Circresaha 119 316303

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Circulation Research

ORIGINAL RESEARCH

Modulation of Mammalian Cardiomyocyte


Cytokinesis by the Extracellular Matrix
Chi-Chung Wu, Sylvia Jeratsch, Johannes Graumann, Didier Y.R. Stainier

RATIONALE: After birth, cycling mammalian CMs (cardiomyocytes) progressively lose the ability to undergo cytokinesis and
hence they become binucleated, which leads to cell cycle exit and loss of regenerative capacity. During late embryonic and
early postnatal heart growth, CM development is accompanied by an expansion of the cardiac fibroblast (cFb) population
and compositional changes in the ECM (extracellular matrix). Whether and how these changes influence cardiomyocyte
cytokinesis is currently unknown.
OBJECTIVE: To elucidate the role of postnatal cFbs and the ECM in cardiomyocyte cytokinesis and identify ECM proteins that
promote cardiomyocyte cytokinesis.
METHODS AND RESULTS: Using primary rat cardiomyocyte cultures, we found that a proportion of postnatal, but not embryonic,
cycling cardiomyocytes fail to progress through cytokinesis and subsequently binucleate, consistent with published reports of
in vitro and in vivo observations. Direct coculture with postnatal cFbs increased cardiomyocyte binucleation, which could be
inhibited by RGD peptide treatment. In contrast, cFb-conditioned medium or transwell coculture did not significantly increase
cardiomyocyte binucleation, suggesting that cFbs inhibit cardiomyocyte cytokinesis through ECM modulation rather than
by secreting diffusible factors. Furthermore, we found that both embryonic and postnatal CMs binucleate at a significantly
higher rate when cultured on postnatal cFb-derived ECM compared with embryonic cFb-derived ECM. These cytokinetic
defects correlate with cardiomyocyte inefficiency in mitotic rounding, a process which is key to successful cytokinesis. To
identify ECM proteins that modulate cardiomyocyte cytokinesis, we compared the composition of embryonic and postnatal
Downloaded from http://ahajournals.org by on July 8, 2022

cFb-derived ECM by mass spectrometry followed by functional assessment. We found that 2 embryonically enriched ECM
proteins, SLIT2 and NPNT (nephronectin), promote cytokinesis of postnatal CMs in vitro and in vivo.
CONCLUSIONS: We identified the postnatal cardiac ECM as a nonpermissive environment for cardiomyocyte cytokinesis
and uncovered novel functions for the embryonic ECM proteins SLIT2 and NPNT (nephronectin) in promoting postnatal
cardiomyocyte cytokinesis.
GRAPHIC ABSTRACT: A graphic abstract is available for this article.

Key Words: cytokinesis ◼ coculture ◼ extracellular matrix ◼ mass spectrometry ◼ myocyte, cardiac

Editorial, see p 908 | In This Issue, see p 851 | Meet the First Author, see p 853

T
he proliferative and regenerative capacity of mam- mouse heart, the extent of cardiomyocyte proliferation
malian CMs (cardiomyocytes) declines with age. and functional recovery after myocardial infarction (MI)
Embryonic cardiomyocytes are highly proliferative1 negatively correlates with the degree of cardiomyocyte
and are able to regenerate after injury.2 Shortly after multinucleation.5 Similar observations have been made in
birth, cycling cardiomyocytes progressively lose the the regenerating zebrafish heart. Adult zebrafish cardio-
ability to complete cytokinesis and become binucle- myocytes are predominantly mononuclear and diploid,6
ated,3 leading to cell cycle exit and loss of regenerative and they can proliferate upon injury.7 However, experi-
capacity.4 Consistent with these findings, in the adult mentally induced cardiomyocyte polyploidization (both

Correspondence to: Chi-Chung Wu, PhD, Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Ludwigstrasse 43, 61231, Bad
Nauheim, Germany, Email chi-chung.wu@mpi-bn.mpg.de; or Didier Y.R. Stainier, PhD, Max Planck Institute for Heart and Lung Research, Department of Developmental
Genetics, Ludwigstrasse 43, 61231, Bad Nauheim, Germany, Email didier.stainier@mpi-bn.mpg.de
The Data Supplement is available with this article at https://www.ahajournals.org/doi/suppl/10.1161/CIRCRESAHA.119.316303.
For Sources of Funding and Disclosures, see page 906.
© 2020 American Heart Association, Inc.
Circulation Research is available at www.ahajournals.org/journal/res

896   September 11, 2020 Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303


Wu et al The ECM Regulates Cardiomyocyte Cytokinesis

Novelty and Significance

Original Research
What Is Known? Mammalian CMs lose the ability to complete cyto-
• Mammalian cardiomyocytes (CMs) lose the ability to kinesis and become binucleated shortly after birth,
complete cytokinesis and become binucleated shortly which correlates with their cell cycle exit and the
after birth. loss of regenerative capacity. The regulatory mecha-
• The degree of multinucleation and polyploidization of nisms of CM cytokinesis, in particular, the role of
mammalian CMs negatively correlates with their prolif- cell nonautonomous factors, is poorly understood.
erative and regenerative capacity. Using primary culture of postnatal CMs, we find that
• cFbs (cardiac fibroblasts) and the cardiac ECM cFbs promote CM binucleation through modulating
(extracellular matrix) influence various aspects of CM the ECM. We find that, unlike the embryonic cFb-
behavior.
derived ECM, the postnatal cFb-derived ECM is a
nonpermissive environment for CM cytokinesis. Fur-
What New Information Does This Article
thermore, we uncover the novel function of 2 embry-
Contribute? onically enriched ECM proteins, SLIT2 and NPNT,
• Cardiac fibroblasts induce CM binucleation through in promoting CM cytokinesis both in vitro and in
modulation of the ECM.
vivo. These findings highlight the importance of the
• Postnatal cFb-derived ECM is a nonpermissive envi-
nonmyocyte population and the ECM in modulating
ronment for CM cytokinesis.
• Two embryonically enriched ECM proteins, SLIT2 and CM cytokinesis, which may provide new therapeutic
NPNT (nephronectin), are able to function as soluble fac- targets to augment adult CM proliferation after myo-
tors to promote CM cytokinesis both in vitro and in vivo. cardial infarction.

hormone signaling,18 p38 MAPK,19,20 ECT2,21 TNNI3K,5


Nonstandard Abbreviations and Acronyms cell cycle genes,22 and hypoxia.23,24 However, little is
known about whether and how nonmyocyte cardiac cells
cFb cardiac fibroblast and the cardiac ECM (extracellular matrix) modulate car-
Downloaded from http://ahajournals.org by on July 8, 2022

ECM extracellular matrix diomyocyte cytokinesis. Fibroblasts constitute ≈20% of


MI myocardial infarction the nonmyocyte population in the mammalian heart25
NPNT nephronectin and play important functions during heart development,
homeostasis, and disease.26 The number of cFbs (car-
diac fibroblasts) increases throughout development,27
multinucleation and increase in nuclear ploidy) in the and it continues to expand after birth with peak prolif-
zebrafish heart impairs its proliferative and regenerative eration at around postnatal day 4 (P4),28 coinciding with
capacity.8 These observations suggest that ploidy is a the onset of cardiomyocyte binucleation.3 Similar to cFbs,
critical factor in determining the degree of endogenous the ECM also plays important roles in the heart.29 The
cardiomyocyte proliferation and regeneration. However, ECM is a dynamic and complex protein network whose
the regulatory mechanisms of cardiomyocyte cytoki- composition30 and complexity31 changes as the heart
nesis are poorly understood. Similar to their postnatal grows. Both cFbs and the ECM are known to interact
counterpart, adult mammalian cardiomyocytes are also with cardiomyocytes and influence their behavior. Embry-
prone to cytokinetic defects. Reactivation of cell cycle onic cFbs have been reported to promote cardiomyocyte
activity in adult cardiomyocytes following MI9 or various proliferation via paracrine signaling in contrast to the
experimental or therapeutic approaches often results in hypertrophy-inducing adult cFbs.27 Several studies have
karyokinesis but not cytokinesis,10,11 which could impact been conducted on ECM proteins in the heart including
the potential of stimulating adult cardiomyogenesis via PERIOSTIN, which has been shown to stimulate cardio-
cardiomyocyte proliferation as a therapeutic approach myocyte proliferation and cardiac regeneration after MI
for MI.10 A thorough understanding of cardiomyocyte cell in neonates32 and adults.33 AGRIN, another ECM protein,
cycle control, in particular of the factors modulating car- whose expression in the heart decreases from P1 to P7,
diomyocyte cytokinesis, is of great interest to the cardiac also promotes CM proliferation and regeneration after
development and regeneration fields. MI.34 Despite their broad roles in CM behavior and car-
In the past 2 decades, a number of factors and sig- diac function, whether and how cFbs and the ECM influ-
naling pathways have been identified as modulators ence CM cytokinesis remains unknown.
of the cardiomyocyte cell cycle including MEIS1,12 the We addressed these questions using primary rat car-
Hippo pathway,13–15 NRG1/ERBB2 signaling,16,17 thyroid diomyocyte cultures. We first show that postnatal cFbs

Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303 September 11, 2020   897


Wu et al The ECM Regulates Cardiomyocyte Cytokinesis

induce cardiomyocyte binucleation through the ECM Percoll gradient were preplated for 30 minutes in complete
rather than through diffusible factors. Using a cell-derived medium. After removal of the unattached cells, cFbs were
Original Research

matrix approach, we further show that the postnatal ECM cultured in complete medium. To improve purity, embryonic
is a nonpermissive environment for cardiomyocyte mitotic cFbs were passaged for 2× to 3× until no beating cells were
observed in the dish. cFbs were then expanded and frozen
rounding and cytokinesis, leading to cardiomyocyte binu-
down until use.
cleation. By comparing the composition of embryonic
and postnatal cFb-derived ECM, we identify 2 ECM pro-
teins, SLIT2 and NPNT (nephronectin), which we show Cardiomyocyte cFb Cocultures
are capable of promoting cardiomyocyte cytokinesis both For the direct cocultures, isolated cardiomyocytes and cFbs
in vitro and in vivo. Our results reveal previously unap- were resuspended in neonatal cardiomyocyte medium con-
taining 5% horse serum and 0.02 mmol/L ara-C and mixed
preciated roles for cFbs and the ECM in regulating mam-
in 4:1 or 2:1 ratio (total cell number=1.2×105 cells/cm2), fol-
malian cardiomyocyte cytokinesis.
lowed by the binucleation assay. For the transwell cocultures,
isolated cardiomyocytes were seeded on a cover glass coated
with 1% gelatin in a 24 well plate. Forty thousand or 80 000
METHODS cFbs were plated on a Transwell permeable insert with a poly-
The data that support the findings of this study are available ester membrane of 0.4 μm pore size (Corning), followed by the
from the corresponding authors upon reasonable request. binucleation assay. For the cFb-conditioned medium experi-
Other methods, sample sizes and the Major Resources Table ments, isolated P3 cFbs were cultured with complete medium
can be found in the Data Supplement. for 4 days. The collected medium was filtered (0.2 μm) and
stored at −20°C until use. For the binucleation assay, cardio-
myocytes were cultured in neonatal cardiomyocyte medium
Animal Procedures with 10% FBS (ctrl) or 50% cFb-conditioned medium (1:1
Sprague Dawley rats (Rattus norvegicus) and C57/Bl6 mice mix with neonatal cardiomyocyte medium with 10% FBS) or
(Mus musculus) were housed at the Max Planck Institute for 100% cFb-conditioned medium.
Heart and Lung Research according to § 11 TierSchG and EU
Directive 2010/63/EU on the protection of animals used for
scientific purposes. Preparation of cFb-Derived ECM
cFb-derived ECM was generated as previously described.36
E16 and P3 cFbs (6.25×104 cells/cm2) were plated in com-
Rat Cell Isolation and Culture plete medium on a cover glass coated with 0.1% gelatin cross-
Downloaded from http://ahajournals.org by on July 8, 2022

To isolate ventricular cardiomyocytes from rats, pooled linked with 1% glutaraldehyde. cFbs were cultured for 11 days
ventricles from E16 embryos or P3 pups of the same litter in complete medium with ascorbic acid (50 μg/mL), which was
were dissociated using the Neonatal heart dissociation kit exchanged every 48 hours. For decellularization, an extraction
(Miltenyi Biotec). Hence, N (number of replicates) indicates buffer (DPBS with 0.5% (v/v) Triton X-100 and 20 mmol/L
the number of independent cardiomyocyte isolations. After NH4OH) was added and incubated for 5 minutes at 37°C. After
dissociation, cardiomyocytes were purified using a 2-step washing, DNA debris were removed by DNase I treatment (10
discontinuous Percoll gradient as previously described.35 U/mL, Roche) for 45 minutes at 37°C. The resulting matrix was
Cardiomyocytes (1×105 cells/cm2 for E16 cardiomyocytes washed with DPBS and stored at 4°C until use.
and 1.2×105 cells/cm2 for P3 cardiomyocytes) were plated To generate a protein mixture of cFb-derived ECM, a solu-
on a cover glass coated with 1% gelatin and cultured in neo- bilization reagent (5 mol/L guanidine containing 10 mmol/L
natal cardiomyocyte medium (DMEM/F12, GlutaMAX with dithiothreitol) was added to the matrix for 5 minutes on ice.
3 mmol/L Na-Pyruvate, 0.1 mmol/L Ascorbic acid, Insulin/ The ECM was scraped off the dish with a cell scraper and kept
transferrin/Na-Selenite solution [1:200], 0.2% BSA and 1× in the solubilization reagent for 1 hour at 4°C. Afterward, the
Pen/Strep) containing 5% horse serum and 0.02 mmol/L solubilized matrix (supernatant) was collected by centrifuga-
ara-C at 37°C and 5% CO2 for 48 hours. To perform the tion (15 minutes, 12 000×g) and stored at 4°C until use. Cover
binucleation assay, unless otherwise specified, cardiomyo- glass was coated with 10 μg/mL protein mix for at least 1
cytes were washed twice and cultured in neonatal cardiomyo- hour at 37°C.
cyte medium with 10% fetal bovine serum (FBS, HyClone)
for 24 hours. 0.02 mmol/L EdU was added to the medium
for 24 hours. Cells were washed twice to remove the EdU RGD Peptide, ITGß1 Blocking Antibody, and
and cultured for another 24 hours in neonatal cardiomyocyte ECM Protein Treatments
medium with 10% FBS until harvesting. The RGD peptide (Gly-Arg-Gly-Asp-Ser-Pro-Lys, Sigma) was
To isolate ventricular cFbs, ventricles were isolated, dis- added to cardiomyocytes at 0.1 and 0.2 mmol/L. Water was
sociated, and cells were purified as described above. For P3 used as a control. Hamster anti-Rat ITGß1 antibody (#555002,
hearts, cFbs collected after the Percoll gradient were preplated BD Pharmingen) or Hamster IgM control (#553957, BD
in uncoated Petri dishes in complete medium (DMEM/F12, Pharmingen) was added to cardiomyocytes at 40 or 80 μg/
GlutaMAX, 1× Pen/Strep supplemented with 10% FBS) at mL. The following recombinant ECM proteins were added to
37°C for 1 hour. After removal of the unattached cells, cFbs cardiomyocytes at 0.5 or 1 μg/mL: mouse NPNT (#4298-NP-
were cultured in complete medium. Fresh cFbs were isolated 050), mouse SLIT2 (#5444-SL-050), mouse NID2 (#6760-
for each experiment. For E16 hearts, cFbs obtained after the ND-050), mouse SPON1 (#7950-SP-050), mouse TGM2

898   September 11, 2020 Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303


Wu et al The ECM Regulates Cardiomyocyte Cytokinesis

(#5418-TG-010), human FN1 (#ACFP4305B; R&D Systems), further cultured for another 24 hours (Figure 1A). The
and human LAMA5 (#ab42326; Abcam). nucleation status of EdU+ cardiomyocytes was then

