardiac reprogramming in vivo give optimism to even-
%CTFKQO[QE[VG
%CTFKQO[QE[VGRTQNKHGTCVKQP tual clinical translation. However, several challenges
remain to be addressed before clinical application,
such as the low reprogramming efficiency, the imma-
turity and heterogeneity of induced cardiomyocyte-​like
(KDTQUKU
cells, and the lack of a feasible delivery method for the
reprogramming cocktail. In addition, in current studies,
4##5 &GFKHHGTGPVKCVKQP %GNNE[ENG 1ZKFCVKXG the induction of reprogramming is limited to a short
TGIWNCVQTU UVTGUU
906 period of time after coronary ligation. This short win-
dow of administration would restrict the therapeutic
time window in a clinical setting. A 2018 study reported
#ITKP /'+5 *KRRQ *[RQZKC /GEJCPKECN that direct cardiac reprogramming could be achieved
UKIPCNNKPI WPNQCFKPI
in both mouse and human cells by delivering repro-
Fig. 4 | approaches to stimulate endogenous regenerative capacity for heart repair. gramming factors with a Sendai virus vector, which is a
Approaches targeting endogenous cardiac regeneration (green boxes) involve activating non-​segmented, negative-​stranded RNA virus138. Sendai
the proliferation of endogenous cardiomyocytes and targeting the cardiac fibrotic virus replicates only in the cytoplasm and does not
response. Cardiomyocyte proliferation can be induced by overexpressing cell integrate into the genome, suggesting a more feasible
cycle-related genes or by inhibiting cell cycle suppressors such as the transcription factor strategy for clinical application compared with delivery
homeobox protein MEIS1 or the Hippo signalling pathway (red boxes). Alternatively , using retrovirus or lentivirus139.
approaches to mimic the neonatal cardiac environment by exposure to hypoxia or
mechanical unloading or by providing the extracellular matrix protein agrin also evoke
Stimulating endogenous cardiac repair
cardiomyocyte proliferation. Transforming growth factor-​β1 (TGFβ1) and angiotensin
have a pivotal role in inducing cardiac fibroblast differentiation into myofibroblasts Proliferation of endogenous cardiomyocytes. To
promote endogenous cardiomyocyte proliferation,
during cardiac injury , thereby inducing cardiac fibrosis. Additionally , other cell lineages
are proposed to transdifferentiate into myofibroblasts. The renin–angiotensin– initial approaches targeted universal cell cycle regulators
aldosterone system (RAAS) and the WNT signalling pathway also contribute to cardiac such as cyclins, cyclin-​dependent kinases (CDKs), and
fibrosis. These fibrotic processes (blue boxes) can be targeted to attenuate cardiac tumour suppressor genes. Although modulating these
remodelling. The dashed arrows indicate incompletely understood mechanisms. factors individually in adult cardiomyocytes did enable
some proliferative activity140,141, a study published in
and action potential; therefore, in  vivo-​generated 2018 reported that overexpression of a combination of
cardiomyocyte-​like cells seem to mimic endoge- cell cycle regulators increases the effect on cardiomyo-
nous cardiomyocytes better than in vitro-​generated cyte proliferation and improves cardiac function after
cardiomyocyte-​like cells. Actually, this finding is MI142 (Fig. 4). These findings suggest that specific factors
not surprising considering that cardiomyocyte-​like involved in postnatal cardiomyocyte cell cycle regulation
cells generated in vivo are exposed to a physiological are also candidates to stimulate the proliferative capacity
microenvironment, including electromechanical stim- in adult cardiomyocytes. For example, the transcription
ulation and interactions with adjacent cells. In vivo-​ factor homeobox protein MEIS1 is reported as a regu­
generated cardiomyocyte-​like cells also form gap lator of postnatal cardiomyocyte cell cycle arrest143.
junctions with endogenous cardiomyocytes, increasing Cardiac-​specific overexpression of Meis1 in neonatal
the potential for electromechanical coupling18. Similar mice reduced cardiomyocyte proliferation and abol-
results were reported with the use of a combination of ished heart regeneration. Conversely, genetic ablation
muscle-​specific miRNAs delivered with lentivirus to the of Meis1 in neonatal mice extended the time window of
murine heart122. postnatal cardiomyocyte proliferation and induced cardio­
Although direct reprogramming in vivo improves myocytes to re-​enter the cell cycle in adult mice143. Gene
cardiac function after MI in mice, the relatively low expression analysis revealed that MEIS1 is required for
reprogramming efficiency compared with direct repro- transcriptional activation of the synergistic CDK inhib-
gramming in vitro suggests that other factors besides itors p15 (also known as CDK inhibitor 2B; encoded by
contractility of induced cardiomyocyte-​like cells Cdkn2b), p16 (also known as CDK inhibitor 2A; encoded
contribute to the therapeutic efficacy of in vivo repro- by Cdkn2a), and p21(also known as CDK inhibitor 1A;
gramming137. For example, a decrease in the size of encoded by Cdkn1a), which probably contribute to
the fibrotic tissue suggests that paracrine effects from postnatal cardiomyocyte cell cycle withdrawal.
induced cardiomyocyte-​like cells or a direct contribu- The Hippo signalling pathway is an evolutionarily
tion of the reprogramming factors suppress the fibrotic conserved pathway shown to control cell proliferation
response. In addition, we cannot exclude the possibility and organ size144. This pathway is activated through
that the reprogramming cocktail affects other cells in phosphorylation of a series of factors, such as mammal­
the heart, such as endothelial cells, immune cells, CSCs, ian STE20-like protein kinase 1 (MST1; also known as

www.nature.com/nrcardio
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
C A R D I A C R E G E N ERREAvTI EI O
wNs

