You are on page 1of 14

Am J Physiol Heart Circ Physiol 309: H1237–H1250, 2015.

First published September 4, 2015; doi:10.1152/ajpheart.00559.2015. Review

CALL FOR PAPERS Cardiac Regeneration and Repair: Mechanisms and Therapy

Cardiomyocyte proliferation in cardiac development and regeneration: a guide


to methodologies and interpretations
Marina Leone,1* Ajit Magadum,2* and Felix B. Engel1
1
Experimental Renal and Cardiovascular Research, Institute of Pathology, Department of Nephropathology, Friedrich-
Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany; and 2Department of Cardiology, Icahn School of
Medicine at Mount Sinai Hospital, New York, New York

Leone M, Magadum A, Engel FB. Cardiomyocyte proliferation in cardiac


development and regeneration: a guide to methodologies and interpretations. Am J
Physiol Heart Circ Physiol 309: H1237–H1250, 2015. First published September 4,
2015; doi:10.1152/ajpheart.00559.2015.—The newt and the zebrafish have the
ability to regenerate many of their tissues and organs including the heart. Thus, a
major goal in experimental medicine is to elucidate the molecular mechanisms
underlying the regenerative capacity of these species. A wide variety of experi-
ments have demonstrated that naturally occurring heart regeneration relies on
cardiomyocyte proliferation. Thus, major efforts have been invested to induce
proliferation of mammalian cardiomyocytes in order to improve cardiac function
after injury or to protect the heart from further functional deterioration. In this
review, we describe and analyze methods currently used to evaluate cardiomyocyte
proliferation. In addition, we summarize the literature on naturally occurring heart
regeneration. Our analysis highlights that newt and zebrafish heart regeneration
relies on factors that are also utilized in cardiomyocyte proliferation during
mammalian fetal development. Most of these factors have, however, failed to
induce adult mammalian cardiomyocyte proliferation. Finally, our analysis of
mammalian neonatal heart regeneration indicates experiments that could resolve
conflicting results in the literature, such as binucleation assays and clonal analysis.
Collectively, cardiac regeneration based on cardiomyocyte proliferation is a prom-
ising approach for improving adult human cardiac function after injury, but it is
important to elucidate the mechanisms arresting mammalian cardiomyocyte pro-
liferation after birth and to utilize better assays to determine formation of new
muscle mass.
cardiac regeneration; cardiomyocyte proliferation; newt; zebrafish

THE MAMMALIAN HEART FORMS from the first and second heart of cardiomyocytes by cardiomyocyte hypertrophy (increase in
field. The heart field precursors migrate and fuse at the midline cell size). In contrast to reversible physiological hypertrophy
of the embryo to form the primary linear heart tube (81). The during pregnancy or endurance training, this compensatory
heart tube then elongates, which occurs primarily via addition hypertrophy results in concentric or dilative remodeling (Fig.
of precursor-derived cardiomyocytes to both poles. Subsequent 1). It is accompanied by inflammation, fibrosis, increased wall
“ballooning” of the chambers and growth of the heart are stiffness, cardiomyocyte slippage, and continuous apoptotic
achieved by proliferation of contracting cardiomyocytes (109). loss of cardiomyocytes, resulting in reduced heart function (47,
After birth, mammalian cardiomyocytes exit the cell cycle and 57, 76).
stop proliferating (Fig. 1) (107, 115). Currently, there are no effective therapies available to re-
Cardiovascular diseases are among the leading causes of verse cardiac damage and remodeling, and, for a large group of
death worldwide, presenting a major socioeconomic burden patients, further deterioration cannot be efficiently prevented.
whose incidence is expected to increase further (39). Upon
Consequently, there is an urgent need for the development of
injury the adult mammalian heart does not regenerate, neither
novel therapies. In contrast to adult mammals, newborn mice,
by activating stem cells nor by induction of cardiomyocyte
proliferation (129). Instead, the heart compensates for the loss newts, and zebrafish regenerate their hearts after experimen-
tally induced injury by the induction of cardiomyocyte prolif-
eration (14, 38, 63). Thus, extensive efforts have been invested
* M. Leone and A. Magadum contributed equally to this work. to determine whether it is possible to induce adult mammalian
Address for reprint requests and other correspondence: F. B. Engel, Experimental
Renal and Cardiovascular Research, Inst. of Pathology, Dept. of Nephropathology,
cardiomyocyte proliferation. A large body of evidence has
Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 12, accumulated in recent years suggesting that in the future it
91054 Erlangen, Germany (e-mail: felix.engel@uk-erlangen.de). might be possible to prevent functional deterioration after
http://www.ajpheart.org 0363-6135/15 Copyright © 2015 the American Physiological Society H1237
Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
H1238 CARDIOMYOCYTE PROLIFERATION

Fetal heart Neonatal heart Adult heart

Polyploidization Polyploidization
H
E
C Pregnancy A
M G2 Sport G2
S S R
G1
T
Hypertrophy
Cell cycle Birth Cell cycle G0 Cell cycle F
G1/0 M A
G2 Injury M I
Injury C
Regeneration G1 G1 L
C U
S R
Binucleation Endomitosis Polynucleation E

Fig. 1. Cell cycle activities in fetal, neonatal, and adult mammalian cardiomyocytes. In development the ballooning of the chamber and the further growth of
the heart are accomplished by fetal cardiomyocyte proliferation. After birth, cardiomyocytes gradually stop proliferating and exit the cell cycle (G0/G1 phase).
Initially this arrest is reversible, as newborn cardiomyocytes can re-enter the cell cycle and proliferate upon injury. Yet during postnatal development
cardiomyocytes undergo one more incomplete cycle, resulting in binucleated or polyploid neonatal cardiomyocytes. In the case of cell cycle re-entry of adult
cardiomyocytes, such as during pathological hypertrophy in contrast to physiological hypertrophy (e.g., sports and pregnancy), cells undergo polyploidization,
endomitosis, and polynucleation but not cell division. This is a common feature of heart failure.

injury and even to reverse cardiac damage by inducing cardi- However, DNA synthesis occurs not only during semiconser-
omyocyte proliferation. vative DNA replication during S phase but also during DNA
repair. Moreover, semiconservative DNA replication is merely
Assays to Determine Cardiomyocyte Proliferation
an indicator of S-phase cell cycle progression (115, 129). Thus,
Cardiomyocyte cell cycle activity does not necessarily a disadvantage of this assay is that it does not predict whether
equate with proliferation as it can also reflect pathological a cell will divide or undergo G2/M arrest, polyploidization, or
hypertrophy, polyploidization, or polynucleation (115, 129). polynucleation. Pulse-chase experiments permit the identifica-
Therefore, it is important to be careful with the interpretation tion of colony formation and thus, by deduction, cell division,
of data from cell cycle assays (Table 1). Misinterpretation of as the label will be reduced by 50% per each cell division. Yet,
cell cycle assays might provide an explanation for the contro- to our knowledge, this approach has so far not been utilized in
versies in the field of cardiac regeneration, as to the extent of the field of cardiac regeneration.
cardiomyocyte proliferation. Therefore, we will discuss here Colorimetric assays. Several colorimetric proliferation as-
their advantages and disadvantages. says are available. These assays, such as MTT, Alamar Blue, or
Nucleotide analog incorporation. A commonly used ap- ViaLight assays, are based on a color change due to the
proach in determining the extent of cardiomyocyte prolifera- cleavage of tetrazolium salts, reduction of resazurin, or the
tion is the use of a nucleotide analog-incorporation assay detection of cellular ATP. An increase in the color change is
(BrdU, EdU, tritiated thymidine). Nucleotide analogs will be interpreted as an increase in cell number. However, these
incorporated during DNA synthesis, and thus cells will be assays work on the assumption that mitochondrial activity or
labeled during S phase. A major advantage of this assay is that the amount of cellular ATP per cell is constant. In this regard,
it permits the ability to perform long-term labeling, as well as these assays have limitations when applied to cardiomyocytes,
pulse-chase experiments. Long-term labeling is especially as mitochondrial activity is increased during hypertrophy or
helpful for the detection of cell cycle entry when the time elevated contractility (100). Moreover, as these assays are not
course of cell cycle activation is unclear and, furthermore, cell-type specific, it cannot be excluded that an increase in the
helps to determine the total number of cycling cardiomyocytes. colorimetric signal results from an increase in proliferation of

Table 1. Assays used to determine cardiomyocyte proliferation and their specificity


Bi-/
DNA Synthesis DNA Repair Polyploidization Mitosis Endomitosis Polynucleation Cytokinesis Proliferation Hypertrophy Cardiac specific

Nucleotide incorporation X X X X X X X
Ki67 X ? X X ? ? X X X
Histone H3 phosphorylation X X X X X X
Aurora B/Anillin X ? X X X X ?
Sarcomere disassembly X X ? X X
FACS X X X X X X
Colorimetric assays X X
Cell count X X
Clonal analysis X X
FACS, fluorescence-activated cell sorting.

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
CARDIOMYOCYTE PROLIFERATION H1239
nonmyocytes, which are present, albeit to a lesser degree, in mitosis. In contrast, during endomitosis the sarcomeric appa-
primary cardiomyocyte cultures. ratus stays intact.
Histone H3 phosphorylation. Histones undergo a wide va- Ki67. Ki67 has been shown to be required for cell prolifer-
riety of posttranslational modifications, some of which are ation, but its function is poorly understood. Ki67 is expressed
specific to mitosis (118). The most common histone modifica- in all cell cycle phases of proliferating cells but is not ex-
tion used to identify mitotic cells is the phosphorylation of pressed in non-cycling cells. Thus, Ki67 has been considered a
histone H3 at serine 10 (Ser-10) (H3P). Phosphorylation at reliable marker for evaluating cellular proliferation (18, 30).
Ser-10 begins in G2 phase and dephosphorylation is completed Furthermore, based on its intranuclear staining pattern it is
“immediately prior to detectable chromosome decondensation even possible to determine in what cell cycle phase a cell
in telophase” (Fig. 2) (24, 44). Thus, the often-used term resides, a feature that has not been utilized yet in cardiomyo-
“mitotic index” for the percentage of H3P-positive cells is cytes. However, there is evidence that Ki67 is not a reliable
misleading, as part of the H3P-positive cells are in G2 phase. marker for determining if a cardiomyocyte can proliferate. For
In addition, as with detection of S-phase activity, mitosis does instance, Meckert and coworkers (78) showed that Ki67-
not predict cell division as it can lead to polyploidization via positive cardiomyocytes undergo endomitosis. Moreover, it
endomitosis (78) or binucleation due to failure of cytokinesis has been shown that cell cycle genes as well as Ki67 are
(34). Furthermore, unlike nucleotide analog incorporation as- re-expressed in hypertrophy (28, 129). Thus, as with nucleotide
says, H3P staining cannot be combined with a cardiac-specific analog incorporation and H3P labeling for mitosis, Ki67 does
nuclear marker (e.g., Nkx2.5), as prometaphase is coupled with not predict whether a cycling cell will divide, undergo polyp-
nuclear envelope breakdown. Therefore, to determine if an loidization, or will binucleate.
H3P-positive cell is a cardiomyocyte, H3P staining is generally Aurora B and Anillin. In 2005 we introduced Aurora B as a
coupled with staining of the sarcomeric apparatus, which loses marker of cytokinesis in cardiomyocytes (33). Aurora B is
in cardiomyocytes its organized structure upon entry into expressed in G2 phase, phosphorylates histone H3 at Ser-10,

Cell division Binucleation

G2 phase
H3P/Anillin
H3P
Anillin
Fig. 2. Schematic representation of H3P, Anil-
Aurora B lin, and Aurora B localization in dividing vs.
DAPI binucleating cardiomyocytes. In G2 phase, his-
Prophase tone H3 becomes phosphorylated (H3P) while
Anillin (diffuse) and Aurora B (dotty) accumu-
late in the nucleus. During prophase, Aurora B
associates with centromeric heterochromatin,
while Anillin is translocated to the cytoplasm.
At this stage, these markers are not suitable to
distinguish proliferating from binucleating car-
diomyocytes. During anaphase, Anillin accu-
mulates at the cell cortex, and Aurora B in the
Anaphase
central spindle. Yet, as in binucleating cells,
cleavage furrow ingression is impaired; one can
find anaphase-like cells that are H3P-negative
(telophase-like nuclei) in contrast to proliferat-
ing cardiomyocytes. In cytokinesis of dividing
cardiomyocytes, Anillin localizes around the
midbody, and Aurora B concentrates at its
flanking regions. In binucleating cardiomyo-
Cytokinesis cytes, Aurora B behaves similarly; however,
Anillin fails to concentrate around the midbody.
Costaining of Aurora B and Anillin highlights
the asymmetric furrow ingression, a character-
istic of binucleating cardiomyocytes.

