Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Apr 6.
Published in final edited form as: Dev Cell. 2020 Mar 26;53(1):102–116.e8. doi: 10.1016/j.devcel.2020.02.019

Dynamic transcriptional responses to injury of regenerative and non-regenerative cardiomyocytes revealed by single-nucleus RNA sequencing

Miao Cui 1,*, Zhaoning Wang 1,*, Kenian Chen 2, Akansha M Shah 1, Wei Tan 1, Lauren Duan 1, Efrain Sanchez-Ortiz 1, Hui Li 1, Lin Xu 2, Ning Liu 1, Rhonda Bassel-Duby 1, Eric N Olson 1,3,**
PMCID: PMC7365574  NIHMSID: NIHMS1572039  PMID: 32220304

SUMMARY

The adult mammalian heart is incapable of regeneration following injury. In contrast, the neonatal mouse heart can efficiently regenerate during the first week of life. The molecular mechanisms that mediate the regenerative response and its blockade in later life are not understood. Here, by single-nucleus RNA sequencing, we map the dynamic transcriptional landscape of five distinct cardiomyocyte populations in healthy, injured and regenerating mouse hearts. We identify immature cardiomyocytes that enter the cell-cycle following injury and disappear as the heart loses the ability to regenerate. These proliferative neonatal cardiomyocytes display a unique transcriptional program dependent on NFYa and NFE2L1 transcription factors, which exert proliferative and protective functions, respectively. Cardiac overexpression of these two factors conferred protection against ischemic injury in mature mouse hearts that were otherwise non-regenerative. These findings advance our understanding of the cellular basis of neonatal heart regeneration and reveal a transcriptional landscape for heart repair following injury.

eTOC Blurb

Using single-nucleus RNA sequencing, Cui and Wang et al. identified a unique immature cardiomyocyte population associated with heart regeneration in newborn mice. The NFYa and NFE2L1 factors are activated in these cardiomyocytes after injury and can confer protection against ischemic injury in mature mouse hearts that are otherwise nonregenerative.

Graphical Abstract

graphic file with name nihms-1572039-f0007.jpg

INTRODUCTION

The adult mammalian heart is among the least regenerative of adult tissues. In contrast, the heart of newborn mammals possesses a unique ability to regenerate following various types of injury (Porrello et al., 2011; Porrello et al., 2013; Xin et al., 2013b). In mice, this regenerative capacity is lost by postnatal day (P) 7. Restoration of the dormant neonatal cardiac regenerative response in adulthood represents a possible therapeutic approach for heart repair, but will require a deeper understanding of the mechanistic underpinnings of regeneration.

Regenerating cardiomyocytes in neonatal hearts are derived from preexisting cardiomyocytes instead of cardiac stem cells (Porrello et al., 2011; Porrello et al., 2013), however, whether all cardiomyocytes in newborn mammals have the ability to regenerate or only a unique subset of cardiomyocytes performs this function is unknown. In addition, it is unclear whether there are molecular distinctions between regenerative and non-regenerative cardiomyocytes and, if so, what molecular mechanisms contribute to the neonatal regenerative response and to its blockade in later life. Here, we used single-nucleus RNA sequencing (snRNA-seq) to reveal distinct populations of cardiomyocytes in regenerating neonatal and non-regenerative later stage hearts. We show that a distinct immature population of cardiomyocytes enters the cell-cycle in response to injury. Transcriptome analysis of these cardiomyocytes exposed a gene regulatory network that supports the neonatal regenerative response, the loss of which coincides with the blockade to regeneration. These findings highlight possible therapeutic inroads into the processes of heart regeneration and repair.

RESULTS

snRNA-seq reveals differential composition of cardiomyocytes in regenerative and non-regenerative hearts

Neonatal mouse hearts subjected to apical resection or myocardial infarction (MI) show induced cardiomyocyte proliferation by 7-days after injury, suggestive of an acute regenerative response (Porrello et al., 2013). By assessing the number of proliferating cardiomyocytes in regenerative hearts at 1, 3, and 7 days post-MI or sham surgery performed at P1, we identified the onset of the regenerative response at which cardiomyocytes start to proliferate as early as 3-days post-MI (Figures 1AC). To profile the neonatal regenerative response at single cell resolution, we performed single-nucleus RNA sequencing (snRNA-seq) on cardiomyocytes of regenerative hearts 1- and 3-days after MI or sham surgery at P1 (Figure 1D). In parallel, we collected cardiomyocytes from non-regenerative P8 hearts at the same timepoints post-surgery. For each time-point, we dissected ventricular tissue below the ligation suture from 8 to 12 hearts to control for the dissection and individual variation. Due to the large size of cardiomyocytes, microfluidics-based single-cell RNA-seq (scRNA-seq) is not feasible. Therefore, we isolated cardiomyocyte nuclei by FACS using an antibody against PCM1, a cardiac nuclear membrane protein, and performed snRNA-seq on cardiomyocyte nuclei (Alkass et al., 2015; Habib et al., 2017; Wu et al., 2019) (Figures S1A and S1B). We captured 21,737 single cardiomyocyte nuclei from all 8 samples with high integrity (Figures 1E, 1F and S1AS1C), and sequenced a median of 982 genes per nucleus (Figures S1D and S1E). Given that nuclear RNA was profiled, 23% of UMIs (Unique Molecular Identifiers) were mapped to introns and 52% to exons, thus the gene expression profile likely reflects nascent transcription as well as the cellular transcriptome (Figure S1D).

Figure 1. Single-nucleus RNA sequencing identifies distinct cardiomyocyte populations in neonatal heart.

Figure 1.

See also Figure S1 and Figure S2.

A. Schematic showing myocardial infarction (MI) or sham surgery performed at P1 and subsequent days (d) of sample collection for pH3 staining.

B. Fold change of pH3+ cardiomyocytes (cTnT+) in MI hearts at 1-, 3-, and 7-days after P1 surgery compared with sham hearts at the same stage (Sham heart n=3, MI heart n=4). *p<0.05, **p<0.01. Data are represented as mean ± SD.

C. Immunohistochemistry showing pH3+ cardiomyocytes (cTnT+) in heart sections collected at 3-days after MI (P1-MI-d3) or sham (P1-Sham-d3) performed at P1. Scale bar, 50um.

D. Schematic of the experimental design for snRNA-seq analysis. MI or sham surgery was performed on P1 and P8 hearts. Ventricles below the ligation plane (indicated by dashed lines) were collected at 1- and 3-days post-surgery.

E. UMAP visualization of cardiomyocyte clusters colored by identity (n =21,737).

F. UMAP visualization of cardiomyocyte clusters colored by timepoint.

G. Heatmap of Myh6 expression in each cardiomyocyte cluster projected on UMAP graph. Color scale represents the expression of Myh6.

H. Gene signatures of cardiomyocyte CM1-CM5 populations based on expression levels of top markers for each cluster, displaying 200 randomly selected cells per cluster.

I. Fraction of cardiomyocyte populations in each snRNA-seq sample.

To allow cross-sample comparisons, we integrated datasets from all 8 samples for cell type clustering (Becht et al., 2018; Stuart et al., 2019) (Figure S2A and S2B). Cell-type identity was assigned based on top differentially expressed genes and expression of known cellular marker genes among cell clusters (Figures S2CS2J). Among the 11 cell clusters identified, five major clusters (designated CM1-CM5) were identified as cardiomyocytes based on expression of Myh6 (Figures 1G and S2J). Non-cardiomyocyte populations, which include fibroblasts and endothelial cells, were also captured but at a much lower frequency (25%) (Figures S2AS2B).