STK4) and MST2 (also known as STK3), protein salva- and differentiation into myofibroblasts, which also gener-
dor homologue 1 (SAV1), and large tumour suppressor ate TGFβ1 (Fig. 4). TGFβ1 triggers myofibroblasts to pro-
homologue 1 (LATS1) and LATS2, which together phos- duce matrix proteins, eventually forming fibrous tissue.
phorylate and inactivate the transcriptional co-​activator Accumulating evidence from clinical trials shows that
Yes-​associated protein 1 (YAP1). Phosphorylation of pharmacologically inhibiting the activity of angiotensin II
YAP1 blocks nuclear translocation and results in YAP1 attenuates cardiac remodelling158. However, blocking
being retained in the cytoplasm. In mice, similar pheno­ fibrosis by inhibiting TGFβ is not straightforward, as
types of heart enlargement owing to cardiomyocyte preclinical studies have shown that TGFβ treatment
proliferation can be evoked by genetic ablation of Mst1, outcomes depend on the timing of intervention after the
Mst2, Lats2, or Sav1, or by activating YAP1 in cardio- MI159. In mice, systemic inhibition of TGFβ in the first
myocytes19,145. Encouragingly, inhibiting the Hippo 24 h after MI increased mortality and impaired cardiac
pathway in the heart after MI promotes cardiomyocyte function, whereas TGFβ inhibition at a later phase atten-
proliferation and improves cardiac function in mice19,146 uated cardiac hypertrophy and remodelling. These results
(Fig. 4). Accordingly, cell cycle-​related genes were upreg- support the concept that the initial fibrotic response after
ulated in the heart after Hippo signalling inhibition, but myocardial injury is a necessary phase for healing.
stress response genes, such as those associated with an Additionally, nonfibroblast cells such as endothe-
antioxidant response, and mitochondrial quality control lial cells, epicardial cells, bone marrow-​derived cells,
genes were also upregulated147,148. Because inhibiting the and perivascular cells have been proposed to generate
Hippo pathway is associated with the risk of off-​target newly activated fibroblasts after MI160–163. For exam-
cell proliferation, antioxidant reagents have been tested ple, endothelial cells acquire a fibroblastic phenotype
and administered into the injured adult mouse heart as through endothelial-​to-mesenchymal transition after
an alternative approach. Interestingly, administration myocardial injury160. Delivery of recombinant human
of antioxidants induced cardiomyocyte proliferation bone morphogenetic protein 7 or pigment epithelium-​
and promoted heart repair, suggesting that target- derived factor attenuates myocardial fibrosis by
ing oxidative stress offers a therapeutic approach for inhibiting endothelial-​to-mesenchymal transition after
heart regeneration147. myocardial injury in animal studies160,164. These find-
ings suggest endothelial cells as potent targets for heart
Targeting the fibrotic response for heart repair. repair. Nevertheless, additional lineage-​tracing studies
Cardiac fibroblasts are activated after myocardial injury, show that the contribution of nonfibroblasts to fibroblast
expressing genes associated with contractile responses activation is more limited than previously surmised149,165.
and becoming myofibroblasts149. In the necrotic tis- Further understanding of the fibrotic response after
sue after MI, myofibroblasts and inflammatory cells myocardial injury is necessary to fulfil the therapeutic
produce a secretome that leads to the formation of a potential of targeting this process for heart regeneration.
reparative fibrotic scar (reviewed elsewhere150). The Interestingly, a study reported that injecting agrin, an
continuing activity of myofibroblasts produces fibrotic extracellular matrix protein that is enriched in the
tissue, leading to progressive pathological cardiac neonatal mouse heart, promoted heart regeneration
remodelling. Multiple signalling pathways, such as after MI in adult mice, partly through cardiomyocyte
the renin–angiotensin–aldosterone system and the dediffer­entiation166 (Table  1). Therefore, a potential
TGFβ and WNT signalling pathways, contribute to this approach for heart regeneration lies in modulating, not
fibrotic process after injury20. Targeting these pathways inhibiting, the fibrotic response.
and inhibiting the progressive phase of fibrosis has been
shown to improve cardiac function by attenuating car- Future perspectives
diac remodelling20,151–153 (Fig. 4). Inhibiting additional New advances in basic research for heart regeneration.
targets such as chymase, which is a serine protease that The identification of a transient regenerative period in
activates the renin–angiotensin–aldosterone system the heart of neonatal mice provided scientists with a new
and TGFβ signalling, also prevents cardiac fibrosis and resource to study the mechanisms governing mammal­
improves cardiac function after MI in rats154. The ian cardiac regeneration. In particular, a postnatal switch
hormone relaxin inhibits TGFβ signalling-​mediated from glycolytic to oxidative metabolism and an increase
fibroblast activation into myofibroblasts and thereby in the oxygenation state of cardiomyocytes induce the
reduces cardiac fibrosis after MI155,156. However, the production of reactive oxygen species (ROS) in mito-
approach of targeting the fibrotic response for heart chondria167. This increase in ROS levels leads to the acti-
repair is not as straightforward as anticipated. One of the vation of the DNA damage response (DDR) pathway,
reasons is that the initial phase of the fibrotic response which induces postnatal cardiomyocyte cell cycle arrest167.
is considered a healing process because transient scar Encouragingly, in this study, the postnatal cardiomyocyte
formation is also detected during heart regeneration proliferation window was extended by pharmacologically
after injury in both zebrafish and neonatal mice157. scavenging ROS or by inhibiting the DDR pathway, con-
At the healing site of the myocardial injury, angio­ sistent with findings from another study147. Moreover,
tensin and TGFβ1 have a pivotal role in the secretome exposing adult mice to gradual, severe, systemic hypoxia
of myofibroblasts and inflammatory cells150. The macro­ led to decreases in ROS production and oxidative DNA
phages at the injury site produce angiotensin II, which damage, which reactivated cardiomyocyte mitosis and
leads to the upregulation and secretion of TGFβ1. led to heart repair after MI168 (Table 1). In addition, an
Macrophage-​derived TGFβ1 induces fibroblast activation increase in cardiac mechanical load after birth has been

Nature Reviews | Cardiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
R e v ie w s

a Mouse in angiogenesis172. Future detailed analyses of the epi­


Wild-type mouse DMD mouse Genome-edited DMD genetic landscape and the transcriptome in neonatal and
heart tissue heart tissue mouse heart tissue adult mouse hearts will define the regulatory networks
that contribute to heart regeneration.
Another novel approach to consider for the treatment
of heart diseases that are caused by specific monogenic
mutations is genome editing. Discovery and engineering
of CRISPR technology has advanced the field by offering
the possibility of genome editing targeted by only one
guide RNA173,174. For example, Duchenne muscular dys-
trophy, an X-​linked recessive monogenic disease caused
by mutations in the dystrophin gene (DMD), is a severe
Dystrophin progressive muscle disease that causes premature death
b Human usually in the mid-​twenties owing to cardiac and respira-
Control iPSC-derived DMD iPSC-derived Genome-edited DMD
cardiomyocytes cardiomyocytes iPSC-derived cardiomyocytes tory failure175. In vivo genome editing with the CRISPR–
Cas9 system restored muscle function by correcting the
Dmd mutation in a mouse model of Duchenne muscu-
lar dystrophy in both the germline and the postnatal
stage176–180. In a proof-​of-principle study, genome editing
technology also corrected mutations in human iPSC-​
derived cardiomyocytes from patients with Duchenne
muscular dystrophy 181–185. Corrected iPSC-​d erived
cardiomyocytes had restored dystrophin expression
and improved cardiomyocyte functional performance
Troponin I Dystrophin in vitro, such as improved contraction and suppression
of arrhythmia (Fig. 5). Given the myriad mutations in car-
Fig. 5 | genome editing as a therapeutic approach to heart disease. Genome editing diac structural and contractile protein genes that cause
offers the possibility of correcting mutations postnatally in congenital muscle diseases, cardiomyopathy in humans, gene editing is an attractive
such as Duchenne muscular dystrophy (DMD), to restore muscle function. DMD is caused means of eliminating such disease-​causing mutations.
by mutations in the dystrophin gene (DMD) that lead to abnormalities in the production
Therefore, CRISPR technology has great potential for
of dystrophin protein and are associated with premature death owing to cardiac and
respiratory failure. The CRISPR–Cas9 system has been successfully used to correct Dmd innovative therapies to treat heart disease186.
mutations and restore the expression of dystrophin in a mouse model of DMD. a | The Genome editing could potentially be adapted for
panels show heart sections from a wild-​type mouse, a DMD mouse model, and a CRISPR– heart regenerative therapies. Genetically engineered
Cas9-edited DMD mouse model. Tissues are immunostained with an antibody for stem cells have been reported to have improved
dystrophin (green). b | The panels show induced pluripotent stem cell (iPSC)-derived cardioprotective effects187,188. Priming stem cells with
cardiomyocytes from a healthy person (control) and from a patient with DMD, and iPSCs site-​targeted modification with CRISPR technology
from a patient with DMD that were edited with CRISPR–Cas9 to correct the DMD before transplantation will reduce the risk of undesired
mutation. Cells were immunostained with antibodies for dystrophin (green) and troponin I insertional mutagenesis. This technology also opens
(red). Adapted with permission from ref.180, AAAS. new avenues to xenotransplantation. Xenografts could
be genome-​edited to silence unfavourable xenogeneic
proposed to activate cardiac mitochondrial biogenesis, genes, which potentially could reduce the risk of immune
thereby adapting the heart to the increase in energetic rejection and infection after cell transplantation189,190.
demand169. Remarkably, mechanical unloading after Additionally, CRISPR technology can be modified to
implantation of LVADs in the failing human heart caused regulate endogenous gene expression by using a cata-
a decrease in mitochondrial content and a reduction of lytically inactive form of Cas9 (dCas9)191,192. It will be
the DDR, with signs of cardiomyocyte proliferation170. interesting to see whether direct cardiac reprogramming
These findings suggest a novel therapeutic approach for can be achieved by activating endogenous gene expres-
heart regeneration whereby the proliferative capacity of sion with the use of the CRISPR–dCas9 system, which
cardiomyocytes that is silenced in the adult mammalian has been successfully reported in the reprogramming of
heart can be reawakened by environmental adjustments other cell lineages193,194. However, much work remains
rather than by directly manipulating specific signalling in addressing major issues before clinical application,
pathways in cardiomyocytes. such as off-​target mutagenesis and unstable editing effi-
A study analysing the transcriptome and the chro- ciency, as well as establishing feasible delivery methods
matin landscape in neonatal and adult mouse hearts and determining the therapeutic target population.
also revealed that noncardiomyocytes activate a
distinct injury-​induced transcriptional programme Challenges for clinical application. How can we account
in response to MI171. Another report demonstrated an for the disparity between the outcomes in preclinical
apparent difference in the immune response after MI studies of cardiac regeneration and the clinical trials of
between newborn mice at day 1 and at week 2 (ref.172). cardiac regeneration in humans? One explanation could
Xenotransplantation
Cell, tissue, or organ
Additionally, depletion of macrophages abolished the be an insufficient delivery of therapeutic factors to the
transplantation across different early neonatal cardiac regenerative capacity, and this target sites, which prevents the therapeutic thresholds
species. regenerative deficiency was associated with a defect needed for cardiac regeneration from being reached

www.nature.com/nrcardio
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
C A R D I A C R E G E N ERREAvTI EI O
wNs