Cycling product

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
H1240 CARDIOMYOCYTE PROLIFERATION

and associates with centromeric heterochromatin early in mi- we have demonstrated that this complex disassembly-reassembly
tosis (Fig. 2). In contrast to H3P as a marker, Aurora B can be process occurs not only in neonatal and adult cardiomyocytes after
used to analyze cytokinesis as it transfers first from the kinet- induction of cell division but also during binucleation (33, 34).
ochore during metaphase to the central spindle during ana- Thus, sarcomere disassembly, often misleadingly called “dedif-
phase, ultimately contributing to midbody formation during ferentiation,” is not an unambiguous marker of cardiomyocyte
telophase (24, 121). Unfortunately, we realized later that Au- proliferation. However, it can be utilized to exclude prolifera-
rora B alone is also not a reliable marker for cell division, as its tion as sometimes H3P-positive cardiomyocytes are reported
staining pattern in dividing cells is indistinguishable from the with an intact sarcomeric apparatus, indicative of endomitosis
pattern in binucleating cells (34). However, Aurora B staining (32, 115).
combined with a sarcomere staining allowed us to visualize Cell count experiments. Cell count experiments are used as
cytokinesis failure during binucleation based on asymmetric a direct measurement of cardiomyocyte proliferation. Yet,
constriction of the cleavage furrow. Subsequently, we intro- often investigators measure only the total cell number, not
duced Anillin as an additional marker to evaluate whether a distinguishing between cardiomyocytes and nonmyocytes. In
cardiomyocyte will binucleate or divide. Like Ki67, Anillin is contrast, we have for example shown that the total number of
absent in G0 phase. In proliferating cells it accumulates in the neonatal cardiomyocyte increases over time after FGF1/p38
nucleus during G1, S, and G2 phase, then localizes to the cell inhibitor stimulation based on total cell number corrected for
cortex at the onset of mitosis, and finally locates around the the percentage of cardiomyocytes as analyzed by FACS (33).
midbody in cytokinesis (36, 111). Aurora B and Anillin In addition, in some studies the cell number after stimulation is
costaining identifies binucleating cells based on asymmetric compared with control stimulation. A higher cell number can
constriction of the cleavage furrow and failure of Anillin to also be explained by a cell-protective effect, as it is known that
focus at the cortex in anaphase and to concentrate around the neonatal as well as adult cardiomyocytes undergo apoptosis in
midbody during cytokinesis (34). Collectively, the sole pres- culture. In addition, cell numbers are often determined by
ence of Aurora B or Anillin is not proof of cell division. isolating cardiomyocytes from tissue or trypsination from cul-
However, the cellular localization of Anillin will indicate if a ture dishes. Here, it should be considered that alterations in
cell will divide or binucleate. extracellular matrix composition, cell-to-cell adhesion, and cell
Fluorescence-activated cell sorting. A method that has size can significantly affect the cell count due to inefficient
rarely been utilized to characterize cell cycle progression in enzymatic digestion or altered efficiency in cell harvest by
cardiomyocytes is fluorescence-activated cell sorting (FACS). centrifugation.
Measuring cell proliferation via FACS is based on labeling Clonal analysis. Clonal analysis from single cells unambig-
isolated cardiomyocytes with a cardiomyocyte-specific marker uously determines if a stimulus induces proliferation. In vitro,
and a fluorescent DNA dye, which permits determination of the clonal analysis has not yet been reported for cardiomyocytes.
DNA content based on fluorescence intensity. Using this ap- However, we have introduced the monitoring of cardiomyo-
proach, one can determine what fraction of the cardiomyocytes cytes over time, demonstrating an increase in cardiomyocyte
is for example in S phase. Time series experiments help to density after FGF1/p38 inhibitor stimulation (33). Recently,
determine whether DNA synthesis results in binucleation (in- Lin and coworkers (65) performed clonal analyses in vivo. The
crease in the G2/M phase peak), polyploidization/polynucle- authors utilized the Brainbow technology (68) to induce, in a
ation (DNA content ⱖ 4n), or cell division (no change in the small number of randomly distributed cardiomyocytes, expres-
G1, S, and G2/M distribution) (34). In addition, one can sion of a fluorescence protein (to obtain individual labeled
perform a BrdU pulse-chase experiment (diluting the BrdU cardiomyocytes). In addition, they utilized a cardiomyocyte-
label due to cell division) or a BrdU-Hoechst assay to prove specific doxycycline-inducible system to induce Yes-associ-
that cell division occurs (29). The principle of a label being ated protein expression in cardiomyocytes (65). If induction of
diluted because of cell division (such as BrdU or membrane proliferation were successful, one would expect the formation
labeling) even enables investigators to determine how often a of clones from the individual labeled cardiomyocytes (Fig. 3).
cell has divided, as each cell division results in a 50% decrease It should be noted, however, that the Brainbow system for
of the label (71). clonal analysis requires that labeled individual cells with the
Sarcomere disassembly/dedifferentiation. Previously, it has same color not be adjacent to one another. Given that nonin-
been suggested that newt and zebrafish cardiomyocytes de- duced/control hearts in the study by Lin and coworkers occa-
differentiate before undergoing proliferation based on the ob- sionally exhibited clones of two adjacent cardiomyocytes of
servation that their sarcomeric apparatus disassembles (52). the same color, this assay is thus useful only if a significant
Yet Ahuja and coworkers (3) have shown in detail that fetal number of clones of at least four labeled cardiomyocytes are
cardiomyocytes disassemble their sarcomeric apparatus in a observed. A more direct proof of cardiomyocyte division has
highly ordered fashion during mitosis and reassemble it during been introduced by Ali and coworkers (4), who have utilized
cell division. This suggests that sarcomere disassembly is not a the “mosaic analysis with double markers” mouse model (132),
sign of dedifferentiation but is simply a necessary process to which can distinguish between parental and daughter cells. In
allow cardiomyocyte proliferation. That adult mammalian car- this system parental cells are GFP- and RFP-double positive.
diomyocytes are also able to restructure or disassemble their After cell division daughter cells will be RFP or GFP positive,
sarcomeric apparatus has been long known. For example, adult while binucleated cells will be GFP- and RFP-double positive
cardiomyocytes with disassembled/disorganized/restructured (Fig. 3). As this system is dependent on Cre recombination,
sarcomeric apparatus has been observed in long-term in vitro proliferation can be analyzed specifically in cardiomyocytes.
cultures (92, 110), hypocontractile hibernating myocardium While this system has been used to determine naturally occur-
(72, 106), and Tako-Tsubo cardiomyopathy (83). Importantly, ring cardiomyocyte proliferation (4), it has not yet been uti-

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
CARDIOMYOCYTE PROLIFERATION H1241

A Myh6CreERT2; MADM-11 GT/TG


Cardiomyocyte proliferation

Myh6CreERT2
No
Tamoxifen
cytokinesis

Mitosis
G2-X

Cytokinesis

B Myh6-MerCreMer; ROSA26 fs.rt TA/+; TRE-YAP; CAG-fsBrainbow


Proliferation
Cardiomyocyte proliferation
Tamoxifen
Doxycycline
rtTA
aYAP

rtTA

Myh6-MCM aYAP

rtTA
aYAP

rtTA
aYAP
Centromere Doxycycline-dependent
rtTA Promoter rtTA reverse tetracycline
LoxP site activator gene
aYAP

rtTA
N-GFP Doxycycline-dependent
aYAP
C-GFP reverse tetracycline
N-RFP activator protein
rtTA C-RFP
aYAP Stop aYAP “Yes-associated
protein” gene
RFP
GFP
Cre Recombinase

Fig. 3. Scheme of 2 available cardiac-specific mouse models for clonal analysis. In the literature, 2 in vivo systems have been described to visualize clonal growth
of cardiomyocytes. A: the mosaic analysis with double markers (MADM) mouse model is based on split genes encoding for NH2- and COOH-terminal parts of
the fluorescent proteins red fluorescent protein (RFP) and green fluorescent protein (GFP). The system is designed such that, upon Cre recombination during
mitosis, the split genes are recombined to allow the expression of RFP in 1 daughter cell and GFP in the other (G2-X). If cell division/cytokinesis is not
completed, cells will be binucleated, expressing both RFP and GFP and appearing yellow. This model has been used to determine homeostasis in the heart of
the Myh6CreERT2 mouse. B: the 2nd model is based on the Brainbow mouse model containing several genes encoding different fluorescent proteins under the
control of a general promoter. Cre recombination results in the random expression of individual fluorescent proteins. Thus, the use of a cardiac-specific promoter
(Myh6) driving Cre will result in the labeling of individual cardiomyocytes. Activation of proliferation in such an individual labeled cardiomyocyte will then
result in single color-labeled clones. Yet this system works well only if few cardiomyocytes are labeled, several different fluorescent proteins are utilized, and
cardiomyocytes are induced to undergo several cell division. However, there is always an uncertainty, especially in the case of small-sized clones, whether a
single color-labeled clone arises from proliferation or by the same recombination event in neighboring cells. This system was utilized as a dual-color system (RFP
and GFP) to determine the effect of Yes-associated protein (YAP) on cardiomyocyte proliferation. YAP was expressed under the control of a doxycycline-
dependent reverse tetracycline activator protein.

lized to test the efficiency of factors to induce cardiomyocyte and discussed, cardiomyocyte-specific cytoplasmic markers
proliferation. are prone to misinterpretations; also the presentation of Z-
Cardiomyocyte specificity. A major issue, especially in vivo, stacks cannot solve this issue (32, 108). Thus, the detection of
is to determine whether the signal of a cell cycle marker indeed Ki67, BrdU, or cell cycle regulators such as cyclins should be
belongs to a cardiomyocyte. This is especially critical as often combined with staining for a cardiomyocyte-specific nuclear
there is an increase of fibroblast proliferation and infiltration of marker such as GATA4 or MEF2. Besides a nuclear marker,
inflammatory cells upon injury. As has been previously shown cardiomyocyte-specific cytoplasmic membrane markers such