The five cardiomyocyte clusters CM1 to CM5 were classified based on expression of different sets of marker genes (Figure 1H). Among them, CM1 was the most abundant population, comprising 50%−75% of all cardiomyocytes in P1 and P8 hearts (Figure 1I). Cluster CM2, which highly expressed cell-cycle genes, represented a small population of cardiomyocytes in both P1 (6%) and P8 (4%) hearts (Figures 1H and 1I). Cluster CM3 uniquely expressed Ddc, a catalase of dopamine, accounting for ∼14% of cardiomyocytes in both P1 and P8 samples (Menheniott et al., 2008). We identified cluster CM4 as the most dynamic cardiomyocyte population between P1 and P8 hearts. Cluster CM4 was enriched in P1 hearts, comprising over 20% of cardiomyocytes, but it sharply declined to less than 4% in P8 samples (Figure 1I). CM4 was marked by expression of genes involved in oxygen-reduction, including Atp5b, Mdh2, Sod2, and Mb, suggesting a unique metabolic state (Figure 1H). Cluster CM5, an injury-induced population, was primarily present in MI samples and was significantly enriched in P8 hearts. The top marker genes of CM5, including Xirp2, Cd44, and Ankrd1, are involved in actin-filament regulation, suggesting changes of cellular migration and adhesion (Figure 1H). Thus, snRNA-seq revealed CM4 and CM5 as distinct and dynamic cardiomyocyte populations associated with the regenerative and injured non-regenerative heart, respectively.

The injury-induced CM5 population elicits an apoptotic and hypertrophic response in the non-regenerative heart

The CM5 population was prominently induced after injury (from an average of 3.5% in all sham samples to an average of 13% in all MI samples) (Figure 2A). We observed that the CM5 population was preferentially induced in P8 hearts (Figure 2A). To identify the anatomical location of CM5 cells in P8 injured hearts, we performed immunohistochemistry on sections of P8 MI hearts using antibodies against the identified marker genes, Xirp2 and Cd44 (Figures 1H). Xirp2+ CM5 cardiomyocytes were detected at the border zone in the infarcted P8 hearts, where they colocalized with Cd44+ cardiomyocytes (Figures 2B and 2C).

Figure 2. Identification of the hypertrophic response of CM5 cardiomyocytes following injury.

Figure 2.

See also Table S1.

A. Percentage of CM5 cardiomyocytes in each snRNA-seq sample.

B. Immunohistochemistry of heart sections from P8 heart 1-day after MI showing Xirp2+ cardiomyocytes in the border zone. TUNEL co-staining marks the infarct zone. Scale bar, 50um.

C. Immunohistochemistry of heart sections from P8 heart 1-day after MI showing colocalization of Xirp2+ and Cd44+ cardiomyocytes. Scale bar, 50um.

D. Monocle2 pseudotime analysis of major cardiomyocyte populations in injured P8 hearts depicting an injury response trajectory of CM1 cells towards CM5 cells. Cell identities are projected on the injury trajectory (left); Injury trajectory of non-regenerative CM5 cells (arrow) is shown in pseudotime (right).

E. Palantir pseudotime analysis also revealed an injury response trajectory (arrow) of CM1 cells to CM5 cells.

F. Activation of Xirp2 and Cd44 expression along the injury response trajectory (arrow) of CM5 cells.

G. Heatmap showing the expression of differentially regulated genes along the CM1-to-CM5 injury trajectory at 1-day post-MI in P8 hearts (left). Top GO terms for genes down-regulated (DOWN) and up-regulated (UP) after injury in CM5 cells compared to CM1 cells are shown (right).

H. Volcano plot showing fold changes and p-values of genes up-regulated (CM5-MI-d3) and down-regulated (CM5-MI-d1) in CM5 cells at 3-days post-MI compared to 1-day post-MI (left) and top enriched GO terms (right).

I. Immunostaining of heart sections from the P8 injured heart 7 days after MI showing the hypertrophic phenotype in Xirp2+ CM5 cardiomyocytes in the border zone. WGA counter staining was used to show cell borders. Scale bar, 50um.

J. Quantification of transverse area of Xirp2 and Xirp2+ cardiomyocytes in P8 hearts 7 days post injury, showing hypertrophy of Xirp2+ CM5 cells. Four different hearts were analyzed. *** p< 0.001.

The emergence of the CM5 cardiomyocyte population in a short time period suggests that it is derived from another cardiomyocyte population that may have undergone transcriptional remodeling in response to injury. Indeed, pseudotime analyses, using both Monocle and Palantir, in the P8 heart at 1-day post-MI revealed an injury-responsive trajectory of CM1 cells toward CM5 cells (Figures 2DF). Differential gene analysis identified 282 downregulated and 333 upregulated genes along the CM5 injury trajectory in the P8 heart (Figure 2G and Table S1). Downregulated genes were mostly related to heart contraction and calcium channels, suggesting decreased cardiac function (Figure 2G). Upregulated genes were associated with cell junction organization, programmed cell death, response to stress, and apoptosis. The activation of apoptotic pathways suggests that death of CM5 cells coincides with the decrease of this population from 1- to 3-days post-MI (Figure 1I).

In response to injury, cardiomyocytes in non-regenerative hearts undergo hypertrophic remodeling (Pangonyte et al., 2008). Differential gene analysis comparing the transcriptome of CM5 cells at 3-days post-MI to that at 1-day post-MI indeed showed activation of genes associated with dilated cardiomyopathy and muscle hypertrophy (Figure 2H). Moreover, WGA staining at 7-days after P8 injury revealed hypertrophy of the Xirp2+ CM5 cardiomyocytes compared to Xirp2 cardiomyocytes (Figures 2I and 2J). Additionally, down-regulated genes involved in cell-matrix adhesion, regeneration, and calcium signaling, further indicated a decline in cardiac function. Thus, our analysis revealed early transcriptomic remodeling that precedes the onset of the hypertrophic phenotype in CM5 cells.

Together, these findings highlight the CM5 population as distinct cardiomyocytes that respond to injury by apoptosis and hypertrophic remodeling. The pronounced injury-induction of CM5 in P8 hearts likely contributes to myocardial loss and hypertrophic growth of cardiomyocytes in non-regenerative hearts.

An immature CM4 cardiomyocyte population proliferates after injury

Analysis of the abundance of each cardiomyocyte population in sham hearts at various ages revealed enrichment of the CM4 population in the regenerative stage (P2 and P4) and a sharp decrease at the non-regenerative stage (P9 and P11) (Figure 3A and 3B). Compared to the other cardiomyocyte populations, CM4 cells express higher levels of markers of immature hearts, including Tnni1, Myh7, and Actc1 (Ames et al., 2013; Cui et al., 2018; Taegtmeyer et al., 2010) (Figure 3C), and lower levels of maturation genes, including Myh6, Ryr2, and Cacna1c (Sun and Nunes, 2017; Taegtmeyer et al., 2010) (Figures 3D), suggesting that they are immature. CM4 cells were also significantly enriched for expression of embryonic genes associated with cell-cycle progression (Figures 3E and S3A) and glycolysis (Figures S3BS3D), consistent with the switch from glycolysis to β-oxidation during cardiomyocyte maturation (Fisher et al., 1980; Lopaschuk et al., 1992; Puente et al., 2014; Wisneski et al., 1985). Genes associated with mitochondrial electron transport and ATP synthesis were also expressed at higher levels in CM4 cells (Figures S3ES3F), whereas genes involved in DNA damage were not differentially expressed (Figure S3G). This suggests that although CM4 cells may have higher energy production, ROS activity was not notably increased (Koopman et al., 2010). Constrained ROS activity in CM4 cells is consistent with their high expression of antioxidant genes, such as Prdx1, Sod1, Sod2, and Cat (Figures S3HS3I).