Box 1 | research models for translational medicine in heart regeneration when translated to clinical settings. To address this issue,
bio­materials have attracted attention for their capacity to
Representative research models in the flow of translational research in heart serve as a matrix to improve graft survival and behaviour,
regeneration are shown in the figure. as well as to protect the therapeutic factors from degrada-
Fish tion and to act as a reservoir for sustained local delivery
Fish such as zebrafish are widely used model organisms in the early phase of translational of therapeutic factors. Biomaterial-​based delivery systems
research. High fertility rates, short gestation period, short lifespan, and low cost of thus provide a scaffold to improve the therapeutic effects
maintenance make fish a suitable model organism for high-​throughput studies using of both cellular and noncellular approaches79,102,195–197.
genetic models of heart disease206. However, fish have substantial differences compared
Furthermore, tissue-​engineering technologies have
with mammals in heart structure, having only two chambers and maintaining
allowed the building of myocardial patches and sheets
regenerative capacity throughout their lifetime. These properties make fish an invalid
model to confirm efficacy of therapeutic approaches for human heart regeneration. and 3D heart tissues recapitulating in vitro the complex
structure of heart tissue. Engineered heart tissues can be
Small animals directly transplanted to the damaged heart and have been
Mammalian model organisms are phylogenetically, anatomically, and physiologically
shown to improve therapeutic outcomes198–201. Meanwhile,
closer to humans, including the heart structure and the minimal regenerative capacity in
the adult heart. Among mammals, rodents are one of the most popular model organisms the number of deliveries of the therapeutic factor might
to study heart regeneration. The short gestation period and lifespan of rodents, also be important, because repetitive delivery of stem cells
associated with well-​understood molecular mechanisms of the cardiac regulatory was shown to improve therapeutic outcomes compared
networks, make these animal models suitable for proof-​of-principle studies using with a single delivery202. Therefore, optimizing the delivery
genetically modified animals207. A fairly low cost of maintenance and the convenience of protocol in preclinical settings is necessary to advance the
easy handling enable testing the reproducibility of regenerative approaches before potential of therapeutic factors to clinical trials.
proceeding to further studies. Nonetheless, rodents are substantially different from Another challenge in the process of clinical applica-
humans in many ways, including body size, cardiac output, and contractile proteins, tion of regenerative therapies is the limitations of current
and have a relatively homogenous genetic background; therefore, therapeutic
animal models (Box 1). Typically, proof-​of-principle
approaches tested in rodent models require an additional confirmatory step before
studies are first performed in fish or rodent models and
clinical application208.
then repeated in larger animal models to test the feasi-
large animals bility, safety, and efficacy of the approach203. Although
Large animals such as pigs, dogs, and sheep are considered to be the bridge between animal models undoubtedly have a pivotal role in trans-
small-​animal studies and clinical trials. Relatively similar to humans in body size and
lational medicine, differences between species, including
cardiac physiology, large-​animal models enable testing the feasibility, efficacy, and
long-​term safety of therapeutic approaches for heart regeneration203. However, major
the molecular basis of disease, anatomy, and physiology,
disadvantages of large animals include the high cost of maintenance and the long life might account for translational failures. In other words,
cycle compared with small animals. In addition, obtaining approval to use some species no universally ideal animal model for preclinical studies
for research purposes might be difficult. These factors hinder the design of large-​animal is available, and for each purpose and use, an appropriate
studies at a large scale with adequate numbers to confirm reproducibility. model should be selected. Additionally, preclinical stud-
Nonhuman primates ies tend to have weak statistical power because rigorously
Nonhuman primates are distinct from other large-​animal models because these animals designed, large-​scale studies require major financial
are phylogenetically closer to humans and their immune system has been well studied in investments. The lack of sufficient scientific rigour of
translational medicine209. These features give nonhuman primate models an important clinical trials of cardiac regeneration stands as a major
role in studies of allogeneic stem cell transplantation, which is considered to be one of challenge for clinical application of cardiac regenerative
the main therapeutic approaches for heart regeneration210. therapies, because positive outcomes in initial clinical
Human samples trials, which tended to be performed on a small scale,
Alternative preclinical models involve the use of human ex vivo cultured cardiac tissues were often not reproduced in later, well-​randomized,
and human pluripotent stem cell (PSC)-derived cardiomyocytes, which theoretically large-​scale clinical trials. Altogether, study designs in
should be suitable for performing in vitro proof-​of-principle studies211. preclinical settings require further refinements, and
establishing a more feasible translational path could
reduce the risk of clinical translational failure204.
(KUJ 5OCNNCPKOCNU .CTIGCPKOCNU %NKPKECN
VTKCNU
Conclusions
<GDTCȰUJ /KEG 4CVU In conclusion, new discoveries and promising preclin-
2KIU &QIU 5JGGR ical outcomes in the field of cardiac regeneration give
0QPJWOCPRTKOCVGU us optimism regarding the establishment of novel effec-
/QFGN

tive regenerative therapies for the failing human heart.


*WOCPUCORNGU However, considering the disappointing results of pre-
r'ZXKXQJWOCPVKUUWG
r25%FGTKXGFECTFKQO[QE[VGU vious clinical trials of cardiac regenerative therapies,
we must acknowledge and address the limitations of
r*KIJVJTQWIJRWV r'HȰECE[CUUGUUOGPV preclinical studies of cardiac regeneration and the dif-
2WTRQUG

r2TQQHQHRTKPEKRNGUVWFKGU r(GCUKDKNKV[CUUGUUOGPV
r.QPIVGTOUCHGV[UVWFKGU ficulties of clinical translation. In addition, mammalian
heart development is a complex and precisely spatio-
r.QYEQUV r#PCVQOKECNN[TGNGXCPV
DQF[ temporally orchestrated process205. Therefore, repairing
#FXCPVCIGU

r*KIJHGTVKNKV[TCVGU UK\GCPFJGCTVUVTWEVWTG
r5JQTVIGUVCVKQPVKOGCPFNKHGURCP r2J[UKQNQIKECNN[TGNGXCPV
the human heart will probably require a combination of
r9GNNFGUETKDGFIGPGTGIWNCVQT[
JGCTVTCVGECTFKCEQWVRWV multiple therapeutic approaches.
PGVYQTMU CPFEQPVTCEVKNGRTQVGKPU
r*GVGTQIGPGQWURQRWNCVKQP Published online xx xx xxxx