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
H1242 CARDIOMYOCYTE PROLIFERATION

as caveolin 3 can be utilized to identify unambiguously a cells are characterized by the absence of thin filaments and Z
cycling cardiomyocyte (32). bands (87). These features have also been observed in dividing
The analysis of commonly used methods to determine car- fetal mammalian cardiomyocytes, indicating that the adult
diomyocyte proliferation shows that while many of these newt heart might indeed regenerate its heart by cardiomyocyte
techniques can detect cell cycle activity, distinguishing be- proliferation. However, this “transient disassembly of the sar-
tween proliferation, polynucleation, or polyploidization is comeric apparatus” occurs also when cells fail to undergo cell
more difficult and in some cases impossible. Not acknowledg- division after mitosis resulting in binucleated cardiomyocytes
ing this issue often results in misinterpretation of results and is (34). Thus, it was important to determine whether the number
likely the reason for the contradictions, and even controversies, of binucleated cardiomyocytes increases upon injury. Ober-
in the field of cardiac regeneration. priller and coworkers (89) reported that the vast majority
(⬎98%) of adult newt cardiomyocytes are mononucleated.
Naturally Occurring Cardiac Regeneration Importantly, they have demonstrated that the majority of newt
cardiomyocytes (⬃85%) that underwent DNA synthesis 45
Pondering regeneration is as old as civilization. First written days after injury (apex resection and reintroduction after minc-
reports by Empedocles and Aristotle describing the observation ing) were still mononucleated and diploid. These data strongly
that certain animals can regenerate their appendages can be indicated that the vast majority of adult newt cardiomyocytes
found as early as 400 BC. In modern times the first known that enter the cell cycle complete cell division.
publication regarding regeneration was by René Antoine Fer- To prove that adult newt cardiomyocytes are indeed able to
chault de Réaumur in 1712, where he described limb regener- complete cell division and to proliferate, Matz and coworkers
ation in crustaceans (crayfish, crabs, and lobsters) (41, 99). In
(77) performed life cell imaging experiments (day 8 –19 in
1740, Abraham Trembley perturbed, analyzed, and dissected
culture). These experiments demonstrated that 80% of mono-
the process of regeneration in hydra, which is considered to be
nucleated cardiomyocytes produced mononucleated daughter
the birth of “experimental biology” (60). In contrast to com-
cells, while the remaining failed to complete cytokinesis,
mon belief, regeneration is a widely distributed event with
resulting in binucleated cells. In addition, 32% of binucleated
regenerative species in almost every phylum (101).
cardiomyocytes produced binucleated daughter cells. Betten-
Urodele heart regeneration. Urodeles (salamanders and
court-Dias and coworkers (12) confirmed the proliferative
newts) are considered to be the champions of regeneration.
They can regenerate a variety of structures such as limbs, tails, competence of adult newt cardiomyocytes in vitro. They ob-
jaws, eyes, and the heart (16, 114). Urodele heart regeneration served that approximately one-third of the cells completed cell
was mentioned in the literature as early as 1974 (9, 88). After division and part of those entered successive cell divisions.
Oberpriller and Oberpriller (87) reported the appearance of These data suggest that the newt heart contains a subpopulation
mitotic adult newt cardiomyocytes in Diemictylus viridescens of cardiomyocytes that retain a higher proliferative potential,
16 days after apical resection, they studied the response of the while the remaining newt cardiomyocytes behave similarly to
adult newt ventricle to injury in Notophthalmus viridescens their mammalian counterparts.
(88). The authors resected around 12.5% of the ventricle. At To determine whether new myocardium is formed during
the site of injury a blood clot occluded the ventricle and newt regeneration, Piatkowski and coworkers (91) introduced
macrophages accumulated at 10 days within and around it. To “scanning electron microscopy analysis in combination with
determine if the injury induced cardiomyocyte proliferation the stereological estimation”. Injury induced by “repeated orthog-
authors injected tritiated thymidine 3 h before tissue collection onal squeezing with fine forceps” was followed by cardiac
to determine the frequency of cycling cardiomyocytes. The regeneration associated with a significant increase in the vol-
maximal thymidine-incorporation index (9 –10%) and mitotic ume fraction of trabeculae. In contrast to initial studies, this
index (0.9%) were observed at 20 days postinjury. Becker and and other studies have demonstrated, by investigating later
coworkers (9) provided additional support for the capacity of time points, that cardiac newt regeneration results in a mor-
urodeles to regenerate their hearts. They demonstrated that phologically fully regenerated heart with no signs of scarring
Triturus viridescens at the adult aquatic stage survives the (91, 124).
resection of 30 –50% for at least 2 wk with a mortality rate of To provide evidence that newts can functionally regenerate
10% (several hundred resections were performed). Normal their hearts, a standardized ventricular resection model was
circulatory dynamics were re-established after 5 h based on established, whereby “a more lateral region of the ventricle, as
microscopic analysis of skin capillaries in the tail fin using opposed to the apex,” (5–10%), was resected (124). Echocar-
transmitted light. These data suggested that, upon injury, the diography was utilized for the analysis of heart function and
adult newt heart might be able to regenerate its heart by revealed that heart function was reduced at day 7 postinjury
cardiomyocyte proliferation. However, the authors observed from around 38-34% fractional shortening. Full function was
the formation of scar tissue at 30 days postinjury, indicating a reported at 23 days and 60 days postinjury (⬃40% fractional
limited regenerative response (88). shortening) (124). However, it remains unclear whether the
To demonstrate that the newt heart regenerates from cardi- functional improvement is indeed due to newly formed heart
omyocyte proliferation several assays have been performed. tissue. Considering that a rather small amount of muscle tissue
Mitotic cardiomyocytes in newts were initially identified by was resected, the rather small drop in function might also be
electron microscopy based on condensed mitotic chromatin, due to the initial formation of the blood clot and scar at day 7,
“fibrils, which appeared to be myofibrils approximately the impairing wall motion. In the future it would be important to
length of a sarcomere” in the periphery of the cell, a basal repeat such a study in a model with more severe damage to the
lamina, and glycogen granules (87). In addition, these mitotic heart. This is especially important as recent studies in neonatal

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
CARDIOMYOCYTE PROLIFERATION H1243
mice suggest that the regenerative response of the heart de- ocytes or by stem cells (or some other cardiomyocyte progen-
pends on the size of injury (19, 26). itor cell) differentiating into cardiomyocytes. In addition, in
Collectively, the available data demonstrate that adult newt contrast to newt cardiomyocytes (12, 77), no protocol exists to
cardiomyocytes can proliferate in vitro and indicate that newts induce proliferation of isolated adult zebrafish cardiomyocytes
can repair their hearts after several different kinds of injury by (64, 102) in vitro.
cardiomyocyte proliferation (Table 2). To further characterize Thus, the source of newly formed cardiomyocytes remained
newt heart regeneration, echocardiographic studies should be controversial for several years until Jopling and coworkers (52)
performed comparing the functional recovery in injury models as well as Kikuchi and coworkers (54) introduced the tamox-
of varying degrees of damage. Finally, it remains unclear how ifen (4-OHT)-inducible Cre/lox system to the zebrafish model
adult proliferating newt cardiomyocytes differ from nonprolif- to conduct lineage-tracing experiments. These lineage-tracing
erating newt or mammalian cardiomyocytes. approaches demonstrated that stem/progenitor cells are not
Zebrafish heart regeneration. Zebrafish are known to regen- significantly involved in zebrafish cardiac regeneration but
erate a variety of organs, and, in contrast to urodeles, zebrafish indicated that regenerated heart muscle in zebrafish is derived
are amenable to molecular analysis and genetic manipulation. from proliferation of differentiated cardiomyocytes. Jopling
In 2002, Poss and coworkers (97) tested whether zebrafish can and coworkers (52) observed a global proliferative response of
regenerate their hearts. Similar to the experiments with urode- cardiomyocytes to mechanical injury, which was supported by
les, Poss and coworkers amputated around 20% of the apex. On the finding that an inhibitor of the cell cycle kinase Plk1
the basis of histological data, nucleotide analog (BrdU)-incor- blocked cardiac regeneration. In contrast, Kikuchi and cowork-
poration assays, and an analysis of H3P for mitosis, the authors ers (54) identified a cardiomyocyte subpopulation (GATA4-
concluded that the zebrafish can regenerate its heart by cardi- positive) throughout the subepicardial ventricular layer that
omyocyte proliferation after mechanical injury. Subsequently, contributed to heart regeneration. The importance of this sub-
similar data were obtained for other injury models including population for cardiac regeneration has been proven by induc-
cryoinjury (21) and genetic ablation (25, 119). That zebrafish ible, tissue-specific overexpression of a dominant-negative
heart regeneration is based on cardiomyocyte proliferation was Gata4 cassette (g4DN), which blocked regeneration and caused
supported by the observation that zebrafish carrying a mutation severe scarring. The data indicate that proliferation of trabec-
in a cell cycle gene failed to regenerate, forming a scar (97). ular myocytes was not affected while cortical cardiomyocyte
However, this mutation was not cell-type specific and might proliferation was markedly reduced (43).
have affected heart regeneration by inhibiting proliferation of Earlier studies were significantly limited by the lack of
another cell type. Thus, it remained unclear if regeneration longitudinal studies (observing the process of regeneration of
occurred by proliferation of remaining endogenous cardiomy- an individual heart over time) and functional studies. As it is

Table 2. Summary of cardiac regeneration in model systems


Cardiomyocyte Proliferation Heart Regeneration/Cardiac Tissue Regrowth after:

in vitro
in vivo after Genetic Mechanical Functional
Species Neonatal Adult Injury Resection Cryoinjury Ablation LAD Injury Improvement Reference

Urodeles X X 88
X X 9
X X 87
X X 89
X 12, 77
X X 91
X X X 124
Zebrafish X X 97
X X X 21
X 25
X X X X 119
X X 52
X X X 54
X X X 43
X X 50
X X 127
X X 40
Mammals (fetal/neonatal) X 61, 115
X X 19
X X X 26
X X 74, 75
X X X 93
X X X 94
X X X 6
X X 5
X 51
X X X X 56
LAD, left anterior descending coronary artery ligation.