Original Research
assessed by immunostaining for cardiac troponin I (car-
Cardiomyocyte Cell Shape Measurements diomyocyte), N-cadherin (cardiomyocyte junctions), and
Mitotic cardiomyocytes at metaphase or anaphase (PH3+ and EdU at 24 and 48 hours after EdU addition (Figure 1B).
cTnI+) throughout the sample were randomly selected and As the cell cycle length of postnatal mammalian cardio-
imaged. Cell boundaries of mitotic and neighboring nonmitotic myocytes has been estimated to be around 35 hours,41
cardiomyocytes were manually outlined for aspect ratio mea- this regimen allows one to assess the degree of cyto-
surements using Fiji. kinesis completion in EdU+ cardiomyocytes within one
cell cycle.
SLIT2 and NPNT Injections Using this assay, we found that the percentage of
About 1.5 μg of recombinant SLIT2 or NPNT protein (R&D binuclear cells within the EdU+ cardiomyocyte population
systems, see above) (PBS as a control) was injected daily increased significantly from 24 to 48 hours for P3 car-
into wild-type C57/Bl6 pups from P1 to P4 while 50 mg/ diomyocytes, indicative of cytokinesis failure (Figure 1C),
kg EdU was injected once on P3. Ventricles were harvested and an observation consistent with previous reports.19,40
on P5, and cells were dissociated using 2 different fixation- In contrast, no significant difference in cytokinesis was
digestion protocols. Detailed cell dissociation protocols can be observed using embryonic day 16 (E16) cardiomyocytes
found in the Data Supplement. Dissociated cells were spotted (Figure 1C). Next, we repeated the above experiment with
on a glass slide and air-dried, which was followed by EdU and P3 cardiomyocytes under serum-free condition to inhibit
alpha-actinin immunostaining. At least 100 EdU+ cardiomyo-
cell cycle activity. Serum starvation did not induce signifi-
cytes were analyzed per sample, blindly. At least 3 crosses were
set up for each round of experiments. Litters were randomly
cant apoptosis in cardiomyocytes (Online Figure IA), and
assigned to receive PBS, SLIT2, or NPNT; hence experiments we found no significant increase in the percentage of total
were not performed with littermate controls. No sex determina- binuclear cardiomyocytes from 24 to 48 hours (Online Fig-
tion was performed and hence both sexes are assumed to have ure IB), suggesting that the observed increase in binuclear
been used in an unknown ratio. P3 cardiomyocytes is cell cycle-dependent. Hereafter, the
degree of binucleation (ie, binucleation index) is calculated
as the relative increase in the percentage of binuclear cells
RESULTS within the EdU+ cardiomyocyte population from 24 to 48
hours (Figure 1D). To further validate this approach, we
Downloaded from http://ahajournals.org by on July 8, 2022

Establishing an EdU Pulse-Chase monitored the cell cycle behavior of E16 and P3 cardiomy-
Cardiomyocyte Binucleation Assay ocytes by confocal time-lapse imaging with live tubulin and
During heart development, changes in the quantity and DNA labeling (Online Movie I). We imaged >500 mitotic
properties (eg, transcriptome) of various cardiac cell events in total (195 for E16 and 372 for P3 cardiomyo-
types, including CMs37 and Fbs,27,28 as well as in the cytes) and found that the majority of the mitotic P3 (58%)
composition and complexity of the ECM,30,31 occur con- and a small minority of the mitotic E16 (4.6%) cardiomyo-
comitantly. It is therefore difficult to decipher their indi- cytes underwent binucleation (Figure 1E), consistent with
vidual influence on cardiomyocyte cytokinesis in vivo. To previous results (Figure 1C). Altogether, these data show
circumvent this complexity, we used primary rat cardio- that the binucleation assay developed here is a reliable
myocyte cultures as a model as they allow one to test readout of cardiomyocyte cytokinesis.
individual variables (eg, culturing cardiomyocytes of a
particular stage with different types of cFbs and ECM). Postnatal Cardiac Fibroblasts Lead to an
Detection of cardiomyocyte cytokinesis is conven-
tionally carried out by immunostaining or lineage tracing.
Increase in Cardiomyocyte Binucleation
However, immunostaining for commonly used cytokinetic Through the Extracellular Matrix
markers like Aurora B-kinase has been shown to mark To investigate the role of cFbs in cardiomyocyte cytokinesis,
both dividing and binucleating cardiomyocytes.38 While we performed binucleation assays on isolated P3 cardio-
lineage tracing-based systems including mosaic analy- myocytes cocultured with increasing amounts of P3 cFbs
sis with double markers39 or live-cell imaging40 are more (Figure 2A). We found that the degree of binucleation posi-
reliable, they are not suitable for routine use because of tively correlated with cFb number (Figure 2B), suggesting
their low efficiency or throughput, respectively. Hence, that postnatal cFbs promote cardiomyocyte binucleation.
we first set out to establish a simple and robust cardio- Communication between cFbs and cardiomyocytes can
myocyte binucleation assay. Isolated rat cardiomyocytes be mediated through diffusible factors, the ECM or direct
from various pre- and postnatal stages were cultured cell-cell contact.42 To investigate whether diffusible fac-
with 10% FBS to stimulate cell cycle activity. To mark tors from cFbs could modulate cardiomyocyte binucleation,
the cycling population, EdU was added to the medium we performed 2 types of indirect coculture experiments.
for 24 hours and after washing out, cardiomyocytes were cFbs were cocultured with cardiomyocytes using Transwell

Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303 September 11, 2020   899


Wu et al The ECM Regulates Cardiomyocyte Cytokinesis
Original Research

Figure 1. Postnatal rat cardiomyocytes binucleate in vitro.


A, Schematic of the CMs (cardiomyocytes) binucleation assay. Isolated CMs are cultured with 10% FBS and treated with EdU for 24 h. EdU
Downloaded from http://ahajournals.org by on July 8, 2022

is then washed out, and CMs are cultured for another 24 h to complete one cell cycle. B, Nucleation status of EdU+ CMs was assessed by
immunostaining for cTnI (cardiac troponin I), EdU, and N-cadherin (N-cad). C, Quantification of CM binucleation. The percentage of binuclear cells
within the EdU+ CM population increases significantly from 24 to 48 h in postnatal (P3) but not embryonic (E16) CM cultures. D, Binucleation
index, defined as the relative increase in the percentage of binuclear cells within the EdU+ CM population from 24 to 48 h, calculated from (C).
E, Cell cycle analysis of E16 and P3 CMs. Cardiomyocyte cell cycle was monitored by confocal time-lapse imaging with live tubulin and DNA
labeling. A majority of the mitotic P3 but not E16 CMs underwent binucleation. Endoreplication denotes failed karyokinesis. n=number of mitotic
CMs imaged. Values represent mean±SEM. (C, D) P values determined by unpaired t-test; (E) P values determined by χ2 test.

inserts with a pore size of 0.4 μm, which physically sepa- cardiomyocyte:cFb cocultures compared with control
rates the cells but allows diffusible factors secreted from (Online Figure IIB). To test whether ECM proteins could
cFbs to reach cardiomyocytes. No significant difference in promote cardiomyocyte binucleation, we decreased
the binucleation index was observed in cocultures of cardio- cardiomyocyte-ECM interactions with the RGD peptide,
myocytes with 40 000 or 80 000 cFbs compared with con- which mimics the RGD motif on a subset of ECM pro-
trol (no extra cFbs; Figure 2C). Consistent with this result, teins.44 Treating 2:1 cardiomyocyte:cFb cocultures with
conditioned medium from P3 cFbs also did not increase the RGD peptide from 24 to 48 hours reduced the car-
cardiomyocyte binucleation (Online Figure IIA). These data diomyocyte binucleation index in a dose-dependent man-
suggest that diffusible factors are not mediating the effects ner (Figure 2D). Altogether, although we cannot rule out
of postnatal cFbs on cardiomyocyte cytokinesis. the effect of cell-cell contacts, these data suggest that
cFbs contribute in a major way to the production, cFbs increase cardiomyocyte binucleation through ECM
secretion, and maintenance of the cardiac ECM, and modulation rather than by secreting diffusible factors.
ECM proteins have been shown to promote cardio-
myocyte proliferation27,34 and hypertrophic growth.43 Postnatal Cardiac Extracellular Matrix
We therefore hypothesized that postnatal cFbs pro-
mote cardiomyocyte binucleation by modulating the
Hinders Cardiomyocyte Mitotic Rounding and
microenvironment (ie, ECM protein expression and/or Cytokinesis
assembly). In line with this hypothesis, we observed by To further investigate the role of the ECM in cardio-
immunostaining increased expression and assembly into myocyte cytokinesis, we employed a cell-derived matrix
fibrillar structures of FN (fibronectin) and COL 1 in 2:1 approach,36 which better mimics aspects of native tissue