Nature Reviews | Cardiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REvIEws
1. Roth, G. A. et al. Global, regional, and national 27. Soonpaa, M. H., Kim, K. K., Pajak, L., Franklin, M. & placebo-controlled trial (MSC-​HF trial). Eur. Heart J.
burden of cardiovascular diseases for 10 causes, Field, L. J. Cardiomyocyte DNA synthesis and 36, 1744–1753 (2015).
1990 to 2015. J. Am. Coll. Cardiol. 70, 1–25 binucleation during murine development. 51. Dixon, J. A. et al. Mesenchymal cell transplantation
(2017). Am. J. Physiol. 271, H2183–H2189 (1996). and myocardial remodeling after myocardial infarction.
2. Cohn, J. N., Ferrari, R. & Sharpe, N. Cardiac 28. Haubner, B. J. et al. Functional recovery of a human Circulation 120, S220–229 (2009).
remodeling — concepts and clinical implications: neonatal heart after severe myocardial infarction. 52. Silva, G. V. et al. Mesenchymal stem cells differentiate
a consensus paper from an international forum Circ. Res. 118, 216–221 (2016). into an endothelial phenotype, enhance vascular
on cardiac remodeling. J. Am. Coll. Cardiol. 35, 29. Bergmann, O. et al. Evidence for cardiomyocyte density, and improve heart function in a canine chronic
569–582 (2000). renewal in humans. Science 324, 98–102 (2009). ischemia model. Circulation 111, 150–156 (2005).
3. Sacks, C. A., Jarcho, J. A. & Curfman, G. D. Paradigm 30. Laflamme, M. A. & Murry, C. E. Heart regeneration. 53. Messina, E. et al. Isolation and expansion of adult
shifts in heart-​failure therapy — a timeline. N. Engl. Nature 473, 326–335 (2011). cardiac stem cells from human and murine heart.
J. Med. 371, 989–991 (2014). 31. Marelli, D., Desrosiers, C., el-​Alfy, M., Kao, R. L. & Circ. Res. 95, 911–921 (2004).
4. Packer, M. et al. Effect of carvedilol on survival in Chiu, R. C. Cell transplantation for myocardial repair: 54. Oh, H. et al. Cardiac progenitor cells from adult
severe chronic heart failure. N. Engl. J. Med. 344, an experimental approach. Cell Transplant. 1, myocardium: homing, differentiation, and fusion
1651–1658 (2001). 383–390 (1992). after infarction. Proc. Natl Acad. Sci. USA 100,
5. The SOLVD Investigators. Effect of enalapril on 32. Taylor, D. A. et al. Regenerating functional 12313–12318 (2003).
survival in patients with reduced left ventricular myocardium: improved performance after skeletal 55. Laugwitz, K. L. et al. Postnatal isl1+ cardioblasts enter
ejection fractions and congestive heart failure. N. Engl. myoblast transplantation. Nat. Med. 4, 929–933 fully differentiated cardiomyocyte lineages. Nature
J. Med. 325, 293–302 (1991). (1998). 433, 647–653 (2005).
6. Pitt, B. et al. The effect of spironolactone on morbidity 33. Al Attar, N. et al. Long-​term (1 year) functional and 56. Johnston, P. V. et al. Engraftment, differentiation, and
and mortality in patients with severe heart failure. histological results of autologous skeletal muscle functional benefits of autologous cardiosphere-​derived
N. Engl. J. Med. 341, 709–717 (1999). cells transplantation in rat. Cardiovasc. Res. 58, cells in porcine ischemic cardiomyopathy. Circulation
7. McMurray, J. J. et al. Angiotensin-​neprilysin inhibition 142–148 (2003). 120, 1075–1083 (2009).
versus enalapril in heart failure. N. Engl. J. Med. 371, 34. Durrani, S., Konoplyannikov, M., Ashraf, M. & 57. Bolli, R. et al. Intracoronary delivery of autologous
993–1004 (2014). Haider, K. H. Skeletal myoblasts for cardiac repair. cardiac stem cells improves cardiac function in a
8. Rose, E. A. et al. Long-​term use of a left ventricular Regen Med. 5, 919–932 (2010). porcine model of chronic ischemic cardiomyopathy.
assist device for end-​stage heart failure. N. Engl. 35. Povsic, T. J. et al. A double-​blind, randomized, Circulation 128, 122–131 (2013).
J. Med. 345, 1435–1443 (2001). controlled, multicenter study to assess the safety and 58. Leong, Y. Y., Ng, W. H., Ellison-​Hughes, G. M. &
9. Bristow, M. R. et al. Cardiac-​resynchronization therapy cardiovascular effects of skeletal myoblast Tan, J. J. Cardiac stem cells for myocardial
with or without an implantable defibrillator in implantation by catheter delivery in patients with regeneration: they are not alone. Front. Cardiovasc.
advanced chronic heart failure. N. Engl. J. Med. 350, chronic heart failure after myocardial infarction. Med. 4, 47 (2017).
2140–2150 (2004). Am. Heart J. 162, 654–662.e1 (2011). 59. Bolli, R. et al. Cardiac stem cells in patients with
10. Yacoub, M. Cardiac donation after circulatory death: 36. Fouts, K., Fernandes, B., Mal, N., Liu, J. & ischaemic cardiomyopathy (SCIPIO): initial results of a
a time to reflect. Lancet 385, 2554–2556 (2015). Laurita, K. R. Electrophysiological consequence of randomised phase 1 trial. Lancet 378, 1847–1857
11. Behfar, A., Crespo-​Diaz, R., Terzic, A. & Gersh, B. J. skeletal myoblast transplantation in normal and (2011).
Cell therapy for cardiac repair — lessons from clinical infarcted canine myocardium. Heart Rhythm 3, 60. Chugh, A. R. et al. Administration of cardiac stem cells
trials. Nat. Rev. Cardiol. 11, 232–246 (2014). 452–461 (2006). in patients with ischemic cardiomyopathy: the SCIPIO
12. Menasche, P. et al. The Myoblast Autologous 37. Sanganalmath, S. K. & Bolli, R. Cell therapy for heart trial: surgical aspects and interim analysis of
Grafting in Ischemic Cardiomyopathy (MAGIC) trial: failure: a comprehensive overview of experimental and myocardial function and viability by magnetic
first randomized placebo-​controlled study of clinical studies, current challenges, and future resonance. Circulation 126, S54–64 (2012).
myoblast transplantation. Circulation 117, 1189–1200 directions. Circ. Res. 113, 810–834 (2013). 61. Ellison, G. M. et al. Adult c-​kit(pos) cardiac stem cells
(2008). 38. Orlic, D. et al. Bone marrow cells regenerate infarcted are necessary and sufficient for functional cardiac
13. Meyer, G. P. et al. Intracoronary bone marrow cell myocardium. Nature 410, 701–705 (2001). regeneration and repair. Cell 154, 827–842 (2013).
transfer after myocardial infarction: eighteen months’ 39. Jackson, K. A. et al. Regeneration of ischemic cardiac 62. van Berlo, J. H. et al. c-​Kit+ cells minimally contribute
follow-​up data from the randomized, controlled muscle and vascular endothelium by adult stem cells. cardiomyocytes to the heart. Nature 509, 337–341