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
H1244 CARDIOMYOCYTE PROLIFERATION

difficult to identify the amputation plane and to identify newly paired wall motion by increased pumping capacity of the
formed cardiomyocytes, it remained unclear if zebrafish can remainder of the ventricular wall. However, it remains unclear
fully restore lost myocardium and whether the observed mor- if lost cardiac tissue is fully replaced by cardiomyocyte pro-
phological regeneration is indeed due to cardiomyocyte prolif- liferation or whether hypertrophy and/or binucleation contrib-
eration. For example, the morphologically repair of a ventricle ute to this process. In the future, it will be important to
might be due to cardiomyocyte migration, which has been demonstrate that adult zebrafish cardiomyocytes can proliferate
shown to be essential for zebrafish cardiac regeneration (50). in vitro and to elucidate how adult zebrafish cardiomyocytes
To address this issue, the group of Ken Poss (22) recently differ from postnatal mammalian cardiomyocytes.
introduced so-called “zebraflash” transgenic lines for in vivo Newborn mouse heart regeneration. Fetal and newborn
bioluminescence imaging. Expression of firefly luciferase un- cardiomyocytes have the ability to proliferate (115). Conse-
der the cardiomyocyte-specific promoter cmcl2 allows for quently, the question was raised if this ability is enough to
direct monitoring of the recovery of heart size. However, this allow the fetal, embryonic, or newborn heart to regenerate after
transgenic line cannot distinguish whether recovery is due to injury. In 1975 the first evidence for fetal heart regeneration
either proliferation or hypertrophy. Moreover, these transgenic after mechanical injury in rabbits was reported (74, 75). In
lines cannot be used to determine functional recovery. 2008, Drenckhahn and coworkers (27) introduced a mouse
To demonstrate that the zebrafish can fully regenerate heart model that demonstrated that the fetal heart can compensate for
function after injury, physiological parameters have been ana- the genetic inhibition of proliferation in around 50% of fetal
lyzed, resulting in contradictory results. Analyses of cardiac cardiomyocytes. Proliferation was inhibited through inactiva-
conduction postresection in regenerating and scarring hearts tion of the X-linked gene encoding holocytochrome c synthase
initially suggested that zebrafish might not fully recover func- (Hccs). While deletion of Hccs in knockout males resulted in
tion. J-point depression was statistically significant 10 days midgestational lethality, heterozygous knockout female mice
postinjury and gradually normalized at 60 days postresection, were not affected, even though around 50% of the cardiomy-
independent of scarring or regeneration. In contrast, QTc ocytes were Hccs deficient because of the random nature of
intervals remained prolonged in both scenarios, indicating X-chromosome inactivation. The gradual increase of complex
delayed electric repolarization (127). However, Kikuchi and III activity of the mitochondrial respiratory chain, the decrease
coworkers (54) reported normalization of conduction 30 days in Hccs-deficient cardiomyocytes, and a detailed cell cycle
postresection, which was later confirmed in the cryoinjury analysis showed that the reduced proliferation rate of Hccs-
model (21). While these studies provided a more physiological negative cells is compensated for by an increased proliferation
insight into cardiac regeneration in zebrafish, recovery of rate of the healthy cardiomyocytes. However, hearts of Hccs-
cardiac conduction is a weak indicator of improved cardiac deficient female mice at postnatal day 1 were still hypoplastic.
function. Importantly, despite an increased proliferation rate adult Hccs-
In 2011, Wang and coworkers (119) introduced “swimming deficient females exhibit a range of cardiac phenotypes (28).
endurance” as a read-out to evaluate functional recovery after These data suggest that the fetal heart has only a limited
injury. Zebrafish were placed in a swim tunnel respirometer capacity for cardiac regeneration.
(MiniSwim-170; Loligo Systems, Tjele, Denmark) and al- While fetal cardiomyocytes contribute significantly to heart
lowed to acclimatize for 20 min at a low flow velocity of 3 growth by proliferation, cardiomyocytes of newborn mice have
cm/s. Swimming speed was then slowly increased to a velocity a limited proliferation capacity, and cell cycle activity gener-
of ⱖ20 cm/s, and the endurance of zebrafish was monitored. ally results in binucleation instead of cell division (61). Despite
The authors utilized this system to test the effect of inducible this fact, Porrello and coworkers (93) observed that neonatal
genetic ablation of a high percentage of cardiomyocytes mice at postnatal day 1, but not day 7, exhibit a transient
through overexpression of cytotoxic diphtheria toxin A chain. cardiac regeneration capacity. In addition, Naqvi and cowork-
They demonstrated that inducible genetic ablation of ⬎60% of ers (82) have recently reported a brief but intense proliferative
cardiomyocytes in contrast to apex resection results in reduced burst of predominantly binuclear cardiomyocytes at postnatal
performance in a swim tunnel. Recovery of this phenotype, day 15 in mice. Moreover, they report that mice at postnatal
considering an initial loss of ⬍60% together with recovery of day 15 were able to partially regenerate upon experimental
the density of cardiomyocyte nuclei, provided strong evidence myocardial infarction. Apex resection (15%) at postnatal day 1
of regeneration from proliferation, even though binucleation was restored within 21 days following a similar process as
had not been excluded (119). described for zebrafish (see above) (93). Cardiomyocyte pro-
Recently, Gonzalez-Rosa and coworkers (40) introduced the liferation was assessed by BrdU incorporation assays, phos-
use of 2D echocardiography to directly determine the pump phorylation of histone H3, aurora B kinase expression, and
function of the zebrafish heart. In a longitudinal study analyz- sarcomere disassembly, indicating an up to sevenfold increase
ing individual zebrafish over time the authors observed that in cell cycle activity. As described above these markers do not
global systolic function but not ventricular wall motion was allow one to distinguish between proliferation and binucleation
fully recovered following cardiac cryoinjury. This was in (34). However, the observation that ventricular weight and
agreement with an increased density of cardiomyocyte nuclei surface area in the absence of hypertrophy were recovered
in the injured ventricular wall that might affect normal con- within 21 days supports the conclusion that the rate of cardi-
traction due to ultrastructural changes. omyocyte proliferation is increased upon resection. In addition,
In conclusion, the available data indicate that zebrafish can, cardiac function of resected animals was comparable to sham-
to a large extent, regenerate their hearts by means of cardio- operated hearts. Yet it remains unclear whether heart function
myocyte proliferation (Table 2). In addition, it appears that in was affected and whether the normal differentiation process
case of incomplete regeneration zebrafish compensate for im- resulting in binucleation was inhibited by the resection or not.

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
CARDIOMYOCYTE PROLIFERATION H1245
In a subsequent study Porrello and coworkers (94) investigated tion it is important to understand the molecular mechanisms
whether the neonatal heart also regenerates after an ischemic controlling proliferation and to determine whether the identi-
insult induced by permanent ligation of the left anterior de- fied molecular mechanisms can be applied to human cardio-
scending (LAD) coronary artery, a more disease-related injury. myocytes. Critical questions are:
A major advantage of this study is the marked decrease in Are adult cardiomyocytes from model organisms compara-
cardiac function 4 days after ligation, which was normalized at ble to adult mammalian/human cardiomyocytes?
day 21. Moreover, the scar that formed within 7 days was Are positive regulators enough to induce adult mammalian/
resolved at day 21. In addition, in contrast to the Hccs-deficient human cardiomyocyte proliferation?
female mice, LAD-ligated mice showed no obvious pathology Urodele and zebrafish heart regeneration. Urodeles have
at 9 mo of age, indicating full regeneration. Utilizing the same been used in classical experiments to study cardiogenesis as its
assays as in their previous studies the authors concluded that development is slow and its embryos are large and develop
regeneration is due to induction of cardiomyocyte prolifera- externally (84). However, due to the lack of molecular tools the
tion. In addition, Aurora and coworkers (6) have shown that the use of urodeles is limited with regard to the elucidation of
regenerative response in the LAD model depends on macro- molecular mechanisms regulating heart development and thus
phages. also heart regeneration. Consequently, so far only microRNA
Recently, it has been suggested that, similar to the study of (miR)-128 (123) and the process of RNA editing (125) have
Drenckhahn and coworkers (28), the regenerative capacity of been implicated in newt heart regeneration. To harness the
newborn mouse hearts is limited. Andersen and coworkers (5) advantages of newt heart regeneration the group of Thomas
published the first study that challenged the apex resection Braun (15, 69, 70) has made significant progress in pioneering
model of regeneration. The authors observed neither a recovery work to elucidate the transcriptome and proteome of newt heart
in heart size and weight nor an increased proliferation rate of development and regeneration.
cardiomyocytes, even though they used the same mouse strain Even though the zebrafish heart has only two chambers, one
and a similar injury protocol as Porrello and coworkers. How- ventricle and one atrium, the myocardial wall is similar in
ever, loss and recovery of heart tissue were assessed differently structure to that of mammals, being composed of three layers:
(heart to body weight vs. ventricular weight and surface). myocardium, epicardium, and endocardium. In addition, as in
Instead of regeneration, the authors observed fibrosis and mammals, the zebrafish myocardium can be divided into com-
cardiac hypertrophy. Interestingly, Jesty and coworkers (51) pact and trabecular layers (48). That the zebrafish is a useful
had already shown that cryoinjury in neonatal mice does not model in which to study the human heart and human disease
result in scar-free regeneration within 3 mo. In addition, signs has been demonstrated in a large number of embryonic studies
of fibrosis and hypertrophy were reported in a diphtheria toxin and forward genetic screens (7). However, in contrast to
receptor-based injury model studying the dependence of neo- mammalian cardiomyocytes, zebrafish cardiomyocytes are
natal regeneration on macrophages (59). Recently, Konfino and small and mononucleated and retain proliferative potential
coworkers (56) challenged the regeneration upon LAD liga- throughout life (96, 122). Electrophysiological studies revealed
tion. While they observed complete regeneration after apical
that the adult zebrafish myocardium is very similar to that of
resection, they reported scar formation after LAD ligation in
humans based on acetylcholine-activated K⫹, Na⫹, L-type
neonatal mice with a lower cardiomyocyte proliferation rate
Ca2⫹, and IKr channels. However, these studies also revealed
than in the apex resection model.
that adult zebrafish cardiomyocytes display a robust T-type
Collectively, these studies demonstrate that, under certain
Ca2⫹ current, which, in mammals, is observed only in devel-
circumstances, neonatal hearts can recover from injury through
opment or disease. These data further suggest that adult ze-
cardiomyocyte proliferation (Table 2). It is possible that the
brafish cardiomyocytes are less mature than adult mammalian
observed differences are due to varying injury protocols. Re-
cently, two reports have indicated that the regenerative re- cardiomyocytes (85).
sponse depends on the severity of the injury. Darehzereshki Whether the underlying mechanisms of adult zebrafish re-
and coworkers (26) demonstrated that, in contrast to superficial generation can be utilized to induce mammalian cardiomyocyte
cryoinjuries, transmural cryoinjuries resulted, even after 120 proliferation remains uncertain. To date only a few regulatory
days, in severe scarring and marked reduced function. In mechanisms underlying zebrafish cardiomyocyte proliferation
addition, Bryant and coworkers (19) reported that the regener- have been identified. Ventricular injury induces raldh2 expres-
ative capacity depends on the size of the resected piece of the sion, which is known to result in retinoic acid (RA)-induced
apex. In the future, it will be important to determine how robust fetal cardiomyocyte proliferation. In addition, induced trans-
the cardiac regenerative capacity of newborn mice is and to genic inhibition of RA signaling blocked cardiac regeneration
identify the reasons for the failure of inducing regeneration (53). Besides RA signaling, it has been suggested that IGF2
upon different injury models in several laboratories. Finally, as signaling (49) and Notch signaling (131) are required for
for zebrafish, it will be important to analyze the effect of the zebrafish cardiomyocyte proliferation and cardiac regenera-
injuries on binucleation. tion. In addition, it has been shown that the cardiomyocyte-
specific miR-133 is a negative regulator of zebrafish cardio-
What Can We Learn from Naturally Occurring Cardiac myocyte proliferation (126). All of these pathways/regulators
Regeneration for Human Cardiac Therapy? have previously been shown to be involved in cardiomyocyte
proliferation during mammalian development (62, 67, 105,
It is assumed that studying animal models that can naturally 112, 116). However, none of these factors has been shown to
regenerate their hearts will result in novel therapies to treat have a significant effect on adult mammalian cardiomyocyte
human cardiac disease. In regard to cardiomyocyte prolifera- proliferation. While they might be necessary, to date they have