Figure 3. An immature cardiomyocyte population (CM4) is enriched in the regenerative heart and becomes proliferative after injury.

Figure 3.

See also Figure S3 and Figure S4.

A. Schematic showing decreasing capacity of heart regeneration during postnatal life. Heart samples at timepoints P2, P4, P9 and P11 were collected for sham samples.

B. Percentage change of each cardiomyocyte population (CM1-CM5) in P2, P4, P9 and P11 hearts. *** p < 0.001, Chi square test.

C. Violin plots showing the expression of embryonic genes, Tnni1, Myh7, and Actc1, in CM1-CM5 cells.

D. Violin plots showing the expression of mature cardiomyocyte genes, Myh6, Ryr2, and Cacna1c in CM1-CM5 cells.

E. Heatmap showing expression of embryonic genes that are highly expressed in E15 hearts compared to P1 hearts in CM1-CM5 populations. Z-scores of RPKM values are plotted for the expression in E15 and P1 hearts (left). Z-scores of the averaged expression of imputation corrected values from cluster CM1-CM5 are plotted (right). Representative genes related to cell-cycle regulation, extracellular matrix organization, and fetal contractile gene isoforms are labeled. Top GO terms enriched for these genes are shown (bottom left).

F. Percentage of cardiomyocytes mapped to the G2/M phase in CM1-CM5 populations from P1 heart at 3-days post MI or Sham. n.s, not significant; *** p< 0.001, Chi square test.

G and H. Flow cytometry graphs showing cardiac nuclei labeled with Hoechst alone to analyze DNA content (G), or Hoechst plus PCM-1 antibody to identify 2n and 4n cardiomyocytes (H). Note that the different fluorescent intensity of Hoechst staining indicates 2n and 4n DNA content, which correspond to G0/G1 and G2/M cell-cycle phases, respectively. PCM1+ 2n and 4n cardiomyocyte nuclei were collected and separately analyzed with snRNA-seq. 2n: diploid; 4n: tetraploid; CM: cardiomyocytes.

I. The percentage of CM4 population in 2n and 4n cardiomyocyte nuclei. *** p< 0.001, Chi square test.

As cardiomyocyte proliferation is a prerequisite of heart regeneration, we next assessed proliferative activity of individual cardiomyocytes using a method that predicts cell-cycle phase based on gene expression (Kowalczyk et al., 2015). Analysis of total cardiomyocytes at each timepoint showed a significant increase of cardiomyocytes mapped to the cycling G2/M phase at 3-days post P1 MI compared to sham, as well as a decrease in P8 hearts compared to P1 hearts (Figure S4A), consistent with previously published data (Porrello et al., 2013). To identify which population contributes to the increase of G2/M cardiomyocytes in P1 hearts following injury, we plotted the fraction of G2/M cells in each cardiomyocyte population at 3-days post MI and sham (Figure 3F). This analysis revealed that only in the CM4 population did the fraction of G2/M cells significantly increase after MI (from 34% to 67%). Moreover, cell-cycle genes, such as Mki67 and Ccnb1, were strongly activated in CM4 cells at 3-days post-MI (Figure S4B). Furthermore, a separately performed snRNA-seq analysis on isolated G0/G1(2n) and G2/M (4n) cardiomyocytes, based on DNA content, at 3-days after MI at P1 revealed an enrichment of CM4 in G2/M cycling cardiomyocytes compared to G0/G1 cardiomyocytes, further supporting the proliferative phenotype of CM4 cells after injury (Figures 3G3I and S4CS4J). The CM2 population, which also contains a high percentage of G2/M cells (Figures 3F and S4J), only comprises ∼4% of total cardiomyocytes. Unlike CM4, CM2 cells do not become more proliferative after MI and were detected at a similar abundancy in P1 and P8 sham hearts. Together, these findings reveal CM4 cells as an immature cardiomyocyte population that is enriched in regenerative hearts and enters the cell-cycle at 3-days following injury.

CM4 cells display a defined transcriptional response following injury

To understand the injury response of CM4 cells that underlies their proliferative phenotype during heart regeneration, we compared their transcriptomes at 1-day post sham surgery (P1-sham-d1) to 1-day (P1-MI-d1) and 3-days (P1-MI-d3) after MI injury (Figure 4A and 4B). We identified >900 differentially regulated genes, clustered into 3 groups by their temporal regulation (Figure 4B and Table S2). Genes downregulated in CM4 after injury (MI-down) were associated with respiratory electron transport and aerobic respiration, indicating reduced oxidative metabolism. In this regard, aerobic metabolism increases ROS activity, which inhibits cardiomyocyte proliferation (Puente et al., 2014). The reduced oxidative metabolism in CM4 cells after injury is likely a prerequisite for cell-cycle re-entry (Nakada et al., 2017; Puente et al., 2014). Genes that showed acute upregulation at 1-day post-MI (Ml-up d1) were associated with GO terms including ribosome, ribonucleoprotein complex biogenesis, actin filament-based process, muscle development, and PI3K-Akt signaling (Figure 4B). Remarkably, genes that were activated at 3-days post-MI (MI-up d3) are highly associated with GO terms related to cell-cycle regulation, further confirming active proliferation of CM4 cells after injury (Figure 4B). Extracellular matrix genes were also activated at this stage, suggesting that CM4 cardiomyocytes may remodel the extracellular environment during heart regeneration (Figures 4B and S5A). Thus, the transcriptional response of CM4 cardiomyocytes following injury involves a decrease of oxidative metabolism and an increase in ribosome biogenesis, followed by enhanced activation of cell-cycle processes.

Figure 4. Regenerative response of CM4 cardiomyocytes following injury.

Figure 4.

See also Figure S4, Figure S5, Table S2, and Table S3.

A. Reclustering of CM4 cardiomyocytes reveals transcriptome differences between injured (P1-MI-d1 and P1-MI-d3) and control (P1-Sham-d1) samples, visualized in UMAP.

B. Hierarchical clustering of differentially expressed genes (p< 0.0001, Wilcoxon rank-sum test) in CM4 cells across timepoints identified 3 groups of genes that showed different dynamic regulation in response to injury. Left, heatmap of gene expression in z-scores; middle, z-scores of the average expression of genes in each group, with shade representing standard error; right, top GO terms enriched for genes in each group.

C. Expression of transcription factors that highly correlate (Spearman’s rank correlation coefficient >0.85) with Mki67 and Ccnb1 expression in CM4 cells. Cells are ordered (from left to right) by their expression level of Mki67 from low to high. The corresponding sample point of each cell is color-coded and shown on the top. Representative genes with known roles in cardiomyocyte proliferation are indicated. The predicted upstream regulators of the CM4 injury response are labeled in bold.

D. UMAP visualization of co-embedded scATAC-seq and snRNA-seq datasets obtained in P1 hearts at 3-days after MI. Cardiomyocytes are colored by experimental technique (left). Cardiomyocytes are colored by the cell type identified after the cell-label transfer in Seurat (right). EC, endothelial cells; EndoEC, endocardial cells; EPI, epicardial cells; FB, fibroblasts.