BOOST (BOne marrOw transfer to enhance ST-​ J. Clin. Invest. 107, 1395–1402 (2001). (2014).
elevation infarct regeneration) trial. Circulation 113, 40. Schachinger, V. et al. Intracoronary bone marrow-​ 63. The Lancet, E. Expression of concern: the SCIPIO trial.
1287–1294 (2006). derived progenitor cells in acute myocardial infarction. Lancet 383, 1279 (2014).
14. Hare, J. M. et al. Comparison of allogeneic versus N. Engl. J. Med. 355, 1210–1221 (2006). 64. Makkar, R. R. et al. Intracoronary cardiosphere-​
autologous bone marrow-​derived mesenchymal 41. Traverse, J. H. et al. Effect of the use and timing of derived cells for heart regeneration after myocardial
stem cells delivered by transendocardial injection bone marrow mononuclear cell delivery on left infarction (CADUCEUS): a prospective, randomised
in patients with ischemic cardiomyopathy: ventricular function after acute myocardial infarction: phase 1 trial. Lancet 379, 895–904 (2012).
the POSEIDON randomized trial. JAMA 308, the TIME randomized trial. JAMA 308, 2380–2389 65. Murry, C. E. & Keller, G. Differentiation of embryonic
2369–2379 (2012). (2012). stem cells to clinically relevant populations: lessons
15. Beltrami, A. P. et al. Adult cardiac stem cells are 42. Perin, E. C. et al. Effect of transendocardial delivery of from embryonic development. Cell 132, 661–680
multipotent and support myocardial regeneration. autologous bone marrow mononuclear cells on (2008).
Cell 114, 763–776 (2003). functional capacity, left ventricular function, and 66. Mummery, C. et al. Differentiation of human
16. Mummery, C. L. et al. Differentiation of human perfusion in chronic heart failure: the FOCUS-​CCTRN embryonic stem cells to cardiomyocytes: role of
embryonic stem cells and induced pluripotent stem trial. JAMA 307, 1717–1726 (2012). coculture with visceral endoderm-​like cells. Circulation
cells to cardiomyocytes: a methods overview. Circ. Res. 43. Surder, D. et al. Intracoronary injection of bone 107, 2733–2740 (2003).
111, 344–358 (2012). marrow-​derived mononuclear cells early or late after 67. Qiao, H. et al. Long-​term improvement in postinfarct
17. Ieda, M. et al. Direct reprogramming of fibroblasts acute myocardial infarction: effects on global left left ventricular global and regional contractile function
into functional cardiomyocytes by defined factors. ventricular function. Circulation 127, 1968–1979 is mediated by embryonic stem cell-​derived
Cell 142, 375–386 (2010). (2013). cardiomyocytes. Circ. Cardiovasc. Imag. 4, 33–41
18. Song, K. et al. Heart repair by reprogramming non-​ 44. Pittenger, M. F. et al. Multilineage potential of adult (2011).
myocytes with cardiac transcription factors. Nature human mesenchymal stem cells. Science 284, 68. Shiba, Y. et al. Human ES-​cell-derived cardiomyocytes
485, 599–604 (2012). 143–147 (1999). electrically couple and suppress arrhythmias in injured
19. Xin, M. et al. Hippo pathway effector Yap promotes 45. Planat-​Benard, V. et al. Spontaneous cardiomyocyte hearts. Nature 489, 322–325 (2012).
cardiac regeneration. Proc. Natl Acad. Sci. USA 110, differentiation from adipose tissue stroma cells. 69. Chong, J. J. et al. Human embryonic-​stem-cell-​derived
13839–13844 (2013). Circ. Res. 94, 223–229 (2004). cardiomyocytes regenerate non-​human primate
20. Gourdie, R. G., Dimmeler, S. & Kohl, P. Novel 46. Antonitsis, P., Ioannidou-​Papagiannaki, E., Kaidoglou, A. hearts. Nature 510, 273–277 (2014).
therapeutic strategies targeting fibroblasts and & Papakonstantinou, C. In vitro cardiomyogenic 70. US National Library of Medicine. ClinicalTrials.gov
fibrosis in heart disease. Nat. Rev. Drug Discov. 15, differentiation of adult human bone marrow https://clinicaltrials.gov/show/NCT02057900 (2014).
620–638 (2016). mesenchymal stem cells. The role of 5-azacytidine. 71. Takahashi, K. & Yamanaka, S. Induction of pluripotent
21. Xin, M., Olson, E. N. & Bassel-​Duby, R. Mending Interact. Cardiovasc. Thorac. Surg. 6, 593–597 stem cells from mouse embryonic and adult fibroblast
broken hearts: cardiac development as a basis for (2007). cultures by defined factors. Cell 126, 663–676 (2006).
adult heart regeneration and repair. Nat. Rev. Mol. 47. Li, X. et al. Bone marrow mesenchymal stem cells 72. Takahashi, K. et al. Induction of pluripotent stem cells
Cell Biol. 14, 529–541 (2013). differentiate into functional cardiac phenotypes by from adult human fibroblasts by defined factors. Cell
22. Porrello, E. R. et al. Transient regenerative potential cardiac microenvironment. J. Mol. Cell Cardiol. 42, 131, 861–872 (2007).
of the neonatal mouse heart. Science 331, 295–303 (2007). 73. Kawamura, M. et al. Feasibility, safety, and therapeutic
1078–1080 (2011). 48. Quevedo, H. C. et al. Allogeneic mesenchymal stem efficacy of human induced pluripotent stem
23. Forte, E., Furtado, M. & Rosenthal, N. The interstitium cells restore cardiac function in chronic ischemic cell-derived cardiomyocyte sheets in a porcine
in cardiac repair: role of the immune–stromal cell cardiomyopathy via trilineage differentiating capacity. ischemic cardiomyopathy model. Circulation 126,
interplay. Nat. Rev. Cardiol. (in the press). Proc. Natl Acad. Sci. USA 106, 14022–14027 S29–S37 (2012).
24. Cao, J. & Poss, K. D. The epicardium as a hub for (2009). 74. Shiba, Y. et al. Allogeneic transplantation of iPS
heart regeneration. Nat. Rev. Cardiol. (in the press). 49. Amado, L. C. et al. Cardiac repair with intramyocardial cell-derived cardiomyocytes regenerates primate
25. Poss, K. D., Wilson, L. G. & Keating, M. T. Heart injection of allogeneic mesenchymal stem cells after hearts. Nature 538, 388–391 (2016).
regeneration in zebrafish. Science 298, 2188–2190 myocardial infarction. Proc. Natl Acad. Sci. USA 102, 75. Miura, K. et al. Variation in the safety of induced
(2002). 11474–11479 (2005). pluripotent stem cell lines. Nat. Biotechnol. 27,
26. Porrello, E. R. & Olson, E. N. A neonatal blueprint for 50. Mathiasen, A. B. et al. Bone marrow-​derived 743–745 (2009).
cardiac regeneration. Stem Cell Res. 13, 556–570 mesenchymal stromal cell treatment in patients 76. Sougawa, N. et al. Immunologic targeting of CD30
(2014). with severe ischaemic heart failure: a randomized eliminates tumourigenic human pluripotent stem cells,