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
H1246 CARDIOMYOCYTE PROLIFERATION

not been shown to be sufficient to induce proliferation in adult or required for neonatal heart regeneration with the potential to
mammalian cardiomyocytes. induce adult mammalian cardiomyocyte proliferation or at
In contrast to the above-mentioned factors, Fang and co- least to improve cardiac function (95, 104). For example,
workers (35) have reported a Stat3-dependent program as Mahmoud and coworkers (73) have shown that the transcrip-
injury-specific regulator of zebrafish cardiomyocyte prolifera- tion factor Meis1 is downregulated upon myocardial infarction
tion. They identified several Jak1/Stat3 pathway members by at postnatal day 1, yet only by around 25%. siRNA-mediated
screening translating RNAs in zebrafish cardiomyocytes during knockdown in rat neonatal cardiomyocytes (age is unclear)
heart regeneration. Cardiomyocyte-specific Stat3 inhibition increased rates of histone H3 phosphorylation. Cardiomyocyte-
blocked proliferation during regeneration but not development. specific (␣MHC) Meis1 knockout mice exhibited increased
Retro-orbital introduction of the secreted protein Rln3 partially cell cycle activity (BrdU, H3P, and Aurora B) at low levels
rescued Stat3-inhibited cardiomyocyte proliferation (35). In (BrdU index of 0.3%) 14 days after birth and contained an
addition, Aguirre and coworkers (2) have recently suggested increased number of total and mononuclear cardiomyocytes.
that a microRNA program (miR-99/100, Let-7a/c, fntb, Analyses of adult Meis1 knockout hearts suggested that cardi-
smarca5) responsible for zebrafish heart regeneration can be omyocyte proliferation markedly decreased with age. Consid-
utilized to regenerate the mammalian heart. Manipulation of ering that the use of ␣MHC-Cre results in Meis1 deletion
these factors indicated that they regulate zebrafish cardiomyo- before birth, these data suggest that deletion of Meis1 might
cyte proliferation after cardiac injury. The authors subse- prevent or delay cardiomyocyte differentiation. Yet the authors
quently utilized these factors to induce adult mammalian car- confirmed induction of cell cycle activity in adult cardiomyo-
diomyocyte proliferation. They provided in vitro-only data cytes upon Meis1 deletion utilizing ␣MHC-MerCreMer mice,
regarding re-expression of proliferating cell nuclear antigen which allowed Meis1 deletion in cardiomyocytes upon tamox-
(PCNA), a DNA clamp that acts as a processivity factor for ifen administration. In fact, the cell number data indicate that
DNA polymerase-␦ in eukaryotic cells and is essential for Meis1 deletion induced the production of 2 million cardiomy-
DNA replication. In vivo, they determined expression of ocytes in less than 5 wk by an H3P as well as Aurora B fold
GATA4, PCNA, Aurora B kinase, and Anillin as well as increase of less than fourfold. This is an increase of around
histone H3 phosphorylation, and BrdU incorporation. While 40% of the total cardiomyocyte. The heart-to-body weight ratio
these data indicate proliferation, they don’t prove cardiomyo- increased by 20%. In conclusion, these data support the idea
cyte proliferation (see above). Yet cardiac function (% frac- that inhibitors control the cell cycle arrest in cardiomyocytes
tional shortening and ejection fraction) was also significantly and that this arrest is reversible. This is in accordance with the
improved, and ventricular wall thickness was recovered to observation that Meis1 deletion results in upregulation of
around 50% (2). Whether this is due to adequate levels of positive cell cycle regulators and downregulation of other
cardiomyocyte proliferation, activation of a compensatory hy- negative regulators (73). Still, it is important that these data are
pertrophic response, or a combination of both remains unclear. confirmed independently, ideally by clonal assays.
Newborn mouse heart regeneration. The mouse heart is Porrello and coworkers (94) reported that the miR-15 family
anatomically highly similar to the human heart (120). How- regulates neonatal heart regeneration. They reported that neo-
ever, the difference in size results in different physiological natal hearts of miR-195-overexpressing transgenic mice failed
demands and thus in differences to human cardiac physiology. to regenerate after LAD ligation because of reduced cardiomy-
In contrast to the human heart, the mouse heart can beat up to ocyte proliferation. Subsequently, they tested whether inhibi-
800 times per minute and exhibits significantly different cal- tion of the miR-15 family would enable adult cardiomyocyte
cium handling. On a molecular level, the human and mouse proliferation. To address this question they injected locked
hearts differ, for example, in the phosphorylation and isoform nucleic acid-modified anti-miRs postnatally at days 1, 7, and
expression of a variety of contractile proteins (e.g., myosin 14 and induced cardiac injury at day 21 by using an LAD
heavy chain, titin, troponin) (79, 86). Whether these differ- ligation/reperfusion model. Even though the authors observed
ences affect cardiomyocyte proliferation is unclear. Yet the an increase from 1 to around 5 H3P-positive cardiomyocytes
human heart might have a greater regeneration potential than per section, the treatment had only a minor effect on cardiac
those of mice and rats, as ⬎60% of adult human cardiomy- function.
ocytes remain mononucleated (but mainly polyploid) (80),
while ⬎90% of adult mouse and rat cardiomyocyte are Critical Open Questions Regarding Human Heart
bi-/polynucleated (61, 107). In addition, it has been sug- Regeneration
gested that low proliferation rates of human cardiomyocytes
can be observed until the second decade of life (80). Yet a Is the cell cycle arrest in adult mammalian/human cardio-
recent paper challenges this view and provides data demon- myocytes actively maintained and reversible? Decades ago it
strating that the final number of human cardiomyocytes is was demonstrated that the expression of a large variety of
reached at 1 mo of birth and remains constant thereafter positive cell cycle factors is downregulated in cardiomyocytes
with a low turnover rate of around 1% (11). during development, while negative regulators, such as cyclin-
Newborn heart regeneration as well as development studies dependent kinase inhibitors (CDKI), are transiently upregu-
will help to identify mechanisms required for cardiomyocyte lated (113, 115, 129). After birth, the majority of cardiomyo-
proliferation. Moreover, they might help to elucidate the mech- cytes become binucleated or polyploid (Fig. 1) (17, 45, 107),
anism underlying the cell cycle arrest. However, it remains the contractile apparatus changes dramatically into a highly
unclear whether this knowledge will lead to reversal of cell ordered structure (66), and cardiomyocytes increase in size
cycle arrest in adult cardiomyocytes, allowing heart regenera- almost 20-fold (13, 37). These changes might explain why the
tion. Yet several genes have been identified as associated with proliferative potential and thus the efficiency to induce cardi-

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
CARDIOMYOCYTE PROLIFERATION H1247
omyocyte proliferation are markedly decreased in adult mam- to be confirmed independently. A major issue here is that often
malian cardiomyocytes. Yet the mechanism that induces the limitations of the different proliferation assays are not
CDKI-dependent cell cycle arrest of mammalian cardiomyo- considered. In addition, there are no studies that follow indi-
cytes after birth, as well as its purpose, remains elusive. Sdek vidual cardiomyocytes to determine if they divide, if the
and coworkers (103) have shown that the regulation of cell resulting daughter cells are viable and functional, and whether
cycle genes in cardiomyocytes is mediated by histone methyl- they can continue to proliferate. Moreover, available clonal
ation. Possible inducers of this epigenetic regulation might be assays that can unambiguously demonstrate cardiomyocyte
alterations of the extracellular matrix around birth, as well as proliferation in vivo are rarely utilized. Importantly, the induc-
elevated levels of reactive oxygen species inducing DNA tion efficiency of mammalian cardiomyocytes decreases dra-
damage (98, 128, 129). Yet there is also evidence that the cell matically with age, which might be at least in part due to
cycle arrest might be initiated even before birth. For example, epigenetic mechanisms (103) and centrosome disassembly
fetal cardiomyocytes cannot be maintained in a proliferative (130).
state in vitro. They exit the cell cycle in vitro in a similar An important goal in achieving mammalian heart regenera-
temporal pattern as it occurs in vivo, suggesting that an tion is to understand naturally occurring heart regeneration.
intrinsic process is activated in cardiomyocytes before birth, While newt and zebrafish regeneration is well established, it
limiting the proliferative capacity of cardiomyocytes to a appears that cardiomyocytes of these species do not undergo a
distinct number of cell divisions (20). Moreover, it has recently similar cell cycle arrest as mammalian cardiomyocytes after
been shown that mammalian cardiomyocytes, in contrast to birth (130). Thus, it might not be possible to learn from newt
newt and zebrafish cardiomyocytes, start to disassemble their and zebrafish regeneration how to reverse the cell cycle arrest
centrosomes in rats as early as embryonic day 15, a process in adult mammalian cardiomyocytes. The same might be true
that is completed shortly after birth (130). As recent data have for neonatal heart regeneration. In addition, it remains unclear
demonstrated that the centrosome is a critical signaling hub for to what extent neonatal mice can regenerate their hearts (19,
the cell cycle regulatory machinery, these data indicate the loss 26, 28).
of centrosomes after birth as a possible reason why the hearts Furthermore, one has to consider carefully the mammalian
of mammals are unable to regenerate after injury. Yet the model system in which one studies the effect of candidate
observations that inhibition of p38 MAP kinase enhances factors in improving cardiac function. For example, pigs are
cardiomyocyte proliferation in the absence of a functional commonly used in large-animal studies for translational car-
centrosome (31, 130) and that newborn mice can regenerate diac studies as they share more similar function and structure
their heart after apex resection through cardiomyocyte prolif- to humans than do rodents. However, porcine cardiomyocytes
eration (93), as well as several other studies (reviewed in Refs. contain 4 –16 nuclei (1, 42). Thus, it is unclear whether
95, 104), provide some optimism for heart regeneration via large-animal models are suitable for studies of cardiac regen-
proliferation of endogenous cardiomyocytes. eration based on cardiomyocyte proliferation. However, data
Does a subpopulation of adult mammalian/human cardio- are accumulating, at least in the adult mouse heart, for the
myocytes with a regenerative potential exist? In recent years existence of a subpopulation of cardiomyocytes with neonatal
evidence has accumulated suggesting that cardiomyocyte pro- characteristics (55). These cells might be responsive to the
liferation turnover occurs in both mice and humans throughout mechanisms utilized during naturally occurring heart regener-
their lifetimes (10, 108). Yet it is unclear if all cardiomyocytes ation. Thus, in future study it will be important to determine in
contribute to this turnover or whether a subpopulation with a which species such subpopulation of cardiomyocytes exist.
higher proliferative capacity exists. The existence of such a Finally, it should be considered that the heart does not
subpopulation of mononucleated cardiomyocytes was sug- consist only of cardiomyocytes. Cardiomyocytes contribute
gested as early as in 2007 (58) and has recently been substan- only around 40 – 60% of the cells in mammalian hearts (8, 23,
tiated by a fate mapping study identifying a “rare population of 46, 90, 117). Thus it appears important to better understand
hypoxic cardiomyocytes that display characteristics of prolif- how the current regenerative approaches contribute to the
erative neonatal cardiomyocytes, such as smaller size, mono- formation of new complex cardiac tissue, how the dynamic of
nucleation and lower oxidative DNA damage” (55). the process is, and whether dividing cardiomyocytes actively
contribute to it.
Conclusions ACKNOWLEDGMENTS