900   September 11, 2020 Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303


Wu et al The ECM Regulates Cardiomyocyte Cytokinesis

Original Research
Figure 2. Postnatal cardiac
fibroblasts increase cardiomyocyte
binucleation through the
extracellular matrix.
A, Direct cocultures of P3 cFbs (cardiac
fibroblasts) and cardiomyocytes (CMs).
Examples of CM (cTnI+) and cFb
(Vimentin+) cocultures shown on the
left. Percentage of cFbs in different
coculture conditions shown on the
right. B, Binucleation index in P3 CMs
cocultured with increasing amounts of
cFbs. C, Transwell cocultures of P3 cFbs
and CMs. Diffusible factors from cFbs do
not increase the CM binucleation index
compared with control. D, RGD peptide
treatment from 24 to 48 h decreases CM
binucleation (2:1 CM:cFb cocultures).
Values represent mean±SEM. P values
determined by 1-way ANOVA followed
by Tukey’s HSD test (P values of overall
ANOVA and post hoc tests indicated
between the parentheses and above the
lines, respectively).
Downloaded from http://ahajournals.org by on July 8, 2022

microenvironments than conventional monolayer cul- on postnatal ECM with 40 or 80 μg/mL of Integrin ß1
tures.45 In brief, isolated E16 and P3 cFbs were cultured (ITGß1) blocking antibody, which decreases cardiomyo-
for 11 days during which ECM proteins were secreted cyte-ECM interactions47 and found that it reduced the
and deposited on the culture surface. After decellular- binucleation index compared with IgM control (Figure 3C,
ization, the native-like ECM was used for cardiomyocyte Online Figure IIIB). These data further support our find-
cultures (Figure 3A; Online Figure IIIA). Binucleation ings that the ECM modulates cardiomyocyte cytokinesis.
assays were performed on E16 and P3 cardiomyocytes Cells undergo a series of cell shape changes dur-
cultured on embryonic and postnatal ECM. We observed ing mitosis including mitotic rounding, and these
that both E16 (compare columns 1 and 2) and P3 (com- changes depend on the remodeling of cell-substrate
pare columns 3 and 4) cardiomyocytes binucleated at a adhesion and are critical for successful cytokinesis.48
significantly higher rate on postnatal ECM (Figure 3B), During early mitosis, postnatal cardiomyocytes retain
suggesting that, compared with the embryonic ECM, the a flattened morphology with prominent sarcomeric
postnatal ECM is a nonpermissive environment for car- structures. From the metaphase stage onward, cardio-
diomyocyte cytokinesis (Figure 3B). In addition, E16 car- myocytes partially lose contact with neighboring cells
diomyocytes consistently binucleated to a lesser extent and the substrate, and they start rounding up (Online
than P3 cardiomyocytes when cultured on the same type Figure IIIC). To investigate the effects of the ECM on
of ECM (compare columns 1 and 3 for E16 ECM, 2 and postnatal cardiomyocyte mitotic rounding, we quanti-
4 for P3 ECM; Figure 3B). Together, these data sug- fied the aspect ratio of mitotic and neighboring non-
gest that both cell-autonomous (ie, intrinsic to the car- mitotic cardiomyocytes. While nonmitotic (PH3−)
diomyocytes) and cell-nonautonomous factors (ie, the cardiomyocytes exhibited a similar aspect ratio on both
ECM) modulate cardiomyocyte cytokinesis. Integrins are embryonic and postnatal ECM, mitotic (PH3+) cardio-
the major receptors that link the ECM to the cytoskel- myocytes reduced their aspect ratio (ie, rounded up) on
eton and they activate a number of intracellular signal- embryonic ECM but failed to do so on postnatal ECM
ing pathways.46 We treated P3 cardiomyocytes cultured (Figure 3D). Moreover, treating P3 cardiomyocytes on

Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303 September 11, 2020   901


Wu et al The ECM Regulates Cardiomyocyte Cytokinesis
Original Research

Figure 3. Postnatal cardiac


extracellular matrix increases
cardiomyocyte binucleation through
inefficient mitotic rounding.
A, Schematic of the production of cardiac
fibroblast (cFb)-derived ECM (extracellular
matrix). Native-like ECM was produced
by culturing cFbs for 11 d followed by
decellularization. B, Cardiomyocyte (CM)
binucleation index on embryonic and
postnatal ECM. Binucleation assay was
performed on CMs cultured on embryonic
or postnatal ECM. Both E16 and P3 CMs
binucleate at a significantly higher rate
on postnatal ECM. C, Integrin ß1 (ITGß1)
blocking antibody treatment from 24 to
48 h partially prevents CM binucleation.
D, CM mitotic rounding defects. Aspect
ratio of mitotic (PH3+, yellow dashed
line) and neighboring nonmitotic P3 CMs
was determined. Mitotic CMs reduce
their aspect ratio (ie, undergo mitotic
rounding) more efficiently on embryonic
compared with postnatal ECM. E, Integrin
ß1 blocking antibody treatment from 24
to 48 h increases CM mitotic rounding.
n=number of CMs analyzed. Values
represent mean±SEM. (B: E16 CMs, C) P
values determined by unpaired t-test; (B:
Downloaded from http://ahajournals.org by on July 8, 2022

P3 CMs, E) P values determined by Mann-


Whitney U test; (D) P values determined by
Kruskal-Wallis test followed by Dunn test
(P values of overall ANOVA and post hoc
tests indicated between the parentheses
and above the lines, respectively).

postnatal ECM with 80 μg/mL of ITGß1 blocking anti- with intact ECM (Figure 3B), P3 cardiomyocytes binu-
body, which promotes cytokinesis (Online Figure IIIB), cleated at a significantly higher rate on postnatal-ECM
rescued their mitotic rounding defects (Figure 3E). coated surfaces than on embryonic-ECM coated sur-
These data suggest that the cardiomyocyte cytokinetic faces (Online Figure IVA).
defects on postnatal cFb-derived ECM can be at least To identify ECM proteins that modulate cardiomyocyte
partly attributed to the perturbation of mitotic rounding. cytokinesis, we compared the composition of the embry-
onic and postnatal cFb-derived ECM by mass spectrome-
try-based quantitative proteomics. We focused on proteins
NPNT and SLIT2 Promote Mitotic Rounding and enriched in the embryonic ECM as it promotes cardio-
Cytokinesis of Postnatal Cardiomyocytes myocyte mitotic rounding and cytokinesis. Out of the 270
Next, we generated protein mixtures of embryonic and proteins that were enriched in the embryonic ECM (Log2
postnatal cFb-derived ECM through solubilization and fold change <1, false discovery rate <0.05; Online Figure
used them to coat the tissue culture surface before IVB), 44 are categorized as ECM or ECM-associated pro-
seeding cardiomyocytes. This treatment eliminates the teins49 (Figure 4A). We then analyzed the gene expression
original 3D organization/arrangement of the ECM and level for each of these proteins using a published data-
allows one to test whether compositional differences set50 and selected for further analysis 7 candidates whose
between two types of ECM alone are sufficient to affect expression went down in the mouse heart within the first
cardiomyocyte cytokinesis. Similar to our observations postnatal week, that is, when cardiomyocyte binucleation

902   September 11, 2020 Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303


Wu et al The ECM Regulates Cardiomyocyte Cytokinesis

begins (Figure 4A). Next, we assessed the ability of these role in cardiomyocyte cell cycle remains unaddressed. To
candidate proteins to promote cardiomyocyte cytokinesis test whether SLIT2 and NPNT are mitogenic, we assessed

Original Research
in vitro. We performed binucleation assays using P3 car- cardiomyocyte cell cycle activity by EdU incorporation upon
diomyocytes cultured on postnatal ECM and added recom- SLIT2 and NPNT treatment. No significant difference in cell
binant proteins at 2 concentrations (0.5 and 1 μg/mL) cycle activity was observed in starved P3 cardiomyocytes
right after EdU removal (Figure 4B). Of the 7 candidate treated with SLIT2 or NPNT compared with control (Fig-
proteins tested, SLIT2 and NPNT consistently promoted ure 4C). As the embryonic ECM supports cardiomyocyte
cardiomyocyte cytokinesis (Figure 4B). While SLIT251 and mitotic rounding (Figure 3D) and subsequent cytokinesis
NPNT52 have been implicated in heart development, their (Figure 3B), we tested whether SLIT2 and/or NPNT could
Downloaded from http://ahajournals.org by on July 8, 2022

Figure 4. NPNT and SLIT2 promote mitotic rounding and cytokinesis of postnatal cardiomyocytes in vitro.
A, Workflow to identify ECM (extracellular matrix) proteins that modulate cardiomyocyte (CM) cytokinesis. The composition of the embryonic
and postnatal cardiac fibroblast (cFb)-derived ECM was compared by quantitative mass spectrometry-based proteomics. Seven embryonic ECM
proteins, whose gene expression went down in P7 hearts compared with P1, were selected for further analysis. B, Functional assessment of
embryonic ECM proteins. Binucleation assay was performed on P3 CMs cultured on postnatal ECM and recombinant ECM proteins were added
after EdU removal. NPNT and SLIT2 consistently promoted CM cytokinesis. C, NPNT and SLIT2 are not mitogenic. Treating starved P3 CMs with
NPNT or SLIT2 failed to promote cell cycle entry, as assessed by EdU incorporation. D, NPNT and SLIT2 promote mitotic rounding. Aspect ratio
of mitotic P3 CMs cultured on postnatal ECM. Treatment with NPNT or SLIT2 from 24 to 48 h promotes mitotic rounding. n=number of CMs
analyzed. Values represent mean±SEM. (B, C) P values determined by 1-way ANOVA followed by Tukey’s HSD test; (B) P values of all pairwise
comparisons shown in Online Table II; (D) P values determined by Kruskal-Wallis test followed by Dunn test (P values of overall ANOVA and post
hoc tests indicated between the parentheses and above the lines, respectively).

Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303 September 11, 2020   903


Wu et al The ECM Regulates Cardiomyocyte Cytokinesis

rescue the mitotic rounding defects of P3 cardiomyocytes. longstanding question in the field. While several cell-auton-
We treated P3 cardiomyocytes cultured on postnatal ECM omous factors have been implicated in cardiomyocyte
Original Research

with SLIT2 and NPNT for 24 hours and determined the cytokinesis, much less is known regarding the role of non-
aspect ratio of mitotic cardiomyocytes. Compared with myocyte cardiac cells and the cardiac ECM in this process.
controls (PBS and NID2, a candidate ECM protein that Although originally considered as a bystander cell type
failed to promote cardiomyocyte cytokinesis [Figure 4B]), providing structural support to the heart, a growing body of
SLIT2 and NPNT promoted mitotic rounding (Figure 4D). evidence has revealed a diverse role for cFbs during heart
Next, we employed an approach similar to our binu- development, homeostasis, and disease.26 Global genetic
cleation assay (Figure 1A) to test the role of SLIT2 and inactivation of Tcf21, a transcription factor gene essential
NPNT in vivo. SLIT2 and NPNT recombinant proteins were for the formation of cFbs, leads to the loss of cFbs in the
injected daily into wild-type mouse pups from P1 to P4. The embryonic mouse heart and hypoplastic ventricular cham-
cycling cardiomyocyte population was marked by a single bers.53 Notably, cFbs can influence cardiomyocyte behav-
injection of EdU on P3 (Figure 5A). Cell cycle entry and ior in an age-dependent manner. In an in vitro coculture
nucleation status were subsequently determined on disso- system, embryonic cFbs reportedly induce cardiomyocyte
ciated P5 cardiomyocytes. Consistent with previous results proliferation while adult cFbs induce cardiomyocyte hyper-
(Figure 4C), the percentage of EdU+ cardiomyocytes was trophy.27 In 3D engineered cardiac tissue, adult cFbs sig-
comparable between all groups (Figure 5A), suggesting nificantly worsen both electrical and mechanical functions
that cardiomyocyte cell cycle entry was not significantly of the cocultured cardiomyocytes compared with embryonic
affected by SLIT2 or NPNT. On the other hand, compared cFbs.54 The role of cFbs during early postnatal heart devel-
with PBS-injected controls, both SLIT2 and NPNT sig- opment has also been the focus of recent studies. While
nificantly increased the percentage of mononuclear cells peak proliferation of cFbs in the postnatal heart coincides
within the EdU+ cardiomyocyte population (PBS: 22.3%, with the onset of cardiomyocyte binucleation,28 evidence for
SLIT2: 41% and NPNT: 40.3%, Figure 5B), suggesting a their role in cardiomyocyte cytokinesis regulation has been
higher degree of successful cytokinesis. lacking. Through direct coculture experiments, we found
In summary, we have found that mammalian car- that cFbs increase cardiomyocyte binucleation in a dose-
diomyocyte cytokinesis is modulated by the postnatal dependent manner (Figure 2B). Although cell density was
cardiac ECM, a nonpermissive environment for cardio- kept constant among the different conditions during cell
myocyte cytokinesis. Moreover, through quantitative pro- seeding, because of the high proliferative rate of cFbs, it
teomics analysis followed by candidate screening, we
Downloaded from http://ahajournals.org by on July 8, 2022

is possible that the 4:1 and 2:1 cardiomyocyte:cFb cocul-


uncovered novel functions for the embryonic cardiac tures have a higher cell density by the end of the experi-
ECM proteins SLIT2 and NPNT in promoting cardiomyo- ment (ie, 96 hours after seeding). And although we cannot
cyte mitotic rounding and cytokinesis. rule out a role for cell density in cardiomyocyte binucle-
ation, the effect of cFbs is mediated at least in part through
RGD-motif containing extracellular proteins as RGD pep-
DISCUSSION tide treatment decreased the binucleation index in the 2:1
Cell cycle control of mammalian cardiomyocytes has been cardiomyocyte:cFb cocultures (Figure 2D). Consistent with
extensively investigated in the past decades. Recent stud- our in vitro data, genetic ablation of cFbs through overex-
ies in mouse5 and zebrafish8 have suggested polyploidy pression of diphtheria toxin A in PDGFRα+ cells in P1 mice
as a critical factor limiting the endogenous proliferative promotes cardiomyocyte cell cycle activity as well as the
and regenerative potential of cardiomyocytes. How post- degree of successful cytokinesis at P5 (Michelle Tallquist,
natal cardiomyocyte cytokinesis is regulated has been a personal communication, 2019). Altogether, these results

Figure 5. NPNT and SLIT2 promote


cytokinesis of postnatal cardiomyocytes
in vivo.
A, Injection of NPNT or SLIT2 recombinant
proteins from P1 to P4 did not significantly alter
the percentage of EdU+ CMs (cardiomyocytes).
B, NPNT and SLIT2 injections significantly
increased the percentage of mononuclear cells
within the EdU+ CM population. N=number of
hearts; n=number of CMs (A) or EdU+ CMs (B)
analyzed. Values represent mean±SEM. P values
determined by 1-way ANOVA followed by Tukey
HSD test (P values of overall ANOVA and post
hoc tests indicated between the parentheses and
above the lines, respectively).

904   September 11, 2020 Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303


Wu et al The ECM Regulates Cardiomyocyte Cytokinesis

reveal a novel role for cFbs as modulators of cardiomyocyte provides a robust and reliable readout for cardiomyocyte
cytokinesis. Recently, multiple adult cFb populations with cytokinesis and is suitable for medium to high throughput

Original Research
distinctive transcriptional signatures have been reported screens of cardiomyocyte cytokinesis-promoting factors.
from a single-cell RNA sequencing study in mouse.55 While FN, COL 3,27 and AGRIN34 have been implicated
Whether such cFb heterogeneity also exists in the early in cardiomyocyte proliferation, ECM proteins that specifi-
postnatal heart and whether different cFb populations have cally modulate cardiomyocyte cytokinesis have not been
specific roles in controlling cardiomyocyte cytokinesis are reported thus far. In this study, we found that SLIT2 and
important questions to pursue in the future. NPNT promote mitotic rounding (Figure 4D) and cytokine-
Embryonic to postnatal heart development is char- sis (Figures 4B and 5B) but not cell cycle entry of postnatal
acterized by concurrent changes intrinsic to cardiomyo- cardiomyocytes (Figures 4C and 5A). While both SLIT251
cytes (eg, sarcomeric protein composition and structure, and NPNT52 have been implicated in heart development, it
metabolic shift from glycolysis to fatty acid oxidation),56 is unclear how they interact with cardiomyocytes to regu-
as well as in the microenvironment (eg, composition and late their cytokinesis. Since acute treatment (24 hours) with
complexity of the ECM).30,31,34 It is therefore challenging SLIT2 or NPNT are sufficient to promote cardiomyocyte
to determine their respective role in the cardiomyocyte mitotic rounding (Figure 4D) and cytokinesis (Figure 4B),
transition from cytokinesis to binucleation in vivo. We took we postulate that they may act directly on cardiomyocytes
advantage of an in vitro system to address this question. and activate intracellular signaling pathways critical for suc-
We found that the postnatal ECM is a nonpermissive envi- cessful cytokinesis. In line with this hypothesis, SLIT2 has
ronment for cardiomyocyte cytokinesis irrespective of the been shown to activate RhoA, a small GTPase essential
developmental stage of cardiomyocytes (Figure 3B). On for cytokinesis,61 by activating Trio GEF62 and/or suppress-
the other hand, P3 cardiomyocytes consistently exhibited ing the Myo9b RhoGAP activity.63 Analyzing the changes in
a higher degree of binucleation than E16 cardiomyocytes cytoskeletal organization and the activity of its regulators in
when cultured on the same substrate, that is, gelatin (Fig- cardiomyocytes upon SLIT2 and NPNT treatment will shed
ure 1D) or embryonic or postnatal ECM (Figure 3B). Of all light on the molecular mechanisms underlying the effects
the conditions tested, the highest binucleation index was of SLIT2 and NPNT on cardiomyocyte cytokinesis.
observed when combining P3 cardiomyocytes with post-
natal ECM (Figure 3B). Altogether, these findings suggest
that both cardiomyocyte maturity and the ECM modulate ARTICLE INFORMATION
cardiomyocyte cytokinesis and that they may synergisti-
Downloaded from http://ahajournals.org by on July 8, 2022