www.nature.com/nrcardio
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
C A R D I A C R E G E N ERREAvTI EI O
wNs

allowing safer clinical application of hiPSC-​based cell 100. Liu, X. et al. miR-222 is necessary for exercise-​induced 125. Liu, Z. et al. Single-​cell transcriptomics reconstructs
therapy. Sci. Rep. 8, 3726 (2018). cardiac growth and protects against pathological fate conversion from fibroblast to cardiomyocyte.
77. Tohyama, S. et al. Distinct metabolic flow enables cardiac remodeling. Cell Metab. 21, 584–595 (2015). Nature 551, 100–104 (2017).
large-​scale purification of mouse and human 101. Aurora, A. B. et al. MicroRNA-214 protects the mouse 126. Lalit, P. A. et al. Lineage reprogramming of fibroblasts
pluripotent stem cell-​derived cardiomyocytes. Cell heart from ischemic injury by controlling Ca(2)(+) into proliferative induced cardiac progenitor cells by
Stem Cell 12, 127–137 (2013). overload and cell death. J. Clin. Invest. 122, defined factors. Cell Stem Cell 18, 354–367 (2016).
78. Tohyama, S. et al. Efficient large-​scale 2D culture 1222–1232 (2012). 127. Islas, J. F. et al. Transcription factors ETS2 and MESP1
system for human induced pluripotent stem cells and 102. Wang, L. L. et al. Sustained miRNA delivery from an transdifferentiate human dermal fibroblasts into
differentiated cardiomyocytes. Stem Cell Rep. 9, injectable hydrogel promotes cardiomyocyte cardiac progenitors. Proc. Natl Acad. Sci. USA 109,
1406–1414 (2017). proliferation and functional regeneration after 13016–13021 (2012).
79. Chow, A. et al. Human induced pluripotent stem ischaemic injury. Nat. Biomed. Engineer. 1, 128. Fu, J. D. et al. Direct reprogramming of human
cell-derived cardiomyocyte encapsulating bioactive 983–992 (2017). fibroblasts toward a cardiomyocyte-​like state.
hydrogels improve rat heart function post myocardial 103. Colombo, M., Raposo, G. & Thery, C. Biogenesis, Stem Cell Rep. 1, 235–247 (2013).
infarction. Stem Cell Rep. 9, 1415–1422 (2017). secretion, and intercellular interactions of exosomes 129. Nam, Y. J. et al. Reprogramming of human fibroblasts
80. Kawamura, M. et al. Enhanced therapeutic effects of and other extracellular vesicles. Annu. Rev. Cell Dev. toward a cardiac fate. Proc. Natl Acad. Sci. USA 110,
human iPS cell derived-​cardiomyocyte by combined Biol. 30, 255–289 (2014). 5588–5593 (2013).
cell-​sheets with omental flap technique in porcine 104. Valadi, H. et al. Exosome-​mediated transfer of mRNAs 130. Wada, R. et al. Induction of human cardiomyocyte-​like
ischemic cardiomyopathy model. Sci. Rep. 7, 8824 and microRNAs is a novel mechanism of genetic cells from fibroblasts by defined factors. Proc. Natl
(2017). exchange between cells. Nat. Cell Biol. 9, 654–659 Acad. Sci. USA 110, 12667–12672 (2013).
81. Chen, T. S. et al. Mesenchymal stem cell secretes (2007). 131. Cao, N. et al. Conversion of human fibroblasts into
microparticles enriched in pre-​microRNAs. 105. Sluijter, J. P., Verhage, V., Deddens, J. C., van den functional cardiomyocytes by small molecules. Science
Nucleic Acids Res. 38, 215–224 (2010). Akker, F. & Doevendans, P. A. Microvesicles and 352, 1216–1220 (2016).
82. Gassmann, M. et al. Aberrant neural and cardiac exosomes for intracardiac communication. 132. Yang, N., Ng, Y. H., Pang, Z. P., Sudhof, T. C. &
development in mice lacking the ErbB4 neuregulin Cardiovasc. Res. 102, 302–311 (2014). Wernig, M. Induced neuronal cells: how to make and
receptor. Nature 378, 390–394 (1995). 106. Chen, L. et al. Cardiac progenitor-​derived exosomes define a neuron. Cell Stem Cell 9, 517–525 (2011).
83. Bersell, K., Arab, S., Haring, B. & Kuhn, B. protect ischemic myocardium from acute ischemia/ 133. Pang, Z. P. et al. Induction of human neuronal cells by
Neuregulin1/ErbB4 signaling induces cardiomyocyte reperfusion injury. Biochem. Biophys. Res. Commun. defined transcription factors. Nature 476, 220–223
proliferation and repair of heart injury. Cell 138, 431, 566–571 (2013). (2011).
257–270 (2009). 107. Ibrahim, A. G., Cheng, K. & Marban, E. Exosomes as 134. Zhou, J. X. & Huang, S. Understanding gene circuits at
84. D’Uva, G. et al. ERBB2 triggers mammalian heart critical agents of cardiac regeneration triggered by cell cell-​fate branch points for rational cell reprogramming.
regeneration by promoting cardiomyocyte therapy. Stem Cell Rep. 2, 606–619 (2014). Trends Genet. 27, 55–62 (2011).
dedifferentiation and proliferation. Nat. Cell Biol. 17, 108. Barile, L. et al. Extracellular vesicles from human cardiac 135. Qian, L. et al. In vivo reprogramming of murine cardiac
627–638 (2015). progenitor cells inhibit cardiomyocyte apoptosis and fibroblasts into induced cardiomyocytes. Nature 485,
85. Gao, R. et al. A Phase II, randomized, double-​blind, improve cardiac function after myocardial infarction. 593–598 (2012).
multicenter, based on standard therapy, Cardiovasc. Res. 103, 530–541 (2014). 136. Inagawa, K. et al. Induction of cardiomyocyte-​like cells
placebo-controlled study of the efficacy and safety of 109. Gallet, R. et al. Exosomes secreted by cardiosphere-​ in infarct hearts by gene transfer of Gata4, Mef2c, and
recombinant human neuregulin-1 in patients with derived cells reduce scarring, attenuate adverse Tbx5. Circ. Res. 111, 1147–1156 (2012).
chronic heart failure. J. Am. Coll. Cardiol. 55, remodelling, and improve function in acute and 137. Nam, Y. J., Song, K. & Olson, E. N. Heart repair by
1907–1914 (2010). chronic porcine myocardial infarction. Eur. Heart J. 38, cardiac reprogramming. Nat. Med. 19, 413–415
86. Reuter, S., Soonpaa, M. H., Firulli, A. B., Chang, A. N. 201–211 (2017). (2013).
& Field, L. J. Recombinant neuregulin 1 does not 110. Arslan, F. et al. Mesenchymal stem cell-​derived 138. Miyamoto, K. et al. Direct in vivo reprogramming with
activate cardiomyocyte DNA synthesis in normal or exosomes increase ATP levels, decrease oxidative Sendai virus vectors improves cardiac function after
infarcted adult mice. PLoS ONE 9, e115871 (2014). stress and activate PI3K/Akt pathway to enhance myocardial infarction. Cell Stem Cell 22, 91–103.e5
87. Harada, K. et al. Vascular endothelial growth factor myocardial viability and prevent adverse remodeling (2018).
administration in chronic myocardial ischemia. after myocardial ischemia/reperfusion injury. Stem Cell 139. Li, H. O. et al. A cytoplasmic RNA vector derived from
Am. J. Physiol. 270, H1791–H1802 (1996). Res. 10, 301–312 (2013). nontransmissible Sendai virus with efficient gene transfer