A large amount of data has been accumulated that demon- We thank the rest of the Engel lab for critical reading and suggestions.
strates that, in newt, zebrafish, and newborn mice, a variety of GRANTS
injuries induce heart regeneration by cardiomyocyte prolifera-
This work was supported by grants from the Emerging Fields Initiative
tion. In addition, it has been shown that it is possible to (EFI) for Cell Cycle in Disease and Regeneration (CYDER) from the
efficiently induce neonatal mammalian cardiomyocyte prolif- Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU to F. B. Engel) and
eration. In contrast, with regard to adult cardiomyocytes, little the Deutsche Forschungsgemeinschaft (IRTG 1566, PROMISE; EN 453/9-1 to
evidence exists supporting true proliferation (i.e., successful F. B. Engel).
completion of cytokinesis resulting in two cells). Moreover, DISCLOSURES
much of the data suggestive of adult cardiomyocyte prolifera-
No conflicts of interest, financial or otherwise, are declared by the author(s).
tion is mainly based on immunofluorescence analysis, which,
depending on the scrutiny and method of analysis, can lead to AUTHOR CONTRIBUTIONS
misinterpretations. Ultimately, much of the data underlying Author contributions: M.L. and F.B.E. analyzed data; M.L. and F.B.E.
claims of regeneration by cardiomycoyte proliferation has yet prepared figures; M.L., A.M., and F.B.E. drafted manuscript; M.L., A.M., and

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
H1248 CARDIOMYOCYTE PROLIFERATION

F.B.E. edited and revised manuscript; M.L., A.M., and F.B.E. approved final 23. Chottova Dvorakova M, Wiegand S, Pesta M, Slavikova J, Grau V,
version of manuscript. Reischig J, Kuncova J, Kummer W. Expression of neuropeptide Y and
its receptors Y1 and Y2 in the rat heart and its supplying autonomic and
spinal sensory ganglia in experimentally induced diabetes. Neuroscience
REFERENCES 151: 1016 –1028, 2008.
1. Adler CP, Friedburg H, Herget GW, Neuburger M, Schwalb H. 24. Crosio C, Fimia GM, Loury R, Kimura M, Okano Y, Zhou H, Sen S,
Variability of cardiomyocyte DNA content, ploidy level and nuclear Allis CD, Sassone-Corsi P. Mitotic phosphorylation of histone H3:
number in mammalian hearts. Virchows Arch 429: 159 –164, 1996. spatio-temporal regulation by mammalian Aurora kinases. Mol Cell Biol
2. Aguirre A, Montserrat N, Zacchigna S, Nivet E, Hishida T, Krause 22: 874 –885, 2002.
MN, Kurian L, Ocampo A, Vazquez-Ferrer E, Rodriguez-Esteban C, 25. Curado S, Anderson RM, Jungblut B, Mumm J, Schroeter E,
Kumar S, Moresco JJ, Yates JR 3rd, Campistol JM, Sancho-Marti- Stainier DY. Conditional targeted cell ablation in zebrafish: a new tool
nez I, Giacca M, Izpisua Belmonte JC. In vivo activation of a for regeneration studies. Dev Dyn 236: 1025–1035, 2007.
conserved microRNA program induces mammalian heart regeneration. 26. Darehzereshki A, Rubin N, Gamba L, Kim J, Fraser J, Huang Y,
Cell Stem Cell 15: 589 –604, 2014. Billings J, Mohammadzadeh R, Wood J, Warburton D, Kaartinen V,
3. Ahuja P, Perriard E, Perriard JC, Ehler E. Sequential myofibrillar Lien CL. Differential regenerative capacity of neonatal mouse hearts
breakdown accompanies mitotic division of mammalian cardiomyocytes. after cryoinjury. Dev Biol 399: 91–99, 2014.
J Cell Sci 117: 3295–3306, 2004. 27. Drenckhahn JD, Schwarz QP, Gray S, Laskowski A, Kiriazis H,
4. Ali SR, Hippenmeyer S, Saadat LV, Luo L, Weissman IL, Ardehali Ming Z, Harvey RP, Du XJ, Thorburn DR, Cox TC. Compensatory
R. Existing cardiomyocytes generate cardiomyocytes at a low rate after growth of healthy cardiac cells in the presence of diseased cells restores
birth in mice. Proc Natl Acad Sci USA 111: 8850 –8855, 2014. tissue homeostasis during heart development. Dev Cell 15: 521–533,
5. Andersen DC, Ganesalingam S, Jensen CH, Sheikh SP. Do neonatal 2008.
mouse hearts regenerate following heart apex resection? Stem Cell 28. Drenckhahn JD, Strasen J, Heinecke K, Langner P, Yin KV, Skole F,
Reports 2: 406 –413, 2014. Hennig M, Spallek B, Fischer R, Blaschke F, Heuser A, Cox TC,
6. Aurora AB, Porrello ER, Tan W, Mahmoud AI, Hill JA, Bassel- Black MJ, Thierfelder L. Impaired myocardial development resulting in
Duby R, Sadek HA, Olson EN. Macrophages are required for neonatal neonatal cardiac hypoplasia alters postnatal growth and stress response in
heart regeneration. J Clin Invest 124: 1382–1392, 2014. the heart. Cardiovasc Res 106: 43–54, 2015.
7. Bakkers J. Zebrafish as a model to study cardiac development and 29. Ebelt H, Hufnagel N, Neuhaus P, Neuhaus H, Gajawada P, Simm A,
human cardiac disease. Cardiovasc Res 91: 279 –288, 2011. Muller-Werdan U, Werdan K, Braun T. Divergent siblings: E2F2 and
8. Banerjee I, Fuseler JW, Price RL, Borg TK, Baudino TA. Determi- E2F4 but not E2F1 and E2F3 induce DNA synthesis in cardiomyocytes
nation of cell types and numbers during cardiac development in the without activation of apoptosis. Circ Res 96: 509 –517, 2005.
neonatal and adult rat and mouse. Am J Physiol Heart Circ Physiol 293: 30. Endl E, Gerdes J. The Ki-67 protein: fascinating forms and an unknown
H1883–H1891, 2007. function. Exp Cell Res 257: 231–237, 2000.
9. Becker RO, Chapin S, Sherry R. Regeneration of the ventricular 31. Engel FB. Cardiomyocyte proliferation: a platform for mammalian
myocardium in amphibians. Nature 248: 145–147, 1974. cardiac repair. Cell Cycle 4: 1360 –1363, 2005.
10. Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabe-Heider 32. Engel FB, Hsieh PC, Lee RT, Keating MT. FGF1/p38 MAP kinase
F, Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, Jovinge S, inhibitor therapy induces cardiomyocyte mitosis, reduces scarring, and
Frisen J. Evidence for cardiomyocyte renewal in humans. Science 324: rescues function after myocardial infarction. Proc Natl Acad Sci USA
98 –102, 2009. 103: 15546 –15551, 2006.
11. Bergmann O, Zdunek S, Felker A, Salehpour M, Alkass K, Bernard 33. Engel FB, Schebesta M, Duong MT, Lu G, Ren S, Madwed JB, Jiang
S, Sjostrom SL, Szewczykowska M, Jackowska T, Dos Remedios C, H, Wang Y, Keating MT. p38 MAP kinase inhibition enables prolifer-
Malm T, Andra M, Jashari R, Nyengaard JR, Possnert G, Jovinge S, ation of adult mammalian cardiomyocytes. Genes Dev 19: 1175–1187,
Druid H, Frisen J. Dynamics of cell generation and turnover in the 2005.
human heart. Cell 161: 1566 –1575, 2015. 34. Engel FB, Schebesta M, Keating MT. Anillin localization defect in
12. Bettencourt-Dias M, Mittnacht S, Brockes JP. Heterogeneous prolif- cardiomyocyte binucleation. J Mol Cell Cardiol 41: 601–612, 2006.
erative potential in regenerative adult newt cardiomyocytes. J Cell Sci 35. Fang Y, Gupta V, Karra R, Holdway JE, Kikuchi K, Poss KD.
116: 4001–4009, 2003. Translational profiling of cardiomyocytes identifies an early Jak1/Stat3
13. Bishop SP, Hine P. Carciac muscle cytoplasmic and nuclear develop- injury response required for zebrafish heart regeneration. Proc Natl Acad
ment during canine neonatal growth. Recent Adv Stud Card Struct Metab Sci USA 110: 13416 –13421, 2013.
8: 77–98, 1975. 36. Field CM, Alberts BM. Anillin, a contractile ring protein that cycles
14. Borchardt T, Braun T. Cardiovascular regeneration in non-mammalian from the nucleus to the cell cortex. J Cell Biol 131: 165–178, 1995.
model systems: what are the differences between newts and man? 37. Fraticelli A, Josephson R, Danziger R, Lakatta E, Spurgeon H.
Thromb Haemost 98: 311–318, 2007. Morphological and contractile characteristics of rat cardiac myocytes
15. Borchardt T, Looso M, Bruckskotten M, Weis P, Kruse J, Braun T. from maturation to senescence. Am J Physiol Heart Circ Physiol 257:
Analysis of newly established EST databases reveals similarities between H259 –H265, 1989.
heart regeneration in newt and fish. BMC Genomics 11: 4, 2010. 38. Gamba L, Harrison M, Lien CL. Cardiac regeneration in model
16. Brockes JP, Kumar A. Appendage regeneration in adult vertebrates and organisms. Curr Treat Options Cardiovasc Med 16: 288, 2014.
implications for regenerative medicine. Science 310: 1919 –1923, 2005. 39. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha
17. Brodsky V, Sarkisov DS, Arefyeva AM, Panova NW, Gvasava IG. MJ, Dai S, Ford ES, Fox CS, Franco S, Fullerton HJ, Gillespie C,
Polyploidy in cardiac myocytes of normal and hypertrophic human Hailpern SM, Heit JA, Howard VJ, Huffman MD, Judd SE, Kissela
hearts; range of values. Virchows Arch 424: 429 –435, 1994. BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Mackey
18. Brown DC, Gatter KC. Ki67 protein: the immaculate deception? RH, Magid DJ, Marcus GM, Marelli A, Matchar DB, McGuire DK,
Histopathology 40: 2–11, 2002. Mohler ER, 3rd Moy CS, Mussolino ME, Neumar RW, Nichol G,
19. Bryant DM, O’Meara CC, Ho NN, Gannon J, Cai L, Lee RT. A Pandey DK, Paynter NP, Reeves MJ, Sorlie PD, Stein J, Towfighi A,
systematic analysis of neonatal mouse heart regeneration after apical Turan TN, Virani SS, Wong ND, Woo D, Turner MB. Heart disease
resection. J Mol Cell Cardiol 79: 315–318, 2015. and stroke statistics–2014 update: a report from the American Heart
20. Burton PB, Raff MC, Kerr P, Yacoub MH, Barton PJ. An intrinsic Association. Circulation 129: e28 –e292, 2014.
timer that controls cell-cycle withdrawal in cultured cardiac myocytes. 40. Gonzalez-Rosa JM, Guzman-Martinez G, Marques IJ, Sanchez-
Dev Biol 216: 659 –670, 1999. Iranzo H, Jimenez-Borreguero LJ, Mercader N. Use of echocardiog-
21. Chablais F, Veit J, Rainer G, Jazwinska A. The zebrafish heart raphy reveals reestablishment of ventricular pumping efficiency and
regenerates after cryoinjury-induced myocardial infarction. BMC Dev partial ventricular wall motion recovery upon ventricular cryoinjury in
Biol 11: 21, 2011. the zebrafish. PLoS One 9: e115604, 2014.
22. Chen CH, Durand E, Wang J, Zon LI, Poss KD. Zebraflash transgenic 41. Goss RJ. The natural history (and mystery) of regeneration. In: A History
lines for in vivo bioluminescence imaging of stem cells and regeneration of Regeneration Research: Milestones in the Evolution of a Science.
in adult zebrafish. Development 140: 4988 –4997, 2013. Cambridge, UK: Cambridge University Press, 1991.