E. Aggregate read counts of scATAC-seq in CM1 and CM4 cells within 2kb range of the CM4-active open chromatin regions.

F. TF motifs enriched at CM4-active open chromatin regions.

G. Relative expression of Srf, Nfe2l, Nfe2l2, Nfya, Nfyb, and Nfic in each cardiomyocyte cluster.

H. Relative expression of Srf, Nfe2l1, Nfe2l2, Nfya, Nfyb, and Nfic in CM4 cells from the sham heart 1-day after surgery (P1-Sham-d1) and hearts at 1-day (P1 -MI-d1 ) and 3-days (P1-MI-d3) after injury.

I. Quantification showing fold-change of the proportion of EdU+ cardiomyocytes in NRVMs overexpressing NFE2L1, NFE2L2, NFIc, NFYa, NFYb, or SRF compared with YFP control (n=3 for each group). * p< 0.05, ** p< 0.01. Data are represented as mean ± SD.

J. EdU (violet) and cTnT (green) immunostaining of NRVMs overexpressing YFP (negative control), SRF, or NFYA followed by EdU incorporation for 24h. Scale bar, 100um.

K. Quantification showing the number of viable NRVMs overexpressing YFP, NFE2L1, NFE2L2, NFIc, NFYa, NFYb, or SRF (n=4 for each group). ** p< 0.01. Data are represented as mean ± SD.

L. Viable NRVMs overexpressing YFP, NFE2L1, or NFE2L2 following H2O2 treatment indicated by Calcerin AM (green). Scale bar, 100um

CM4 cells activate proliferative (NFYa) and pro-survival (NFE2L1) factors in response to injury

To delineate the gene program underlying the proliferative phenotype in CM4 cells, we next analyzed genes whose expression highly correlated (Spearman’s rank correlation coefficient > 0.85) with two cell-cycle genes: Mki67 and Ccnb1. We identified 887 correlated genes, including transcriptional regulators of the Hippo/YAP pathway (Tead2 and Tead4) and the hedgehog signaling pathway (Gli3 and Mycn), all of which can regulate cardiomyocyte proliferation (Figures 4C and Table S3) (Gemberling et al., 2015; Singh et al., 2018; von Gise et al., 2012; Xin et al., 2013a). To identify the potential drivers of the proliferative gene program in CM4 cells, we next performed motif enrichment analysis at promoter regions (±5kb from transcription start sites) of cell-cycle correlated genes. The top enriched transcription factor (TF) binding motifs were the NFE2L binding motif, ETS binding motifs, the SRF CArG-box binding motif, and the CCAAT motif that can be recognized by the NFY and NFI transcription factor complexes (Figure S5BS5C) (Fleming et al., 2013; Maity and de Crombrugghe, 1998; Pjanic et al., 2013).

To identify active chromatin regions that contribute to the unique pattern of CM4 gene expression underlying its regenerative response, we performed single-cell ATAC sequencing (scATAC-seq) on cardiac nuclei 3-days post MI at P1 when we observed increased cell cycle activity in CM4 cells following injury (Figure 3F). We captured 714 cardiomyocytes and identified cells corresponding to all five clusters based on chromatin accessibility of marker genes for each cluster (Figure 4D). We identified 726 chromatin peaks that showed higher accessibility in CM4 compared to CM1 cells, which contribute to the unique gene expression profile of CM4 cells (Figure 4E). Motif analysis on these peaks showed enrichment of NFY, TEAD1 and ETS binding sites, as well as a binding site of NFE2L1 with its cofactor MAFG (Figure 4F) (Johnsen et al., 1996), further supporting these TFs as upstream regulators of the CM4 gene program.

To identify the specific members of each TF family that might associate with these motifs, we searched for family members with enriched expression in CM4 cells that were also up-regulated upon injury. This analysis identified Srf, Nfya, Nfyb, Nfic, Nfe2l1, and Nfe2l2 as candidate upstream regulators with gene expression enriched in CM4 (Figures 4G), upregulated after MI (Figure 4H), and correlated with Mki67 expression (Figure 4C). To investigate the functions of these TFs, we overexpressed them individually in neonatal rat ventricular cardiomyocytes (NRVMs) using adenoviruses and assessed their ability to promote proliferation measured by EdU incorporation. Overexpression of NFYa and SRF significantly increased the number of EdU+ cardiomyocytes compared to a YFP control (Figures 4I and 4J). Additionally, genes involved in mitosis, such as Ccna2, Aurka, Aurkb, Cdc2, and Cdc20, were significantly upregulated (Figure S5D).

Although overexpression of NFE2L1 and NFE2L2 did not increase EdU incorporation, we reasoned that they could play a protective role under hypoxic conditions caused by MI, given their known ability to regulate antioxidant gene expression (Bartelt et al., 2018; Biswas and Chan, 2010; Ohtsuji et al., 2008; Widenmaier et al., 2017). To test this, we performed a cardiomyocyte survival assay under H2O2-induced oxidative stress conditions. Overexpression of NFE2L1 (also known as Nrf1) in NRVMs resulted in a significant increase of viable NRVMs after treatment with 50uM H2O2 for 2h, compared to NRVMs overexpressing YFP, as visualized with Calcerin AM dye (Figures 4K and 4L). Overexpression of NFE2L2 also exerted a pro-survival effect, but to a lesser degree than NFE2L1 (Figures 4K and 4L). Indeed, neonatal hearts depleted of NFE2L2 (also known as NRF2) were unable to regenerate, indicating that induction of the anti-oxidant response is required for heart regeneration (Tao et al., 2016). In summary, by profiling the transcriptional response following injury and the genes correlated with cell-cycle activation in CM4 cells, we identified NFYa and SRF as regulators of cardiomyocyte proliferation, and NFE2L1 and NFE2L2 as regulators of cardiomyocyte survival.

Combined overexpression of NFYa and NFE2L1 elicits proliferative and protective effects in cardiomyocytes

To identify a potential therapeutic cocktail that might confer both proliferative and protective benefits after cardiac injury, we simultaneously overexpressed combinations of SRF, NFYa, and NFE2L1 in NRVMs followed by EdU labeling and H2O2 treatment to analyze cell proliferation and survival, respectively. Overexpression of NFYa and NFE2L1 significantly increased the number of cardiomyocytes that were positive for EdU (Figure 5A), pH3 (Figure 5B), and Aurora B (Figures S5E and S5F). The total number of NRVMs overexpressing NFYA and NFE2L2 or NFYa alone was also increased (Figure S5G). Moreover, overexpression of these factors resulted in a substantial protective effect in NRVMs treated with H2O2, as measured with Calcerin AM live-cell dye and TUNEL staining, similar to the effect of overexpressing NFE2L1 alone (Figures 5C5E and Figure 4L). Furthermore, the protective effect was also observed in NRVMs treated with methyl methanesulfonate (MMS, an agent that induces alkylating DNA damage) (Lundin et al., 2005) and peroxynitrite (a critical contributor to ischemia-reperfusion-dependent tissue injury) (Pacher et al., 2007; Yasmin et al., 1997) (Figures S5H and S5I). Although SRF added in the cocktail further increased proliferation, it did not improve the survival of cardiomyocytes following H2O2 treatment (Figures 5A and 5C). We conclude that simultaneous overexpression of NFYa and NFE2L1 elicits a proliferative and protective effect in cardiomyocytes in vitro.

Figure 5. Combined overexpression of NFYa and NFE2L1 promotes cardiomyocyte proliferation and survival.

Figure 5.

See also Figure S5.