88. Gyongyosi, M. et al. NOGA-​guided analysis of regional 111. Alvarez-​Erviti, L. et al. Delivery of siRNA to the mouse and expression. J. Virol. 74, 6564–6569 (2000).
myocardial perfusion abnormalities treated with brain by systemic injection of targeted exosomes. 140. Pasumarthi, K. B., Nakajima, H., Nakajima, H. O.,
intramyocardial injections of plasmid encoding Nat. Biotechnol. 29, 341–345 (2011). Soonpaa, M. H. & Field, L. J. Targeted expression of
vascular endothelial growth factor A-165 in patients 112. Kaufmann, K. B., Buning, H., Galy, A., Schambach, A. cyclin D2 results in cardiomyocyte DNA synthesis and
with chronic myocardial ischemia: subanalysis of the & Grez, M. Gene therapy on the move. EMBO Mol. infarct regression in transgenic mice. Circ. Res. 96,
EUROINJECT-​ONE multicenter double-​blind Med. 5, 1642–1661 (2013). 110–118 (2005).
randomized study. Circulation 112, I157–165 (2005). 113. Pinto, A. R. et al. Revisiting cardiac cellular 141. Chaudhry, H. W. et al. Cyclin A2 mediates
89. House, S. L. et al. Cardiac-​specific overexpression of composition. Circ. Res. 118, 400–409 (2016). cardiomyocyte mitosis in the postmitotic myocardium.
fibroblast growth factor-2 protects against myocardial 114. Sadahiro, T., Yamanaka, S. & Ieda, M. Direct cardiac J. Biol. Chem. 279, 35858–35866 (2004).
dysfunction and infarction in a murine model of low-flow reprogramming: progress and challenges in basic 142. Mohamed, T. M. A. et al. Regulation of cell cycle to
ischemia. Circulation 108, 3140–3148 (2003). biology and clinical applications. Circ. Res. 116, stimulate adult cardiomyocyte proliferation and
90. Simons, M. et al. Pharmacological treatment of 1378–1391 (2015). cardiac regeneration. Cell 173, 104–116.e12 (2018).
coronary artery disease with recombinant fibroblast 115. Nam, Y. J. et al. Induction of diverse cardiac cell types 143. Mahmoud, A. I. et al. Meis1 regulates postnatal
growth factor-2: double-​blind, randomized, controlled by reprogramming fibroblasts with cardiac transcription cardiomyocyte cell cycle arrest. Nature 497,
clinical trial. Circulation 105, 788–793 (2002). factors. Development 141, 4267–4278 (2014). 249–253 (2013).
91. Garbayo, E. et al. Catheter-​based Intramyocardial 116. Zhou, H., Dickson, M. E., Kim, M. S., Bassel-​Duby, R. 144. Pan, D. The hippo signaling pathway in development
Injection of FGF1 or NRG1-loaded MPs Improves & Olson, E. N. Akt1/protein kinase B enhances and cancer. Dev. Cell 19, 491–505 (2010).
Cardiac Function in a Preclinical Model of transcriptional reprogramming of fibroblasts to 145. Heallen, T. et al. Hippo pathway inhibits Wnt signaling
Ischemia-Reperfusion. Sci. Rep. 6, 25932 (2016). functional cardiomyocytes. Proc. Natl Acad. Sci. USA to restrain cardiomyocyte proliferation and heart size.
92. Zangi, L. et al. Modified mRNA directs the fate of 112, 11864–11869 (2015). Science 332, 458–461 (2011).
heart progenitor cells and induces vascular 117. Muraoka, N. et al. MiR-133 promotes cardiac 146. Heallen, T. et al. Hippo signaling impedes adult heart
regeneration after myocardial infarction. reprogramming by directly repressing Snai1 and regeneration. Development 140, 4683–4690 (2013).
Nat. Biotechnol. 31, 898–907 (2013). silencing fibroblast signatures. EMBO J. 33, 147. Tao, G. et al. Pitx2 promotes heart repair by activating
93. Bartel, D. P. MicroRNAs: target recognition and 1565–1581 (2014). the antioxidant response after cardiac injury. Nature
regulatory functions. Cell 136, 215–233 (2009). 118. Yamakawa, H. et al. Fibroblast growth factors and 534, 119–123 (2016).
94. Liu, N. & Olson, E. N. MicroRNA regulatory networks vascular endothelial growth factor promote cardiac 148. Leach, J. P. et al. Hippo pathway deficiency reverses
in cardiovascular development. Dev. Cell 18, 510–525 reprogramming under defined conditions. Stem Cell systolic heart failure after infarction. Nature 550,
(2010). Rep. 5, 1128–1142 (2015). 260–264 (2017).
95. van Rooij, E. & Olson, E. N. MicroRNA therapeutics 119. Abad, M. et al. Notch inhibition enhances cardiac 149. Tallquist, M. D. & Molkentin, J. D. Redefining the
for cardiovascular disease: opportunities and reprogramming by increasing MEF2C transcriptional identity of cardiac fibroblasts. Nat. Rev. Cardiol. 14,
obstacles. Nat. Rev. Drug Discov. 11, 860–872 activity. Stem Cell Rep. 8, 548–560 (2017). 484–491 (2017).
(2012). 120. Mohamed, T. M. et al. Chemical enhancement of 150. Weber, K. T., Sun, Y., Bhattacharya, S. K., Ahokas, R. A.
96. Porrello, E. R. et al. Regulation of neonatal and adult in vitro and in vivo direct cardiac reprogramming. & Gerling, I. C. Myofibroblast-​mediated mechanisms of
mammalian heart regeneration by the miR-15 family. Circulation 135, 978–995 (2017). pathological remodelling of the heart. Nat. Rev.
Proc. Natl Acad. Sci. USA 110, 187–192 (2013). 121. Zhao, Y. et al. High-​efficiency reprogramming of Cardiol. 10, 15–26 (2013).
97. Eulalio, A. et al. Functional screening identifies fibroblasts into cardiomyocytes requires suppression of 151. Moon, J. et al. Blockade to pathological remodeling of
miRNAs inducing cardiac regeneration. Nature 492, pro-​fibrotic signalling. Nat. Commun. 6, 8243 (2015). infarcted heart tissue using a porcupine antagonist.
376–381 (2012). 122. Jayawardena, T. M. et al. MicroRNA-​mediated in vitro Proc. Natl Acad. Sci. USA 114, 1649–1654 (2017).
98. Chen, J. et al. mir-17-92 cluster is required for and in vivo direct reprogramming of cardiac fibroblasts 152. Cittadini, A. et al. Aldosterone receptor blockade
and sufficient to induce cardiomyocyte proliferation to cardiomyocytes. Circ. Res. 110, 1465–1473 (2012). improves left ventricular remodeling and increases
in postnatal and adult hearts. Circ. Res. 112, 123. Zhou, H. et al. ZNF281 enhances cardiac ventricular fibrillation threshold in experimental heart
1557–1566 (2013). reprogramming by modulating cardiac and inflammatory failure. Cardiovasc. Res. 58, 555–564 (2003).
99. Tian, Y. et al. A microRNA-​Hippo pathway that gene expression. Genes Dev. 31, 1770–1783 (2017). 153. Duan, J. et al. Wnt1/betacatenin injury response
promotes cardiomyocyte proliferation and cardiac 124. Zhou, Y. et al. Bmi1 is a key epigenetic barrier to activates the epicardium and cardiac fibroblasts to
regeneration in mice. Sci. Transl Med. 7, 279ra38 direct cardiac reprogramming. Cell Stem Cell 18, promote cardiac repair. EMBO J. 31, 429–442
(2015). 382–395 (2016). (2012).