Received November 4, 2019; revision received May 28, 2020; accepted June
cally induce cardiomyocyte binucleation. As ploidy was 19, 2020.
not assessed in the current study, whether cFbs and/ Affiliations
or the ECM also influence cardiomyocyte karyokinesis From the Department of Developmental Genetics (C.-C.W., D.Y.R.S.), German
remains to be determined. Centre for Cardiovascular Research (DZHK) Partner site Rhein Main (C.-C.W.,
Although adult cardiomyocytes are able to undergo S.J., J.G., D.Y.R.S.), and Biomolecular Mass Spectrometry (S.J., J.G.), Max Planck
Institute for Heart and Lung Research, Bad Nauheim, Germany.
self-renewal throughout life,57 their low proliferation rate
does not support efficient cardiac repair after MI. Thus, Acknowledgments
identifying ways to induce adult cardiomyocyte cell cycle We thank Arica Beisaw, Felix Gunawan, Paolo Panza, Rashmi Priya, and Michelle
Collins for critical reading of the manuscript, Mathilda Mommersteeg, Susann
activity has been a major goal in the cardiac repair field. Bruche, Eldad Tzahor, and Kfir-Baruch Umansky for collaboration, and all Stainier
However, mammalian cardiomyocyte cell cycle activity lab members for helpful discussion and sharing of reagents.
rarely leads to successful cytokinesis and the production
Sources of Funding
of new cells. In the adult heart, multi-isotope imaging mass
This work was supported by a Croucher fellowship for postdoctoral research (C.-
spectrometry revealed that only about 14% of the cycling C. Wu) and the Max Planck Society and Leducq foundation (D.Y.R. Stainier).
cardiomyocytes adjacent to the infarct zone after MI were
mononuclear and diploid, suggestive of a low cytokinesis Disclosures
None.
rate.9 Attempts to augment cardiomyocyte cell cycle activ-
ity by overexpressing cell cycle regulators including cyclin Supplemental Materials
D, A2, or the transcription factor c-Myc likewise result in References64–75
multinucleation rather than cytokinesis.58 Consequently,
identifying factors that can promote cardiomyocyte cytoki-
REFERENCES
nesis, in addition to cell cycle re-entry, should be beneficial
1. Sereti KI, Nguyen NB, Kamran P, Zhao P, Ranjbarvaziri S, Park S, Sabri
for the development of efficient therapeutic strategies to S, Engel JL, Sung K, Kulkarni RP, et al. Analysis of cardiomyocyte clonal
stimulate cardiac repair via cardiomyocyte proliferation. expansion during mouse heart development and injury. Nat Commun.
However, commonly used readouts of cardiomyocyte pro- 2018;9:754. doi: 10.1038/s41467-018-02891-z
2. Meilhac SM, Kelly RG, Rocancourt D, Eloy-Trinquet S, Nicolas JF,
liferation like Ki67,59 EdU,59,60 and Aurora B-kinase38 are
Buckingham ME. A retrospective clonal analysis of the myocardium reveals
not suitable to unambiguously detect cytokinesis. In con- two phases of clonal growth in the developing mouse heart. Development.
trast, the binucleation assay introduced here (Figure 1A) 2003;130:3877–3889. doi: 10.1242/dev.00580

Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303 September 11, 2020   905


Wu et al The ECM Regulates Cardiomyocyte Cytokinesis

3. Soonpaa MH, Kim KK, Pajak L, Franklin M, Field LJ. Cardiomyocyte DNA oxygen-rich postnatal environment induces cardiomyocyte cell-cycle
synthesis and binucleation during murine development. Am J Physiol. arrest through DNA damage response. Cell. 2014;157:565–579. doi:
Original Research

1996;271:H2183–H2189. doi: 10.1152/ajpheart.1996.271.5.H2183 10.1016/j.cell.2014.03.032


4. Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA, Olson EN, 24. Kimura W, Xiao F, Canseco DC, Muralidhar S, Thet S, Zhang HM,
Sadek HA. Transient regenerative potential of the neonatal mouse heart. Abderrahman Y, Chen R, Garcia JA, Shelton JM, et al. Hypoxia fate mapping
Science. 2011;331:1078–1080. doi: 10.1126/science.1200708 identifies cycling cardiomyocytes in the adult heart. Nature. 2015;523:226–
5. Patterson M, Barske L, Van Handel B, Rau CD, Gan P, Sharma A, Parikh S, 230. doi: 10.1038/nature14582
Denholtz M, Huang Y, Yamaguchi Y, et al. Frequency of mononuclear diploid 25. Pinto AR, Ilinykh A, Ivey MJ, Kuwabara JT, D’Antoni ML, Debuque
cardiomyocytes underlies natural variation in heart regeneration. Nat Genet. R, Chandran A, Wang L, Arora K, Rosenthal NA, et al. Revisit-
2017;49:1346–1353. doi: 10.1038/ng.3929 ing cardiac cellular composition. Circ Res. 2016;118:400–409. doi:
6. Wills AA, Holdway JE, Major RJ, Poss KD. Regulated addition of new 10.1161/CIRCRESAHA.115.307778
myocardial and epicardial cells fosters homeostatic cardiac growth and 26. Furtado MB, Nim HT, Boyd SE, Rosenthal NA. View from the heart: car-
maintenance in adult zebrafish. Development. 2008;135:183–192. doi: diac fibroblasts in development, scarring and regeneration. Development.
10.1242/dev.010363 2016;143:387–397. doi: 10.1242/dev.120576
7. Kikuchi K, Holdway JE, Werdich AA, Anderson RM, Fang Y, Egnaczyk GF, 27. Ieda M, Tsuchihashi T, Ivey KN, Ross RS, Hong TT, Shaw RM, Srivastava
Evans T, Macrae CA, Stainier DY, Poss KD. Primary contribution to zebrafish D. Cardiac fibroblasts regulate myocardial proliferation through beta1
heart regeneration by gata4(+) cardiomyocytes. Nature. 2010;464:601– integrin signaling. Dev Cell. 2009;16:233–244. doi: 10.1016/j.
605. doi: 10.1038/nature08804 devcel.2008.12.007
8. González-Rosa JM, Sharpe M, Field D, Soonpaa MH, Field LJ, 28. Ivey MJ, Kuwabara JT, Pai JT, Moore RE, Sun Z, Tallquist MD. Resident fibro-
Burns CE, Burns CG. Myocardial polyploidization creates a barrier to blast expansion during cardiac growth and remodeling. J Mol Cell Cardiol.
heart regeneration in zebrafish. Dev Cell. 2018;44:433–446.e7. doi: 2018;114:161–174. doi: 10.1016/j.yjmcc.2017.11.012
10.1016/j.devcel.2018.01.021 29. Rienks M, Papageorgiou AP, Frangogiannis NG, Heymans S. Myocar-
9. Senyo SE, Steinhauser ML, Pizzimenti CL, Yang VK, Cai L, Wang M, Wu dial extracellular matrix: an ever-changing and diverse entity. Circ Res.
TD, Guerquin-Kern JL, Lechene CP, Lee RT. Mammalian heart renewal 2014;114:872–888. doi: 10.1161/CIRCRESAHA.114.302533
by pre-existing cardiomyocytes. Nature. 2013;493:433–436. doi: 30. Williams C, Quinn KP, Georgakoudi I, Black LD 3rd. Young developmen-
10.1038/nature11682 tal age cardiac extracellular matrix promotes the expansion of neo-
10. Cahill TJ, Choudhury RP, Riley PR. Heart regeneration and repair after myo- natal cardiomyocytes in vitro. Acta Biomater. 2014;10:194–204. doi:
cardial infarction: translational opportunities for novel therapeutics. Nat Rev 10.1016/j.actbio.2013.08.037
Drug Discov. 2017;16:699–717. doi: 10.1038/nrd.2017.106 31. Hanson KP, Jung JP, Tran QA, Hsu SP, Iida R, Ajeti V, Campagnola PJ,
11. Zebrowski DC, Becker R, Engel FB. Towards regenerating the mamma- Eliceiri KW, Squirrell JM, Lyons GE, et al. Spatial and temporal analysis
lian heart: challenges in evaluating experimentally induced adult mam- of extracellular matrix proteins in the developing murine heart: a blue-
malian cardiomyocyte proliferation. Am J Physiol Heart Circ Physiol. print for regeneration. Tissue Eng Part A. 2013;19:1132–1143. doi:
2016;310:H1045–H1054. doi: 10.1152/ajpheart.00697.2015 10.1089/ten.TEA.2012.0316
12. Mahmoud AI, Kocabas F, Muralidhar SA, Kimura W, Koura AS, Thet S, 32. Chen Z, Xie J, Hao H, Lin H, Wang L, Zhang Y, Chen L, Cao S, Huang X,
Porrello ER, Sadek HA. Meis1 regulates postnatal cardiomyocyte cell cycle Liao W, et al. Ablation of periostin inhibits post-infarction myocardial regen-
arrest. Nature. 2013;497:249–253. doi: 10.1038/nature12054 eration in neonatal mice mediated by the phosphatidylinositol 3 kinase/
13. Monroe TO, Hill MC, Morikawa Y, Leach JP, Heallen T, Cao S, Krijger PHL, glycogen synthase kinase 3β/cyclin D1 signalling pathway. Cardiovasc Res.
de Laat W, Wehrens XHT, Rodney GG, et al. YAP partially reprograms chro- 2017;113:620–632. doi: 10.1093/cvr/cvx001
Downloaded from http://ahajournals.org by on July 8, 2022