A. Quantification showing the fold change of the proportion of EdU+ NRVMs overexpressing single factors or combinations of NFYa, NFE2L1, and SRF compared with control YFP (n=5 for each group). ** p< 0.001; n.s, not significant. Data are represented as mean ± SD.

B. Immunofluorescent staining of cTnT (green) and pH3 (white) in NRVMs overexpressing YFP or NFYa plus NFE2L1 (left). Quantification of the percentage of pH3+ and cTnT+ cells (right). ** p< 0.01, *** p< 0.001. Scale bar, 100um. Data are represented as mean ± SD.

C. Quantification showing the number of viable NRVMs overexpressing single factors or combinations of NFYa, NFE2L1, and SRF compared with control YFP (n=5 for each group). ** p< 0.01; n.s, not significant. Data are represented as mean ± SD.

D. Viable NRVMs overexpressing YFP with and without H2O2 treatment, or NFYa plus NFE2L1 after H2O2 treatment, indicated by Calcerin AM (green). Scale bar, 100um

E. Immunostaining showing TUNEL+ (purple) and cTnT+ (green) NRVMs overexpressing YFP or NFYa plus NFE2L1 after H2O2 treatment. Scale bar, 100um

F. Heatmap showing expression of selected genes related to injury-activated pathways in CM4 cells. Z-scores of averaged normalized expression values of each gene in CM1 and CM4 at 3-days post Sham and MI (left). Z-scores of RPKM of each gene in three replicates of NRVMs expressing GFP, NFYa, or NFE2L1 (right).

To understand the mechanisms by which NFYa and NFE2L1 function to promote proliferation and survival, we performed RNA-seq analysis on NRVMs that overexpressed GFP (control), NFYa, or NFE2L1. Genes that were specifically up-regulated in response to NFYa overexpression included many cell-cycle genes, with top enriched GO terms all related to mitotic cell-cycle processes (Figure S6A and S6B). Genes specifically up-regulated by NFE2L1 were related to microtubule movement, amino acid metabolic processes, and skeletal muscle regeneration (Figure S6C). Additionally, we identified many genes up-regulated by both NFYa and NFE2L1 that were related to extracellular organization and the immune response (Figure S6D). We compared the genes up-regulated in response to overexpression of NFYa and NFE2L1 to the genes enriched in CM4 cells in MI samples, and found that genes activated in CM4 cells after MI were also markedly up-regulated by overexpression of NFYa or NFE2L1 (Figure 5F). These gene are associated with antioxidant, glycolysis, extracellular matrix, PI3K-Akt-mTOR pathway, and cell-cycle regulation. Thus, overexpression of NFYa and NFE2L1 promotes cardiomyocyte proliferation and survival, at least in part, by activating pathways involved in the injury response of CM4 cells.

Overexpression of NFYa and NFE2L1 improved heart function after acute injury

We next determined whether overexpression of NFYa and NFE2L1 could confer cardio-protection against ischemic injury in nonregenerative hearts. To do so, we generated adeno-associated virus 9 (AAV9) vectors that expressed NFYa, NFE2L1, or TdTomato (control) under the regulatory control of the muscle creatine kinase 8 (CK8e) promoter (Martari et al., 2009), which directs muscle-specific expression (Chamberlain et al., 1985; Jaynes et al., 1986). AAV9 injections were performed intraperitoneally on P4 mice, and expression was analyzed in cardiac ventricles at P8 (Figure 6A). Mice injected with both AAV9-NFYa and AAV9-NFE2L1 showed more than 40-fold overexpression of NFYa and NFE2L1 mRNA, compared with littermates that received the control AAV9 (Figures S6E and S6F). We estimated that ∼50–70% cardiomyocytes were infected with AAV9 based on TdTomato expression in control hearts (Figure S6G). Next, we performed MI on P8 mice pre-injected with AAV9-NFYa and AAV9-NFE2L1 or AAV9-TdTomato and analyzed cardiomyocyte proliferation at P11 by pH3 staining and cardiac function at P29 by echocardiography (Figure 6A). We also performed TUNEL staining on heart samples collected at 6h post- MI to analyze the acute myocardial loss induced by ischemic injury. AAV9 overexpression of both NFYa and NFE2L1 significantly reduced the relative area of TUNEL-positive myocardium and increased the number of pH3+ cardiomyocytes compared with overexpression of TdTomato (Figures 6B, 6C and S6H). Additionally, cardiac function, assessed by fractional shortening and ejection fraction 21 days after injury was significantly improved in mice overexpressing NFYa and NFE2L1 compared with controls (Figures 6D and 6E). Trichrome staining at this timepoint also revealed a reduction of both fibrotic tissue and cardiac dilation in mice that received AAV9-NFYa and AAV9-NFE2L1 compared with controls (Figures 6F and 6G). Furthermore, H&E staining on sections from the same hearts did not show obvious hypertrophy with overexpression of NFYa and NFE2L1 (Figure S6I). Together, these data suggest that overexpression of NFYa and NFE2L1 in vivo confers a proliferative and protective effect in hearts that would otherwise be non-regenerative following injury.

Figure 6. Combined overexpression of NFYa and NFE2L1 confers cardio-protection.

Figure 6.

See also Figure S6.

A. Schematic showing experimental design for AAV9 injection and timepoints of sample collections.

B. Comparison of TUNEL positive area in hearts of mice that received AAV9-TdTomato or AAV9-NFYa+NFE2L1. Samples were collected at 6h post-MI. Left, TUNEL staining of heart sections collected at 400um below the ligation suture. Right, quantification of TUNEL relative signal intensity normalized by heart section size at 0um, 200um, 400um, and 600um below the suture (n=3 for each group). * p< 0.05, ** p< 0.01, n.s not significant. Data are represented as mean ± SD. Scale bar, 100um.

C. Immunostaining of cTnT (purple) and pH3 (white) on transverse sections from hearts of mice treated with AAV9-TdTomato or AAV9-NFYa+NFE2L1 at P11 (left). Fold change of the percentage of pH3+ and cTnT+ cells in AAV9-NFYa+NFE2L1 treated hearts compared with AAV9-TdTomato hearts (n=4 for each group). * p< 0.05. Data are represented as mean ± SD. Scale bar, 50um.

D and E. Fractional shortening (D) and ejection fraction (E) measurements of mice injected with AAV9-TdTomato and AAV9-NFYa+NFE2L1 at P4 and analyzed at 3wks after MI at P8 (n=16 for AAV9-TdTomato, n=17 for AAV9- NFYa+NFE2L1). ** p< 0.01. Data are represented as mean ± SD.

F. Masson’s trichrome staining of transverse sections of hearts of mice injected with AAV9-TdTomato or AAV9-NFYa+NFE2L1. Samples were collected at 3wks after P8 MI. Images of heart sections at 0um, 200um, 400um, 600um, and 800um below the ligation point are depicted. Five representative hearts are shown for each group, and their fractional shortening (FS) measurements are noted. Scale bar, 500um.

G. Quantification of fibrotic regions of heart sections in F (n=3 for each group). * p< 0.05. Data are represented as mean ± SD.