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
CARDIOMYOCYTE PROLIFERATION H1249
42. Grabner W, Pfitzer P. Number of nuclei in isolated myocardial cells of 64. Lien CL, Schebesta M, Makino S, Weber GJ, Keating MT. Gene
pigs. Virchows Arch B Cell Pathol Incl Mol Pathol 15: 279 –294, 1974. expression analysis of zebrafish heart regeneration. PLoS Biol 4: e260,
43. Gupta V, Gemberling M, Karra R, Rosenfeld GE, Evans T, Poss KD. 2006.
An injury-responsive gata4 program shapes the zebrafish cardiac ventri- 65. Lin Z, von Gise A, Zhou P, Gu F, Ma Q, Jiang J, Yau AL, Buck JN,
cle. Curr Biol 23: 1221–1227, 2013. Gouin KA, van Gorp PR, Zhou B, Chen J, Seidman JG, Wang DZ, Pu
44. Hendzel MJ, Wei Y, Mancini MA, Van Hooser A, Ranalli T, Brin- WT. Cardiac-specific YAP activation improves cardiac function and sur-
kley BR, Bazett-Jones DP, Allis CD. Mitosis-specific phosphorylation vival in an experimental murine MI model. Circ Res 115: 354 –363, 2014.
of histone H3 initiates primarily within pericentromeric heterochromatin 66. Linke WA, Hamdani N. Gigantic business: titin properties and function
during G2 and spreads in an ordered fashion coincident with mitotic through thick and thin. Circ Res 114: 1052–1068, 2014.
chromosome condensation. Chromosoma 106: 348 –360, 1997. 67. Liu N, Bezprozvannaya S, Williams AH, Qi X, Richardson JA,
45. Herget GW, Neuburger M, Plagwitz R, Adler CP. DNA content, Bassel-Duby R, Olson EN. microRNA-133a regulates cardiomyocyte
ploidy level and number of nuclei in the human heart after myocardial proliferation and suppresses smooth muscle gene expression in the heart.
infarction. Cardiovasc Res 36: 45–51, 1997. Genes Dev 22: 3242–3254, 2008.
46. Higuchi K, Hashizume H, Aizawa Y, Ushiki T. Scanning electron 68. Livet J, Weissman TA, Kang H, Draft RW, Lu J, Bennis RA, Sanes
microscopic studies of the vascular smooth muscle cells and pericytes in JR, Lichtman JW. Transgenic strategies for combinatorial expression of
the rat heart. Arch Histol Cytol 63: 115–126, 2000. fluorescent proteins in the nervous system. Nature 450: 56 –62, 2007.
47. Hou J, Kang YJ. Regression of pathological cardiac hypertrophy: 69. Looso M, Michel CS, Konzer A, Bruckskotten M, Borchardt T,
signaling pathways and therapeutic targets. Pharmacol Ther 135: 337– Kruger M, Braun T. Spiked-in pulsed in vivo labeling identifies a new
354, 2012. member of the CCN family in regenerating newt hearts. J Proteome Res
48. Hu N, Yost HJ, Clark EB. Cardiac morphology and blood pressure in 11: 4693–4704, 2012.
the adult zebrafish. Anat Rec 264: 1–12, 2001. 70. Looso M, Preussner J, Sousounis K, Bruckskotten M, Michel CS,
49. Huang Y, Harrison MR, Osorio A, Kim J, Baugh A, Duan C, Sucov Lignelli E, Reinhardt R, Hoffner S, Kruger M, Tsonis PA, Borchardt
HM, Lien CL. Igf signaling is required for cardiomyocyte proliferation T, Braun T. A de novo assembly of the newt transcriptome combined
during zebrafish heart development and regeneration. PLoS One 8: with proteomic validation identifies new protein families expressed
e67266, 2013. during tissue regeneration. Genome Biol 14: R16, 2013.
50. Itou J, Oishi I, Kawakami H, Glass TJ, Richter J, Johnson A, Lund 71. Lyons AB. Analysing cell division in vivo and in vitro using flow
TC, Kawakami Y. Migration of cardiomyocytes is essential for heart cytometric measurement of CFSE dye dilution. J Immunol Meth 243:
regeneration in zebrafish. Development 139: 4133–4142, 2012. 147–154, 2000.
51. Jesty SA, Steffey MA, Lee FK, Breitbach M, Hesse M, Reining S, Lee 72. Maes A, Flameng W, Nuyts J, Borgers M, Shivalkar B, Ausma J,
JC, Doran RM, Nikitin AY, Fleischmann BK, Kotlikoff MI. c-kit⫹ Bormans G, Schiepers C, De Roo M, Mortelmans L. Histological
precursors support postinfarction myogenesis in the neonatal, but not alterations in chronically hypoperfused myocardium. Correlation with
adult, heart. Proc Natl Acad Sci USA 109: 13380 –13385, 2012. PET findings. Circulation 90: 735–745, 1994.
52. Jopling C, Sleep E, Raya M, Marti M, Raya A, Izpisua Belmonte JC. 73. Mahmoud AI, Kocabas F, Muralidhar SA, Kimura W, Koura AS,
Zebrafish heart regeneration occurs by cardiomyocyte dedifferentiation Thet S, Porrello ER, Sadek HA. Meis1 regulates postnatal cardiomy-
and proliferation. Nature 464: 606 –609, 2010. ocyte cell cycle arrest. Nature 497: 249 –253, 2013.
53. Kikuchi K, Holdway JE, Major RJ, Blum N, Dahn RD, Begemann G, 74. Malyshev II. [Mitotic activity of the myocardium in early ontogenesis of
Poss KD. Retinoic acid production by endocardium and epicardium is an rabbits after minor mechanical injury to the heart]. Biull Eksp Biol Med
injury response essential for zebrafish heart regeneration. Dev Cell 20: 97: 734 –736, 1984.
397–404, 2011. 75. Malyshev II. [The recovery process in the fetal rabbit myocardium
54. Kikuchi K, Holdway JE, Werdich AA, Anderson RM, Fang Y, following mechanical trauma]. Biull Eksp Biol Med 80: 111–113, 1975.
Egnaczyk GF, Evans T, Macrae CA, Stainier DY, Poss KD. Primary 76. Mann N, Rosenzweig A. Can exercise teach us how to treat heart
contribution to zebrafish heart regeneration by gata4(⫹) cardiomyocytes. disease? Circulation 126: 2625–2635, 2012.
Nature 464: 601–605, 2010. 77. Matz DG, Oberpriller JO, Oberpriller JC. Comparison of mitosis in
55. Kimura W, Xiao F, Canseco DC, Muralidhar S, Thet S, Zhang HM, binucleated and mononucleated newt cardiac myocytes. Anat Rec 251:
Abderrahman Y, Chen R, Garcia JA, Shelton JM, Richardson JA, 245–255, 1998.
Ashour AM, Asaithamby A, Liang H, Xing C, Lu Z, Zhang CC, 78. Meckert PC, Rivello HG, Vigliano C, Gonzalez P, Favaloro R,
Sadek HA. Hypoxia fate mapping identifies cycling cardiomyocytes in Laguens R. Endomitosis and polyploidization of myocardial cells in the
the adult heart. Nature 523: 226 –230, 2015. periphery of human acute myocardial infarction. Cardiovasc Res 67:
56. Konfino T, Landa N, Ben-Mordechai T, Leor J. The type of injury 116 –123, 2005.
dictates the mode of repair in neonatal and adult heart. J Am Heart Assoc 79. Milani-Nejad N, Janssen PM. Small and large animal models in cardiac
4: e001320, 2015. contraction research: advantages and disadvantages. Pharmacol Ther
57. Kostin S, Hein S, Arnon E, Scholz D, Schaper J. The cytoskeleton and 141: 235–249, 2014.
related proteins in the human failing heart. Heart Fail Rev 5: 271–280, 2000. 80. Mollova M, Bersell K, Walsh S, Savla J, Das LT, Park SY, Silber-
58. Kuhn B, del Monte F, Hajjar RJ, Chang YS, Lebeche D, Arab S, stein LE, Dos Remedios CG, Graham D, Colan S, Kuhn B. Cardio-
Keating MT. Periostin induces proliferation of differentiated cardiomy- myocyte proliferation contributes to heart growth in young humans. Proc
ocytes and promotes cardiac repair. Nat Med 13: 962–969, 2007. Natl Acad Sci USA 110: 1446 –1451, 2013.
59. Lavine KJ, Epelman S, Uchida K, Weber KJ, Nichols CG, Schilling 81. Moorman AF, Christoffels VM. Cardiac chamber formation: develop-
JD, Ornitz DM, Randolph GJ, Mann DL. Distinct macrophage lin- ment, genes, and evolution. Physiol Rev 83: 1223–1267, 2003.
eages contribute to disparate patterns of cardiac recovery and remodeling 82. Naqvi N, Li M, Calvert JW, Tejada T, Lambert JP, Wu J, Kesteven
in the neonatal and adult heart. Proc Natl Acad Sci USA 111: 16029 – SH, Holman SR, Matsuda T, Lovelock JD, Howard WW, Iismaa SE,
16034, 2014. Chan AY, Crawford BH, Wagner MB, Martin DI, Lefer DJ, Gra-
60. Lenhoff SG, Lenhoff HM. Hydra and the Birth of Experimental Biol- ham RM, Husain A. A proliferative burst during preadolescence estab-
ogy, 1744: Abraham Trembley’s Memoires Concerning the Polyps. lishes the final cardiomyocyte number. Cell 157: 795–807, 2014.
Pacific Grove, CA: Boxwood, 1986. 83. Nef HM, Mollmann H, Kostin S, Troidl C, Voss S, Weber M, Dill T,
61. Li F, Wang X, Capasso JM, Gerdes AM. Rapid transition of cardiac Rolf A, Brandt R, Hamm CW, Elsasser A. Tako-Tsubo cardiomyop-
myocytes from hyperplasia to hypertrophy during postnatal development. athy: intraindividual structural analysis in the acute phase and after
J Mol Cell Cardiol 28: 1737–1746, 1996. functional recovery. Eur Heart J 28: 2456 –2464, 2007.
62. Li P, Cavallero S, Gu Y, Chen TH, Hughes J, Hassan AB, Bruning 84. Neff AW, Dent AE, Armstrong JB. Heart development and regenera-
JC, Pashmforoush M, Sucov HM. IGF signaling directs ventricular tion in urodeles. Int J Dev Biol 40: 719 –725, 1996.
cardiomyocyte proliferation during embryonic heart development. De- 85. Nemtsas P, Wettwer E, Christ T, Weidinger G, Ravens U. Adult
velopment 138: 1795–1805, 2011. zebrafish heart as a model for human heart? An electrophysiological
63. Lien CL, Harrison MR, Tuan TL, Starnes VA. Heart repair and study. J Mol Cell Cardiol 48: 161–171, 2010.
regeneration: recent insights from zebrafish studies. Wound Repair Regen 86. Noujaim SF, Lucca E, Munoz V, Persaud D, Berenfeld O, Meijler
20: 638 –646, 2012. FL, Jalife J. From mouse to whale: a universal scaling relation for the