matin accessibility to directly induce adult cardiogenesis in vivo. Dev Cell. 33. Kühn B, del Monte F, Hajjar RJ, Chang YS, Lebeche D, Arab S, Keating
2019;48:765–779.e7. doi: 10.1016/j.devcel.2019.01.017 MT. Periostin induces proliferation of differentiated cardiomyocytes and pro-
14. Heallen T, Zhang M, Wang J, Bonilla-Claudio M, Klysik E, Johnson RL, motes cardiac repair. Nat Med. 2007;13:962–969. doi: 10.1038/nm1619
Martin JF. Hippo pathway inhibits Wnt signaling to restrain cardiomyo- 34. Bassat E, Mutlak YE, Genzelinakh A, Shadrin IY, Baruch Umansky K,
cyte proliferation and heart size. Science. 2011;332:458–461. doi: Yifa O, Kain D, Rajchman D, Leach J, Riabov Bassat D, et al. The extra-
10.1126/science.1199010 cellular matrix protein agrin promotes heart regeneration in mice. Nature.
15. Heallen T, Morikawa Y, Leach J, Tao G, Willerson JT, Johnson RL, Martin 2017;547:179–184. doi: 10.1038/nature22978
JF. Hippo signaling impedes adult heart regeneration. Development. 35. Golden HB, Gollapudi D, Gerilechaogetu F, Li J, Cristales RJ, Peng X,
2013;140:4683–4690. doi: 10.1242/dev.102798 Dostal DE. Isolation of cardiac myocytes and fibroblasts from neonatal
16. Bersell K, Arab S, Haring B, Kühn B. Neuregulin1/ErbB4 signaling induces rat pups. Methods Mol Biol. 2012;843:205–214. doi: 10.1007/978-1-
cardiomyocyte proliferation and repair of heart injury. Cell. 2009;138:257– 61779-523-7_20
270. doi: 10.1016/j.cell.2009.04.060 36. Franco-Barraza J, Beacham DA, Amatangelo MD, Cukierman E. Preparation
17. D’Uva G, Aharonov A, Lauriola M, Kain D, Yahalom-Ronen Y, Carvalho S, of extracellular matrices produced by cultured and primary fibroblasts. Curr
Weisinger K, Bassat E, Rajchman D, Yifa O, et al. ERBB2 triggers mam- Protoc Cell Biol. 2016;71:10.9.1–10.9.34. doi: 10.1002/cpcb.2
malian heart regeneration by promoting cardiomyocyte dedifferentiation and 37. DeLaughter DM, Bick AG, Wakimoto H, McKean D, Gorham JM, Kathiriya
proliferation. Nat Cell Biol. 2015;17:627–638. doi: 10.1038/ncb3149 IS, Hinson JT, Homsy J, Gray J, Pu W, et al. Single-cell resolution of temporal
18. Hirose K, Payumo AY, Cutie S, Hoang A, Zhang H, Guyot R, Lunn D, Bigley gene expression during heart development. Dev Cell. 2016;39:480–490.
RB, Yu H, Wang J, et al. Evidence for hormonal control of heart regenerative doi: 10.1016/j.devcel.2016.10.001
capacity during endothermy acquisition. Science. 2019;364:184–188. doi: 38. Hesse M, Doengi M, Becker A, Kimura K, Voeltz N, Stein V, Fleischmann
10.1126/science.aar2038 BK. Midbody positioning and distance between daughter nuclei enable
19. Engel FB, Schebesta M, Keating MT. Anillin localization defect in car- unequivocal identification of cardiomyocyte cell division in mice. Circ Res.
diomyocyte binucleation. J Mol Cell Cardiol. 2006;41:601–612. doi: 2018;123:1039–1052. doi: 10.1161/CIRCRESAHA.118.312792
10.1016/j.yjmcc.2006.06.012 39. Ali SR, Hippenmeyer S, Saadat LV, Luo L, Weissman IL, Ardehali R.
20. Engel FB, Schebesta M, Duong MT, Lu G, Ren S, Madwed JB, Jiang H, Existing cardiomyocytes generate cardiomyocytes at a low rate after
Wang Y, Keating MT. p38 MAP kinase inhibition enables proliferation of birth in mice. Proc Natl Acad Sci U S A. 2014;111:8850–8855. doi:
adult mammalian cardiomyocytes. Genes Dev. 2005;19:1175–1187. doi: 10.1073/pnas.1408233111
10.1101/gad.1306705 40. Leone M, Musa G, Engel FB. Cardiomyocyte binucleation is associated
21. Liu H, Zhang CH, Ammanamanchi N, Suresh S, Lewarchik C, Rao K, Uys with aberrant mitotic microtubule distribution, mislocalization of RhoA
GM, Han L, Abrial M, Yimlamai D, et al. Control of cytokinesis by beta-adren- and IQGAP3, as well as defective actomyosin ring anchorage and cleav-
ergic receptors indicates an approach for regulating cardiomyocyte endow- age furrow ingression. Cardiovasc Res. 2018;114:1115–1131. doi:
ment. Sci Transl Med. 2019;11:eaaw6419. 10.1093/cvr/cvy056
22. Mohamed TMA, Ang YS, Radzinsky E, Zhou P, Huang Y, Elfenbein A, Foley 41. Hashimoto H, Yuasa S, Tabata H, Tohyama S, Hayashiji N, Hattori F, Muraoka
A, Magnitsky S, Srivastava D. Regulation of cell cycle to stimulate adult N, Egashira T, Okata S, Yae K, et al. Time-lapse imaging of cell cycle
cardiomyocyte proliferation and cardiac regeneration. Cell. 2018;173:104– dynamics during development in living cardiomyocyte. J Mol Cell Cardiol.
116.e12. doi: 10.1016/j.cell.2018.02.014 2014;72:241–249. doi: 10.1016/j.yjmcc.2014.03.020
23. Puente BN, Kimura W, Muralidhar SA, Moon J, Amatruda JF, Phelps 42. Kakkar R, Lee RT. Intramyocardial fibroblast myocyte communication. Circ
KL, Grinsfelder D, Rothermel BA, Chen R, Garcia JA, et al. The Res. 2010;106:47–57. doi: 10.1161/CIRCRESAHA.109.207456

906   September 11, 2020 Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303


Wu et al The ECM Regulates Cardiomyocyte Cytokinesis

43. Laser M, Willey CD, Jiang W, Cooper G 4th, Menick DR, Zile MR, Kuppuswamy 60. Magadum A, Ding Y, He L, Kim T, Vasudevarao MD, Long Q, Yang K,
D. Integrin activation and focal complex formation in cardiac hypertrophy. J Wickramasinghe N, Renikunta HV, Dubois N, et al. Live cell screening plat-