DISCUSSION

The source of regenerating cardiomyocytes in neonatal hearts has remained an important question, although an endogenous stem cell origin has been previously ruled out (Li et al., 2018; Maliken and Molkentin, 2018). We showed previously that the neonatal regenerative heart retains aspects of an earlier developmental gene program, but the cellular origin of this gene program was elusive (Wang et al., 2019). Here, we applied snRNA-seq to interrogate the transcriptome landscape of postnatal cardiomyocytes at regenerative and non-regenerative ages. Our findings identify distinct cardiomyocyte populations that elicit differential injury responses that lead to either proliferation or hypertrophic remodeling after injury. We identify a unique pool of immature cardiomyocytes, CM4, present in neonatal hearts, that represents a cellular source of cardiomyocyte proliferation following injury. These conclusions are supported by three observations. First, among 5 different cardiomyocyte populations, only the CM4 population upregulated cell-cycle genes and showed an increased proportion of cycling cardiomyocytes after injury. Second, by separately profiling cardiomyocytes in G2/M or G0/G1 phases from regenerating hearts, we conversely show that cycling G2/M cardiomyocytes are significantly enriched in the CM4 population. Third, we observed a fractional increase of the CM4 population 3-days after MI at P1 compared to sham (from 19% to 28%), which coincides with its increased proliferative activity following injury.

CM4 cardiomyocytes have high glycolytic metabolism, but also appear to utilize oxidative phosphorylation as seen by high levels of genes regulating mitochondrial electron transport and ATP synthesis. This unique metabolic state has been recently reported in lung and ovarian cancer stem cells, which show the most energetically activated phenotype, characterized by enhanced glycolysis as well as increased mitochondrial oxidative metabolism (Bonuccelli et al., 2017; Martinez-Outschoorn et al., 2017; Sancho et al., 2016). Thus, CM4 cells may adopt a unique metabolic state to cope with the postnatal metabolic demand.

Our study also identifies another proliferative population CM2. Unlike CM4, the abundance of the CM2 population is not proportionally different in regenerative and non-regenerative hearts, nor was cell-cycle activity increased in CM2 cells after injury. Although, it is possible that the CM2 population represents another source of proliferating cardiomyocytes during neonatal heart regeneration, it is unlikely the sole source given its small percentage (∼5%) in the cardiomyocyte pool.

Differential response to injury in regenerative and non-regenerative cardiomyocytes

It was previously suggested that the regeneration of neonatal cardiomyocytes is associated with a developmentally permissive state rather than activation of a regenerative program following injury (Quaife-Ryan et al., 2017). Here, our snRNA-seq data show that the regenerative and non-regenerative cardiomyocytes in neonatal hearts in fact display defined and distinct injury responses.

Our data show that down-regulation of oxidative metabolism and up-regulation of ribosomal synthesis precede cell-cycle activation in CM4 cells and thus are likely required for injury-induced proliferation. Indeed, scavenging ROS or inhibiting DNA damage response pathways can induce postnatal cardiomyocyte proliferation, and increased ribosome biogenesis and protein synthesis in tumors leads to elevated cell proliferation (Martinez-Outschoorn et al., 2017; Puente et al., 2014; Zhou et al., 2015). On the other hand, CM5 cells respond to injury by downregulation of contractile genes and upregulation of genes involved in migration, apoptosis, and hypertrophy. We also observed upregulation of a fetal gene program, including Myh7 and Actc1, in CM5 cells, consistent with the reported reversion to fetal gene program during cardiomyocyte disease remodeling (Cox and Marsh, 2014; Taegtmeyer et al., 2010).

The therapeutic potentials of NFYa and NFE2L1 overexpression

The ability of the regenerative heart to repair damage through cellular proliferation has led to attempts to induce cell-cycle re-entry of terminally differentiated cardiomyocytes, including manipulations of microRNAs, Hippo signaling, and cell-cycle regulators (Aguirre et al., 2014; Cui et al., 2018; Mohamed et al., 2018; Morikawa et al., 2015; Tian et al., 2015; Xin et al., 2013a). Our findings demonstrate that a combination of two TFs, NFYa and NFE2L1, that are injury-induced in CM4 regenerative cardiomyocytes, can efficiently induce cardiomyocyte proliferation and cell survival in vitro and in vivo. Systemic AAV9 delivery of these two factors resulted in improved cardiac function after injury. The combination of proliferative and pro-survival factors to treat cardiac injury represents an appealing therapeutic strategy.

NFYa appears to directly activate cell-cycle progression, as seen by strong up-regulation of many cell-cycle genes and mitosis-related GO terms. NFYa is a subunit of the NF-Y complex, which has been shown to regulate proliferation of hematopoietic stem cells and spermatogonial stem cells (Bungartz et al., 2012; Iyer et al., 2016). NFE2L1 promotes cardiomyocyte survival partially by activating antioxidant genes, including Prdx5 and Ccs, as seen in our RNA-seq data, but may also regulate the endoplasmic reticulum (ER) stress mechanism. In brown adipocytes, NFE2L1 is also localized in the ER and regulates the proteasomal activity to efficiently dispose of misfolded and toxic proteins, therefore protecting cells from metabolic stress (Bartelt et al., 2018). This is in line with our RNA-seq data where we saw upregulation of genes involved in amino acid biosynthesis and metabolism by overexpression of NFE2L1 (Hamel et al., 2003; Suraweera et al., 2012). Together, we show that studying the innate response to injury in regenerating cardiomyocytes can lead to identification of factors that can be leveraged to improve heart repair and function.

Several recent studies have suggested that long-term induction of cardiomyocyte proliferation has detrimental effects (Gabisonia et al., 2019; Monroe et al., 2019). In our study, we assessed the recovery of hearts 3-weeks after infarction and observed improved morphology as well as cardiac function. Although we did not follow the long-term effect, the combined expression of NFYa and NFE2L1 conferred proliferative as well as protective functions, thus providing an additional cell survival benefit to the injured heart.

Single-cell RNA sequencing technology has been widely applied to interrogate cellular heterogeneity of tissues and to construct developmental trajectories of organ differentiation. Here we show that this is also a powerful approach to study the differential response to injury among diverse cell populations within an organ. Future applications to other injury models and in different organs should provide new insights into physiological and pathological responses to various injuries during the mammalian postnatal life.

STAR METHODS

LEAD CONTACT AND MATERIALS AVAILABILITY

Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Eric Olson ([email protected]).

EXPERIMENTAL MODEL AND SUBJECT DETAILS

Experimental Animals

All animal work described in this manuscript has been approved and conducted under the oversight of the UT Southwestern Institutional Animal Care and Use Committee. Timed-pregnant ICR/CD-1 mice (Charles River Laboratories) were used to deliver pups for surgical procedures on postnatal day 1 or 8. Timed-pregnant C57BL/6 mice (Charles River Laboratories) were used to deliver pups for systemic AAV9 injection on P4 and surgical procedures on P8. Neonatal Sprague-Dawley rats (Envigo) were used to isolate neonatal rat ventricular cardiomyocytes (NRVMs). Animals were housed in a 12 h light/dark cycle in a temperature-controlled room in the Animal Research Center of UT Southwestern, with ad libitum access to water and food. Sex was not determined for neonatal pups.

Cell Lines

All cells were cultured at 37°C with 5% CO2. NRVMs were maintained in Dulbecco’s modified Eagle’s medium (DMEM) (Sigma, D5796)/199 medium (Gibco, 11150–059) (3:1) with 3% fetal bovine serum (FBS) (Gemini Bio Products, 100–106), and 1% penicillin-streptomycin (Sigma, P0781). Adeno-X 293 cells (Clontech, 632271) were maintained in DMEM with 10% FBS, and 1% penicillin-streptomycin.