Nature Reviews | Cardiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
REvIEws
154. Kanemitsu, H. et al. Chymase inhibition prevents model of muscular dystrophy. Science 351, 400–403 200. Shudo, Y. et al. Novel regenerative therapy using
cardiac fibrosis and dysfunction after myocardial (2016). cell-sheet covered with omentum flap delivers a huge
infarction in rats. Hypertens. Res. 29, 57–64 178. Tabebordbar, M. et al. In vivo gene editing in number of cells in a porcine myocardial infarction
(2006). dystrophic mouse muscle and muscle stem cells. model. J. Thorac. Cardiovasc. Surg. 142, 1188–1196
155. Sassoli, C. et al. Relaxin prevents cardiac fibroblast-​ Science 351, 407–411 (2016). (2011).
myofibroblast transition via notch-1-mediated 179. Nelson, C. E. et al. In vivo genome editing improves 201. Riegler, J. et al. Human engineered heart
inhibition of TGF-​beta/Smad3 signaling. PLoS ONE 8, muscle function in a mouse model of Duchenne muscles engraft and survive long term in a rodent
e63896 (2013). muscular dystrophy. Science 351, 403–407 (2016). myocardial infarction model. Circ. Res. 117,
156. Samuel, C. S. et al. Relaxin remodels fibrotic healing 180. Amoasii, L. et al. Single-​cut genome editing restores 720–730 (2015).
following myocardial infarction. Lab. Invest. 91, dystrophin expression in a new mouse model of 202. Tokita, Y. et al. Repeated administrations of cardiac
675–690 (2011). muscular dystrophy. Sci. Transl Med. 9, eaan8081 progenitor cells are markedly more effective than a
157. Chablais, F. & Jazwinska, A. The regenerative capacity (2017). single administration: a new paradigm in cell therapy.
of the zebrafish heart is dependent on TGFbeta 181. Kazuki, Y. et al. Complete genetic correction of ips cells Circ. Res. 119, 635–651 (2016).
signaling. Development 139, 1921–1930 (2012). from Duchenne muscular dystrophy. Mol. Ther. 18, 203. Milani-​Nejad, N. & Janssen, P. M. Small and large
158. Pitt, B. et al. Effect of losartan compared with 386–393 (2010). animal models in cardiac contraction research:
captopril on mortality in patients with symptomatic 182. Young, C. S. et al. A single CRISPR-​Cas9 deletion advantages and disadvantages. Pharmacol. Ther. 141,
heart failure: randomised trial — the Losartan strategy that targets the majority of DMD patients 235–249 (2014).
Heart Failure Survival Study ELITE II. Lancet 355, restores dystrophin function in hiPSC-​derived muscle 204. Chamuleau, S. A. J. et al. Translational research in
1582–1587 (2000). cells. Cell Stem Cell 18, 533–540 (2016). cardiovascular repair: a call for a paradigm shift.
159. Ikeuchi, M. et al. Inhibition of TGF-​beta signaling 183. Kyrychenko, V. et al. Functional correction of Circ. Res. 122, 310–318 (2018).
exacerbates early cardiac dysfunction but prevents dystrophin actin binding domain mutations by genome 205. Olson, E. N. Gene regulatory networks in the
late remodeling after infarction. Cardiovasc. Res. 64, editing. JCI Insight 2, e95918 (2017). evolution and development of the heart. Science 313,
526–535 (2004). 184. Zhang, Y. et al. CRISPR-​Cpf1 correction of muscular 1922–1927 (2006).
160. Zeisberg, E. M. et al. Endothelial-​to-mesenchymal dystrophy mutations in human cardiomyocytes and 206. Major, R. J. & Poss, K. D. Zebrafish heart regeneration
transition contributes to cardiac fibrosis. Nat. Med. mice. Sci. Adv. 3, e1602814 (2017). as a model for cardiac tissue repair. Drug Discov.
13, 952–961 (2007). 185. Long, C. et al. Correction of diverse muscular Today Dis. Models 4, 219–225 (2007).
161. Zhou, B. et al. Adult mouse epicardium modulates dystrophy mutations in human engineered heart 207. Rosenthal, N. & Brown, S. The mouse ascending:
myocardial injury by secreting paracrine factors. muscle by single-​site genome editing. Sci. Adv. 4, perspectives for human-​disease models. Nat. Cell Biol.
J. Clin. Invest. 121, 1894–1904 (2011). eaap9004 (2018). 9, 993–999 (2007).
162. Mollmann, H. et al. Bone marrow-​derived cells 186. Strong, A. & Musunuru, K. Genome editing in 208. Gandolfi, F. et al. Large animal models for cardiac
contribute to infarct remodelling. Cardiovasc. Res. 71, cardiovascular diseases. Nat. Rev. Cardiol. 14, 11–20 stem cell therapies. Theriogenology 75, 1416–1425
661–671 (2006). (2017). (2011).
163. Kramann, R. et al. Perivascular Gli1+ progenitors are 187. McGinley, L. M. et al. Mesenchymal stem cell survival 209. Daadi, M. M., Barberi, T., Shi, Q. & Lanford, R. E.
key contributors to injury-​induced organ fibrosis. in the infarcted heart is enhanced by lentivirus Nonhuman primate models in translational
Cell Stem Cell 16, 51–66 (2015). vector-mediated heat shock protein 27 expression. regenerative medicine. Stem Cells Dev. 23 (Suppl. 1),
164. Zhang, H. et al. Pigment epithelium-​derived factor Hum. Gene Ther. 24, 840–851 (2013). 83–87 (2014).
attenuates myocardial fibrosis via inhibiting 188. Mohsin, S. et al. Human cardiac progenitor cells 210. Chong, J. J. & Murry, C. E. Cardiac regeneration using
Endothelial-​to-Mesenchymal Transition in rats with engineered with Pim-​I kinase enhance myocardial pluripotent stem cells — progression to large animal
acute myocardial infarction. Sci. Rep. 7, 41932 (2017). repair. J. Am. Coll. Cardiol. 60, 1278–1287 (2012). models. Stem Cell Res. 13, 654–665 (2014).
165. Kanisicak, O. et al. Genetic lineage tracing defines 189. Yang, Y. G. & Sykes, M. Xenotransplantation: current 211. Sayed, N., Liu, C. & Wu, J. C. Translation of human-​
myofibroblast origin and function in the injured heart. status and a perspective on the future. Nat. Rev. induced pluripotent stem cells: from clinical trial in a
Nat. Commun. 7, 12260 (2016). Immunol. 7, 519–531 (2007). dish to precision medicine. J. Am. Coll. Cardiol. 67,
166. Bassat, E. et al. The extracellular matrix protein agrin 190. Yang, L. et al. Genome-​wide inactivation of porcine 2161–2176 (2016).
promotes heart regeneration in mice. Nature 547, endogenous retroviruses (PERVs). Science 350,
179–184 (2017). 1101–1104 (2015). Acknowledgements
167. Puente, B. N. et al. The oxygen-​rich postnatal 191. Qi, L. S. et al. Repurposing CRISPR as an RNA-​guided The authors thank L. Amoasii and Y.-L. Min (University of
environment induces cardiomyocyte cell-​cycle arrest platform for sequence-​specific control of gene Texas Southwestern Medical Center, USA) and S. Tohyama
through DNA damage response. Cell 157, 565–579 expression. Cell 152, 1173–1183 (2013). (Keio University School of Medicine, Japan) for constructive
(2014). 192. Gilbert, L. A. et al. CRISPR-​mediated modular scientific discussions, J. Cabrera (University of Texas
168. Nakada, Y. et al. Hypoxia induces heart regeneration RNA-guided regulation of transcription in eukaryotes. Southwestern Medical Center, USA) for assistance with fig-
in adult mice. Nature 541, 222–227 (2017). Cell 154, 442–451 (2013). ures, and A. McKenzie (University of Texas Southwestern
169. Piquereau, J. et al. Mitochondrial dynamics in the 193. Black, J. B. et al. Targeted epigenetic remodeling Medical Center, USA) for help with editing. Work in the
adult cardiomyocytes: which roles for a highly of endogenous loci by CRISPR/Cas9-based authors’ laboratory is supported by grants from the NIH (AR-
specialized cell? Front. Physiol. 4, 102 (2013). transcriptional activators directly converts fibroblasts 067294, HL-130253, HD-087351, and HL-138426),
170. Canseco, D. C. et al. Human ventricular unloading to neuronal cells. Cell Stem Cell 19, 406–414 (2016). Fondation Leducq Transatlantic Networks of Excellence in
induces cardiomyocyte proliferation. J. Am. Coll. 194. Chakraborty, S. et al. A CRISPR/Cas9-based system Cardiovascular Research, and the Robert A. Welch
Cardiol. 65, 892–900 (2015). for reprogramming cell lineage specification. Stem Cell Foundation (grant 1–0025 to E.N.O.). H.H. is supported by
171. Quaife-​Ryan, G. A. et al. Multicellular transcriptional Rep. 3, 940–947 (2014). the Uehara Memorial Foundation Postdoctoral Fellowship
analysis of mammalian heart regeneration. Circulation 195. Pascual-​Gil, S., Garbayo, E., Diaz-​Herraez, P., and the Kanae Foreign Study Grant.
136, 1123–1139 (2017). Prosper, F. & Blanco-​Prieto, M. J. Heart regeneration
172. Aurora, A. B. et al. Macrophages are required for after myocardial infarction using synthetic biomaterials. Author contributions
neonatal heart regeneration. J. Clin. Invest. 124, J. Control. Release 203, 23–38 (2015). All authors researched the data for this article, discussed the
1382–1392 (2014). 196. Oduk, Y. et al. VEGF nanoparticles repair heart after content, wrote the manuscript, and reviewed and/or edited
173. Jinek, M. et al. A programmable dual-​RNA-guided myocardial infarction. Am. J. Physiol. Heart Circ. the manuscript before submission.
DNA endonuclease in adaptive bacterial immunity. Physiol. 314, H278–H284 (2018).
Science 337, 816–821 (2012). 197. Seif-​Naraghi, S. B. et al. Safety and efficacy of an Competing interests
174. Cong, L. et al. Multiplex genome engineering using injectable extracellular matrix hydrogel for treating The authors declare no competing interests.
CRISPR/Cas systems. Science 339, 819–823 (2013). myocardial infarction. Sci. Transl Med. 5, 173ra25
175. Guiraud, S. et al. The pathogenesis and therapy of (2013). Publisher’s note
muscular dystrophies. Annu. Rev. Genom. Hum. Genet. 198. Weinberger, F. et al. Cardiac repair in guinea pigs with Springer Nature remains neutral with regard to jurisdictional
16, 281–308 (2015). human engineered heart tissue from induced claims in published maps and institutional affiliations.
176. Long, C. et al. Prevention of muscular dystrophy in pluripotent stem cells. Sci. Transl Med. 8, 363ra148
mice by CRISPR/Cas9-mediated editing of germline (2016). Reviewer information
DNA. Science 345, 1184–1188 (2014). 199. Zimmermann, W. H. et al. Engineered heart tissue Nature Reviews Cardiology thanks T. Eschenhagen and the
177. Long, C. et al. Postnatal genome editing grafts improve systolic and diastolic function in other, anonymous reviewers for their contribution to the peer
partially restores dystrophin expression in a mouse infarcted rat hearts. Nat. Med. 12, 452–458 (2006). review of this work.

www.nature.com/nrcardio
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

Das könnte Ihnen auch gefallen