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.
Review
H1250 CARDIOMYOCYTE PROLIFERATION

PR Interval of the electrocardiogram of mammals. Circulation 110: and proliferation characterizes early chamber formation. Circ Res 99:
2802–2808, 2004. 545–552, 2006.
87. Oberpriller J, Oberpriller JC. Mitosis in adult newt ventricle. J Cell 110. Spahr R, Piper HM, Schwartz P, Probst I, Spieckermann PG.
Biol 49: 560 –563, 1971. Morphological dedifferentiation of adult cardiac myocytes in coculture
88. Oberpriller JO, Oberpriller JC. Response of the adult newt ventricle to with hepatocytes. Basic Res Cardiol 80: 83–86, 1985.
injury. J Exp Zool 187: 249 –253, 1974. 111. Straight AF, Field CM, Mitchison TJ. Anillin binds nonmuscle myosin
89. Oberpriller JO, Oberpriller JC, Arefyeva AM, Mitashov VI, Carlson II and regulates the contractile ring. Mol Biol Cell 16: 193–201, 2005.
BM. Nuclear characteristics of cardiac myocytes following the prolifer- 112. Sucov HM, Dyson E, Gumeringer CL, Price J, Chien KR, Evans RM.
ative response to mincing of the myocardium in the adult newt, Notoph- RXR alpha mutant mice establish a genetic basis for vitamin A signaling
thalmus viridescens. Cell Tissue Res 253: 619 –624, 1988. in heart morphogenesis. Genes Dev 8: 1007–1018, 1994.
90. Pauziene N, Pauza DH, Stropus R. Morphology of human intracardiac 113. Takeuchi T. Regulation of cardiomyocyte proliferation during develop-
nerves: an electron microscope study. J Anat 197: 437–459, 2000. ment and regeneration. Dev Growth Diff 56: 402–409, 2014.
91. Piatkowski T, Muhlfeld C, Borchardt T, Braun T. Reconstitution of 114. Tanaka EM. Regeneration: if they can do it, why can’t we? Cell 113:
the myocardium in regenerating newt hearts is preceded by transient 559 –562, 2003.
deposition of extracellular matrix components. Stem Cells Dev 22: 115. van Amerongen MJ, Engel FB. Features of cardiomyocyte proliferation and
1921–1931, 2013. its potential for cardiac regeneration. J Cell Mol Med 12: 2233–2244, 2008.
92. Piper HM, Jacobson SL, Schwartz P. Determinants of cardiomyocyte 116. VanDusen NJ, Casanovas J, Vincentz JW, Firulli BA, Osterwalder
development in long-term primary culture. J Mol Cell Cardiol 20: M, Lopez-Rios J, Zeller R, Zhou B, Grego-Bessa J, De La Pompa JL,
825–835, 1988. Shou W, Firulli AB. Hand2 is an essential regulator for two notch-
93. Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA, dependent functions within the embryonic endocardium. Cell Rep 9:
Olson EN, Sadek HA. Transient regenerative potential of the neonatal 2071–2083, 2014.
mouse heart. Science 331: 1078 –1080, 2011. 117. Vliegen HW, van der Laarse A, Cornelisse CJ, Eulderink F. Myo-
94. Porrello ER, Mahmoud AI, Simpson E, Johnson BA, Grinsfelder D, cardial changes in pressure overload-induced left ventricular hypertro-
Canseco D, Mammen PP, Rothermel BA, Olson EN, Sadek HA. phy. A study on tissue composition, polyploidization and multinucle-
Regulation of neonatal and adult mammalian heart regeneration by the ation. Eur Heart J 12: 488 –494, 1991.
miR-15 family. Proc Natl Acad Sci USA 110: 187–192, 2013. 118. Wang F, Higgins JM. Histone modifications and mitosis: countermarks,
95. Porrello ER, Olson EN. A neonatal blueprint for cardiac regeneration. landmarks, and bookmarks. Trends Cell Biol 23: 175–184, 2013.
Stem Cell Res 13: 556 –570, 2014. 119. Wang J, Panakova D, Kikuchi K, Holdway JE, Gemberling M,
96. Poss KD. Getting to the heart of regeneration in zebrafish. Semin Cell Burris JS, Singh SP, Dickson AL, Lin YF, Sabeh MK, Werdich AA,
Dev Biol 18: 36 –45, 2007. Yelon D, Macrae CA, Poss KD. The regenerative capacity of zebrafish
97. Poss KD, Wilson LG, Keating MT. Heart regeneration in zebrafish. reverses cardiac failure caused by genetic cardiomyocyte depletion.
Science 298: 2188 –2190, 2002. Development 138: 3421–3430, 2011.
98. Puente BN, Kimura W, Muralidhar SA, Moon J, Amatruda JF, 120. Wessels A, Sedmera D. Developmental anatomy of the heart: a tale of
Phelps KL, Grinsfelder D, Rothermel BA, Chen R, Garcia JA, mice and man. Physiol Genomics 15: 165–176, 2003.
Santos CX, Thet S, Mori E, Kinter MT, Rindler PM, Zacchigna S, 121. Wheatley SP, Carvalho A, Vagnarelli P, Earnshaw WC. INCENP is
Mukherjee S, Chen DJ, Mahmoud AI, Giacca M, Rabinovitch PS, required for proper targeting of Survivin to the centromeres and the
Aroumougame A, Shah AM, Szweda LI, Sadek HA. The oxygen-rich anaphase spindle during mitosis. Curr Biol 11: 886 –890, 2001.
postnatal environment induces cardiomyocyte cell-cycle arrest through 122. Wills AA, Holdway JE, Major RJ, Poss KD. Regulated addition of new
DNA damage response. Cell 157: 565–579, 2014. myocardial and epicardial cells fosters homeostatic cardiac growth and
99. Réaumur RAF. Sur les diverses reproductions qui se font dans les Ecre- maintenance in adult zebrafish. Development 135: 183–192, 2008.
visse, les Omars, les Crabes, etc. et entr’autres sur celles de leurs Jambes et 123. Witman N, Heigwer J, Thaler B, Lui WO, Morrison JI. miR-128
de leurs Ecailles. Memoirs Academic Royal Sciences 223–245, 1712. regulates non-myocyte hyperplasia, deposition of extracellular matrix
100. Rosca MG, Tandler B, Hoppel CL. Mitochondria in cardiac hypertro- and Islet1 expression during newt cardiac regeneration. Dev Biol 383:
phy and heart failure. J Mol Cell Cardiol 55: 31–41, 2013. 253–263, 2013.
101. Sanchez Alvarado A. Regeneration in the metazoans: why does it 124. Witman N, Murtuza B, Davis B, Arner A, Morrison JI. Recapitula-
happen? Bioessays 22: 578 –590, 2000. tion of developmental cardiogenesis governs the morphological and
102. Sander V, Sune G, Jopling C, Morera C, Izpisua Belmonte JC. functional regeneration of adult newt hearts following injury. Dev Biol
Isolation and in vitro culture of primary cardiomyocytes from adult 354: 67–76, 2011.
zebrafish hearts. Nat Protoc 8: 800 –809, 2013. 125. Witman NM, Behm M, Ohman M, Morrison JI. ADAR-related
103. Sdek P, Zhao P, Wang Y, Huang CJ, Ko CY, Butler PC, Weiss JN, activation of adenosine-to-inosine RNA editing during regeneration.
Maclellan WR. Rb and p130 control cell cycle gene silencing to Stem Cells Dev 22: 2254 –2267, 2013.
maintain the postmitotic phenotype in cardiac myocytes. J Cell Biol 194: 126. Yin VP, Lepilina A, Smith A, Poss KD. Regulation of zebrafish heart
407–423, 2011. regeneration by miR-133. Dev Biol 365: 319 –327, 2012.
104. Senyo SE, Lee RT, Kuhn B. Cardiac regeneration based on mechanisms 127. Yu F, Li R, Parks E, Takabe W, Hsiai TK. Electrocardiogram signals
of cardiomyocyte proliferation and differentiation. Stem Cell Res 13: to assess zebrafish heart regeneration: implication of long QT intervals.
532–541, 2014. Ann Biomed Eng 38: 2346 –2357, 2010.
105. Shen H, Cavallero S, Estrada KD, Sandovici I, Kumar SR, Makita T, 128. Zacchigna S, Giacca M. Extra- and intracellular factors regulating cardiomy-
Lien CL, Constancia M, Sucov HM. Extracardiac control of embryonic ocyte proliferation in postnatal life. Cardiovasc Res 102: 312–320, 2014.
cardiomyocyte proliferation and ventricular wall expansion. Cardiovasc 129. Zebrowski DC, Engel FB. The cardiomyocyte cell cycle in hypertrophy,
Res 105: 271–8, 2015. tissue homeostasis, and regeneration. Rev Physiol Biochem Pharmacol
106. Sherman AJ, Klocke FJ, Decker RS, Decker ML, Kozlowski KA, 165: 67–96, 2013.
Harris KR, Hedjbeli S, Yaroshenko Y, Nakamura S, Parker MA, 130. Zebrowski DC, Vergarajauregui S, Wu CC, Piatkowski T, Becker R,
Checchia PA, Evans DB. Myofibrillar disruption in hypocontractile Leone M, Hirth S, Ricciardi F, Falk N, Giessl A, Just S, Braun T,
myocardium showing perfusion-contraction matches and mismatches. Weidinger G, Engel FB. Developmental alterations in centrosome
Am J Physiol Heart Circ Physiol 278: H1320 –H1334, 2000. integrity contribute to the post-mitotic state of mammalian cardiomyo-
107. Soonpaa MH, Kim KK, Pajak L, Franklin M, Field LJ. Cardiomyo- cytes. eLife 4: 2015.
cyte DNA synthesis and binucleation during murine development. Am J 131. Zhao L, Borikova AL, Ben-Yair R, Guner-Ataman B, MacRae CA,
Physiol Heart Circ Physiol 271: H2183–H2189, 1996. Lee RT, Burns CG, Burns CE. Notch signaling regulates cardiomyo-
108. Soonpaa MH, Rubart M, Field LJ. Challenges measuring cardiomyo- cyte proliferation during zebrafish heart regeneration. Proc Natl Acad Sci
cyte renewal. Biochim Biophys Acta 1833: 799 –803, 2013. USA 111: 1403–1408, 2014.
109. Soufan AT, van den Berg G, Ruijter JM, de Boer PA, van den Hoff 132. Zong H, Espinosa JS, Su HH, Muzumdar MD, Luo L. Mosaic
MJ, Moorman AF. Regionalized sequence of myocardial cell growth analysis with double markers in mice. Cell 121: 479 –492, 2005.

AJP-Heart Circ Physiol • doi:10.1152/ajpheart.00559.2015 • www.ajpheart.org


Downloaded from journals.physiology.org/journal/ajpheart (192.167.074.006) on March 28, 2023.

You might also like