METHOD DETAILS

Neonatal MI

Neonatal mice were anesthetized by hypothermia, and neonatal MI was performed as described previously (Mahmoud et al., 2014; Porrello et al., 2011; Porrello et al., 2013). Mice were euthanized at 1 d (24 h) or 3 d (72 h) post-surgery. Each heart was dissected in ice-cold PBS and a transverse cut on the ligation plane (for MI hearts) or similar level (for sham hearts) was made. Heart tissue below the ligation plane was collected for nuclei isolation.

Histology

Postnatal mouse hearts were fixed in 4% paraformaldehyde (Electron Microscopy Sciences, 15710) in PBS (Sigma, D8537), embedded in paraffin, and sectioned at 5 μm intervals. For trichrome staining, samples were sectioned at different levels below the suture (for MI hearts) or comparable levels (for sham hearts), and stained using Masson’s trichrome staining method.

Cardiomyocyte Nuclei Isolation

Cardiomyocyte nuclei isolation was performed as previously described (Quaife-Ryan et al., 2017) with modifications. All chemicals used were purchased from Sigma Aldrich, unless otherwise indicated. For each nuclear sample, 8–12 tissue samples collected from the same litter were pooled together and minced with a razor blade. Minced samples were resuspended in 5 mL Lysis Buffer (320 mM sucrose, 10 mM Tris-HCl, 5 mM CaCl2, 5 mM Magnesium Acetate, 2 mM EDTA, 0.5 mM EGTA, 1 mM DTT, 1x complete protease inhibitor (Roche), 200U/mL RNase OUT Recombinant Ribonuclease Inhibitor (Invitrogen, 10777–019), pH=8) and homogenized with a dounce grinder for 15 strokes. The lysate was sequentially filtered through 70 μm and 40 μm cell strainers (Falcon, 352350 and 352340) and centrifuged at 1,000 × g for 5 min at 4 °C to pellet nuclei. The nuclear pellet was subsequently resuspended in 2 mL Sucrose Buffer (1 M sucrose, 10 mM Tris-HCl, 5 mM magnesium acetate, 1mM DTT, 1x complete protease inhibitor, 200 U/mL RNase OUT Recombinant Ribonuclease Inhibitor, pH=8) and the suspension was cushioned on top of 4 mL Sucrose Buffer, followed by centrifugation at 1,000 × g for 5 min at 4 °C to pellet nuclei. The n uclear pellet was subsequently washed once in 1 mL of Nuclei Storage Buffer (NSB) (440 mM sucrose, 10 mM Tris-HCl, 70 mM KCl, 10 mM MgCl2, 1.5 mM spermine, 1x complete protease inhibitor, 200 U/mL RNase OUT Recombinant Ribonuclease Inhibitor, pH=7.2). The washed nuclei were resuspended in 1 mL NSB and stained with anti-PCM1 antibody (Sigma, HPA023374, 1:200) for 45 min at 4 °C. After primary antibody staining, nuclei were centrifuged at 1,000 × g for 5 min at 4 °C, washed twice with 1 mL NSB, and stained with secondary antibody conjugated with Alexa Fluor 647 (Invitrogen, A21244, 1:500) for 30 min at 4 °C. Subsequently, nuclei were washed twice with 1 mL NSB and resuspended in 2% BSA/PBS before fluorescence-activated cell sorting (FACS). For FACS, PCM1+ nuclei were sorted using a BD FACSARIA cell sorter in the UT Southwestern Flow Cytometry Facility. Nuclei without primary antibody staining were used as a negative gating control. After sorting, PCM1+ nuclei were pelleted at 1,500 × g for 15 min at 4 °C, resuspended in 2% BSA, and counted using a Countess II Automated Cell Counter (Thermo Scientific).

To isolate 2n and 4n cardiomyocyte nuclei, we followed the same cardiomyocyte nuclear isolation protocol with a modification of adding Hoechst (Invitrogen, H3570, 1:2,000) during the secondary antibody incubation to stain DNA. Nuclei without PCM1 antibody staining, or without Hoechst staining were used as negative controls for gating (Figure S4CS4F).

Single Nucleus RNA-seq Library Preparation and Sequencing

Single nucleus RNA-seq libraries were generated using Single Cell 3’ Reagent Kits v2 (10xGenomics) according to the manufacturer’s protocol. Briefly, cells, reagents (10xGenomics, PN-120237), gel beads (10xGenomics, 220104), and partitioning oil (10xGenomics, 220088) were loaded into the Chromium Single Cell A Chip (10xGenomics, 2000019), targeting 2000∼5000 nuclei to be recovered per channel. Chip was loaded to the Chromium Controller (10xGenomics) in the Next Generation Sequencing Core of Children’s Research Institute at UT Southwestern to generate Gel Bead-In-Emulsions (GEMs). After reverse transcription using GEM-RT incubation protocol, GEMs were broken with recovery reagent (10xGenomics, 220016), cDNA was purified with DynaBeads MyOne Silane beads (Thermo Scientific, 37002D), amplified with PCR for 14 cycles, and further purified with SPRIselect reagent (Beckman Coulter, B23318). Quality control of recovered PCR product was performed on a Bioanalyzer (Agilent) with D5000 high sensitivity screentapes (Agilent, 5067–5592), and cDNA yield was measured by Qubit DNA high sensitivity assay (Invitrogen, Q32854). After fragmentation, end-repair, A-tailing and size selection purification, cDNA was ligated with adaptor and subsequently purified with SPRIselect reagents. Libraries were amplified using PCR with sample indexes from Chromium i7 Multiplex kit (10xGenomics, PN-120262), for a total of ∼14 sample index cycles, as estimated from original cDNA yield using Qubit assay. Final PCR products were subjected to double-sided size selection with SPRIselect reagents. Library quality control was performed on the Agilent Bioanalyzer with D1000 screentapes (Agilent, 5067–5582), and library yield was quantified by Qubit DNA high sensitivity assay. Sequencing was performed on an Illumina Nextseq 500 system operated by the Next Generation Sequencing Core of Children’s Research Institute at UT Southwestern using the 150bp high output sequencing kit (Illumina), with the following pair-end sequencing settings: Read1 – 26 cycles, i7 index - 8 cycles, Read2 – 124 cycles.

Pre-processing of snRNA-seq Data

The Cell Ranger Single-Cell Software Suit (https://support.10xgenomics.com/single-cell-geneexpression/software/pipelines/latest/what-is-cell-ranger) was used to perform sample demultiplexing, barcode processing and single-cell 3′ gene counting. The cDNA reads were aligned to the mm10/GRCm38 premRNA reference genome. Only confidently mapped reads with valid barcodes and unique molecular identifiers were used to generate the gene-barcode matrix. Further analyses for quality filtering was performed using the Seurat R package (Butler et al., 2018). Doublets were identified using Scrublet and removed from downstream analysis (Wolock et al., 2019). We further calculated the distribution of detected genes per cell and removed cells in the top 1% quantile or had fewer than 200 detected genes. We also removed cells with more than 25% of the transcripts coming from mitochondrial genes. The co-isolation of mitochondria during the nucleus isolation process is likely due their association with ER. This is consistent with reports from other groups where mitochondrial DNA was detected in single-nucleus RNA-seq (Hu et al., 2018; Lake et al., 2017) as well as ATAC-seq (which also profiles nuclei) (Rickner et al., 2019). In particular, cardiomyocytes are among cell types that have the most mitochondrial content with ∼30% of the cellular volume being filled with mitochondria (Piquereau et al., 2013). However, these mitochondrial transcripts were removed from our data before gene expression analyses, thus they do not affect our results. After quality filtering and removing unwanted cells from the dataset, we normalized the data by the total expression, multiplied by a scale factor of 10,000 and log-transformed the result. To account for variation in the number of genes and UMI detected in each cell, we normalized the scaled expression matrix using the ScaleData function and set the vars.to.regress to nUMI and nGenes.