Original Research
Biol Chem. 2000;275:35624–35630. doi: 10.1074/jbc.M006124200 form identifies PPARδ as a regulator of cardiomyocyte proliferation and
44. Ruoslahti E. RGD and other recognition sequences for integrins. Annu Rev cardiac repair. Cell Res. 2017;27:1002–1019. doi: 10.1038/cr.2017.84
Cell Dev Biol. 1996;12:697–715. doi: 10.1146/annurev.cellbio.12.1.697 61. Piekny A, Werner M, Glotzer M. Cytokinesis: welcome to the Rho zone.
45. Cukierman E, Pankov R, Stevens DR, Yamada KM. Taking cell-matrix Trends Cell Biol. 2005;15:651–658. doi: 10.1016/j.tcb.2005.10.006
adhesions to the third dimension. Science. 2001;294:1708–1712. doi: 62. Backer S, Lokmane L, Landragin C, Deck M, Garel S, Bloch-Gallego E. Trio
10.1126/science.1064829 gef mediates rhoa activation downstream of slit2 and coordinates telence-
46. Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell. phalic wiring. Development. 2018;145:dev153692.
2002;110:673–687. doi: 10.1016/s0092-8674(02)00971-6 63. Kong R, Yi F, Wen P, Liu J, Chen X, Ren J, Li X, Shang Y, Nie Y, Wu K, et al.
47. Milner R, Edwards G, Streuli C, Ffrench-Constant C. A role in migration Myo9b is a key player in SLIT/ROBO-mediated lung tumor suppression. J
for the alpha V beta 1 integrin expressed on oligodendrocyte precursors. J Clin Invest. 2015;125:4407–4420. doi: 10.1172/JCI81673
Neurosci. 1996;16:7240–7252. 64. Lukinavičius G, Reymond L, D’Este E, Masharina A, Göttfert F, Ta H, Güther
48. Lancaster OM, Le Berre M, Dimitracopoulos A, Bonazzi D, Zlotek-Zlotkiewicz A, Fournier M, Rizzo S, Waldmann H, et al. Fluorogenic probes for live-
E, Picone R, Duke T, Piel M, Baum B. Mitotic rounding alters cell geometry to cell imaging of the cytoskeleton. Nat Methods. 2014;11:731–733. doi:
ensure efficient bipolar spindle formation. Dev Cell. 2013;25:270–283. doi: 10.1038/nmeth.2972
10.1016/j.devcel.2013.03.014 65. Lukinavičius G, Blaukopf C, Pershagen E, Schena A, Reymond L, Derivery E,
49. Naba A, Clauser KR, Hoersch S, Liu H, Carr SA, Hynes RO. The matrisome: Gonzalez-Gaitan M, D’Este E, Hell SW, Wolfram Gerlich D, et al. SiR-Hoechst
in silico definition and in vivo characterization by proteomics of normal and is a far-red DNA stain for live-cell nanoscopy. Nat Commun. 2015;6:8497.
tumor extracellular matrices. Mol Cell Proteomics. 2012;11:M111.014647. doi: 10.1038/ncomms9497
doi: 10.1074/mcp.M111.014647 66. Graumann J, Hubner NC, Kim JB, Ko K, Moser M, Kumar C, Cox J,
50. O’Meara CC, Wamstad JA, Gladstone RA, Fomovsky GM, Butty VL, Schöler H, Mann M. Stable isotope labeling by amino acids in cell cul-
Shrikumar A, Gannon JB, Boyer LA, Lee RT. Transcriptional reversion of ture (SILAC) and proteome quantitation of mouse embryonic stem cells
cardiac myocyte fate during mammalian cardiac regeneration. Circ Res. to a depth of 5,111 proteins. Mol Cell Proteomics. 2008;7:672–683. doi:
2015;116:804–815. doi: 10.1161/CIRCRESAHA.116.304269 10.1074/mcp.M700460-MCP200
51. Zhao J, Mommersteeg MTM. Slit-Robo signalling in heart development. Car- 67. Billing AM, Ben Hamidane H, Graumann J. Quantitative proteomic
diovasc Res. 2018;114:794–804. doi: 10.1093/cvr/cvy061 approaches in mouse: stable isotope incorporation by metabolic (SILAC)
52. Patra C, Diehl F, Ferrazzi F, van Amerongen MJ, Novoyatleva T, Schaefer L, or chemical labeling (reductive dimethylation) combined with high-res-
Mühlfeld C, Jungblut B, Engel FB. Nephronectin regulates atrioventricular olution mass spectrometry. Curr Protoc Mouse Biol. 2015;5:1–20. doi:
canal differentiation via Bmp4-Has2 signaling in zebrafish. Development. 10.1002/9780470942390.mo140156
2011;138:4499–4509. doi: 10.1242/dev.067454 68. Boersema PJ, Raijmakers R, Lemeer S, Mohammed S, Heck AJ. Multiplex
53. Acharya A, Baek ST, Huang G, Eskiocak B, Goetsch S, Sung CY, Banfi S, peptide stable isotope dimethyl labeling for quantitative proteomics. Nat
Sauer MF, Olsen GS, Duffield JS, et al. The bHLH transcription factor Tcf21 Protoc. 2009;4:484–494. doi: 10.1038/nprot.2009.21
is required for lineage-specific EMT of cardiac fibroblast progenitors. Devel- 69. Hsu JL, Huang SY, Chow NH, Chen SH. Stable-isotope dimethyl label-
opment. 2012;139:2139–2149. doi: 10.1242/dev.079970 ing for quantitative proteomics. Anal Chem. 2003;75:6843–6852. doi:
54. Li Y, Asfour H, Bursac N. Age-dependent functional crosstalk between car- 10.1021/ac0348625
diac fibroblasts and cardiomyocytes in a 3D engineered cardiac tissue. Acta 70. Rappsilber J, Ishihama Y, Mann M. Stop and go extraction tips for matrix-
Biomater. 2017;55:120–130. doi: 10.1016/j.actbio.2017.04.027 assisted laser desorption/ionization, nanoelectrospray, and LC/MS
Downloaded from http://ahajournals.org by on July 8, 2022

55. Farbehi N, Patrick R, Dorison A, Xaymardan M, Janbandhu V, Wystub-Lis sample pretreatment in proteomics. Anal Chem. 2003;75:663–670. doi:
K, Ho JW, Nordon RE, Harvey RP. Single-cell expression profiling reveals 10.1021/ac026117i
dynamic flux of cardiac stromal, vascular and immune cells in health and 71. Kiweler M, Looso M, Graumann J. MARMoSET - extracting publication-
injury. eLife. 2019;8:e43882. ready mass spectrometry metadata from raw files. Mol Cell Proteomics.
56. Tzahor E, Poss KD. Cardiac regeneration strategies: staying young at heart. 2019;18:1700–1702. doi: 10.1074/mcp.TIR119.001505
Science. 2017;356:1035–1039. doi: 10.1126/science.aam5894 72. Cox J, Mann M. MaxQuant enables high peptide identification rates, indi-
57. Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabé-Heider vidualized p.p.b.-range mass accuracies and proteome-wide protein quanti-
F, Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, et al. Evidence fication. Nat Biotechnol. 2008;26:1367–1372. doi: 10.1038/nbt.1511
for cardiomyocyte renewal in humans. Science. 2009;324:98–102. doi: 73. Cox J, Neuhauser N, Michalski A, Scheltema RA, Olsen JV, Mann M.
10.1126/science.1164680 Andromeda: a peptide search engine integrated into the MaxQuant environ-
58. Ahuja P, Sdek P, MacLellan WR. Cardiac myocyte cell cycle control in devel- ment. J Proteome Res. 2011;10:1794–1805. doi: 10.1021/pr101065j
opment, disease, and regeneration. Physiol Rev. 2007;87:521–544. doi: 74. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. limma
10.1152/physrev.00032.2006 powers differential expression analyses for RNA-sequencing and microar-
59. Eulalio A, Mano M, Dal Ferro M, Zentilin L, Sinagra G, Zacchigna S, Giacca ray studies. Nucleic Acids Res. 2015;43:e47. doi: 10.1093/nar/gkv007
M. Functional screening identifies miRNAs inducing cardiac regeneration. 75. Xie Y, Allaire JJ, Grolemund G. R markdown: The definitive guide. Boca
Nature. 2012;492:376–381. doi: 10.1038/nature11739 Raton: Taylor & Francis, CRC Press; 2018.

Circulation Research. 2020;127:896–907. DOI: 10.1161/CIRCRESAHA.119.316303 September 11, 2020   907

You might also like