In total, we analyzed 12,021 cardiomyocyte nuclei from control hearts and 9,716 cardiomyocyte nuclei from injured hearts. We sequenced an average of 48,896 reads with 75% mapping to the genome and a median of 925 genes per nucleus (Figure S1D).

Although the numbers and distribution of detected genes per nucleus were overall comparable across samples, we observed reduced UMI counts, possibly reflecting decreased nuclear mRNA content, and a slight increase of detected transcripts per nucleus in the injured samples, suggesting activated gene transcription after injury (Figures S1D and S1E).

Integrated Analysis of snATAC and snRNA datasets

Cardiac nuclei were isolated using the same procedure as described above. Single cell ATAC-seq libraries were generated using Chromium Single Cell ATAC library & Gel Bead Kit V1.0 (10xGenomics, 1000111) according to the manufacturer’s protocol. Briefly, nuclei were combined with ATAC enzyme to form transposition mix and were incubated for 60 min at 37°3. Transposed nuclei were then loaded to the Chromium Chip E (10xGenomics, 1000086) and run on the Chromium Controller (10xGenomics) for generation of Gel bead in emulsion (GEM). GEM was incubated and broken. GEM reaction mixture was cleaned up using Dynabeads MyOne Silane magnetic beads (Thermo Fisher Scientific, 37002D). DNA was amplified using PCR with specific primers from Chromium i7 Multiplex Kit N Set A (10xGenomics, 1000084), and purified with SPRIselect beads (Beckman Coulter, B23318). Final sequencing library size was determined using D1000 tape on the Agilent Bioanalyzer. The final library yield was measured using the Qubit DNA high sensitivity assay. Next generation sequencing was performed on an Illumina Nextseq 500 system operated by the Next Generation Sequencing Core of Children’s Research Institute at UT Southwestern using the 150bp high output sequencing kit (Illumina), with the following pair-end sequencing settings: Readl – 70 cycles, i7 index – 8 cycles, i5 index – 16 cycles, Read2 – 70 cycles.

The CellRanger ATAC v1.1.0 (https://support.10xgenomics.com/single-cell-atac/software/pipelines/latest/what-is-cell-ranger-atac) was used for primary analysis of the scATAC-seq data. Raw base call (BCL) files were converted to FASTQ files and filtered reads were aligned to the mouse mm10 reference genome. Transposase cut sites in each cell were quantified using barcoded UMIs and 10x cell barcode sequences. Count matrix for accessible chromatin peaks was generated for downstream analysis using the R package Signac (v0.1.6)

To classify cells in snATAC-seq dataset based on cell types identified in snRNA-seq, we followed the data transfer method developed in Seurat package. The detailed procedure can be found at (https://satiialab.orq/seurat/v3.1/atacseq_integration_vignette.html). In brief, LSI (Latent Semantic Indexing (Cusanovich et al., 2015) was used to learn the structure of ATAC-seq data, then FindTransferAnchors function was used to identify anchors between the scATAC-seq dataset and snRNA-seq dataset. The identified anchors were used to transfer the cell type labels learned from snRNA-seq data to snATAC-seq data with function TransferData. We filtered out cells with a prediction score < 0.4. At this threshold > 96% of cells were transferred with a cell type label from snRNA-seq dataset.

Motif enrichment analysis of differential accessible peaks

To identified differential accessible peaks between CM4 and CM1 cell clusters, we used the FindMarkers function within Seurat with parameters min.pct = 0.2, test.use = ‘LR’, and latent.vars = ‘peak_region_fragments’. Peaks that had p valued < 0.05 were designated as differential accessible peaks. We performed motif enrichment analysis using the findMotifsGenome.pl program in HOMER (Version 4.8) and searched for motif enrichment around the 200bp window of the peaks summit.

Integrated Analysis of snRNA Datasets

To account for potential batch effects, we used the IntegrateData function implemented in Seurat V3 which utilizes canonical correlation analysis that identifies a linear combination of features to construct a shared correlation structure and align the global transcriptome across datasets (Butler et al., 2018). Briefly, we performed standard preprocessing (log-normalization), and identified the top 2000 variable features for each individual dataset. We then identified integration anchors using the FindIntegrationAnchors function. We used default parameters and dimension 30 to find anchors. We then passed these anchors to the IntegrateData function to generate integrated Seurat object.

Visualization and Clustering

To visualize the data, we used Uniform Manifold Approximation and Projection (UMAP) to project cells in 2D space on the basis of the aligned canonical correlation analysis (Becht et al., 2018). Aligned canonical correlation vectors (1:30) were used to identify clusters using a shared nearest neighborhood modularity optimization algorithm (Butler et al., 2018). Using the graph-based clustering, we divided cells into 12 clusters using the FindCluster function in Seurat with resolution 0.6. We identified cardiomyocyteclusters based on expression of Myh6 and Tnnt2. We identified seven clusters expressing markers of non-cardiomyocyte populations, which likely represent contaminating non-cardiomyocyte nuclei after PCM1 sorting. These non-cardiomyocyte clusters were removed, and the clustering processes were repeated and resulted in the identification of 8 subpopulations. We merged three clusters that did not show clear molecular distinctions and were called as a single cluster when reducing the cluster resolution to 0.4. We also removed one small cluster that could not be merged based on the biological identity and was composed of too few cells (< 50) to represent a real subpopulation. The resulting datasets were divided into 5 cardiomyocyte subpopulations. Cluster markers were identified using Findmarker function with Wilcoxon rank sum test.

Analysis of 2n/4n Cardiomyocyte snRNA Datasets

We performed an additional analysis of 2n and 4n cardiomyocyte snRNA datasets. After quality filtering, we retained 3890 2n cardiomyocyte nuclei and 5540 4n cardiomyocyte nuclei. The average number of detected genes per cell in 2n and 4n datasets was 1001 and 685, respectively. To map cells in 2n and 4n datasets to the cardiomyocyte populations identified in the previous analysis, we integrated 2n, 4n, and P1-MI-d3 datasets for dimensionality reduction and subspace alignment using the first 15 canonical correlation vectors. Using the graph-based clustering, we initially divided cells into 7 clusters using the FindCluster function in Seurat with resolution 0.2. Two clusters were later identified as contaminating macrophages and endothelial cells based on their expression of C1qa and Fabp5, respectively. The identities of the remaining new clusters were assigned to the corresponding cardiomyocyte population based on 1) de novo cluster identification and 2) their integration of cells in the P1-MI-d3 dataset that had cell type labels transferred from the previous analysis. CM1, CM3, CM4, and CM5 were identified as separate clusters and their identities were assigned based on the transferred identities of P1-MI-d3 cells that were embedded in each cluster. Although the CM2 cluster was not initially identified as a separate cluster, it contains the majority of the CM2 cells from P1-MI-d3 and thus was manually selected as a separate cluster. We visualized the integrated data in UMAP (Seurat V3) and colored cells by their sample of origin (i.e. 2n, 4n, P1-MI-d3, Figure S4G) and cluster identity (Figure S4H and S4I). The proportion of each individual cluster in each sample was calculated and the Chi square test was used for enrichment analysis.

Analysis of Injury Response in CM4 Cells

CM4 cells from P1-sham-d1, P1-MI-d1 and P1-MI-d3 datasets were isolated for further cluster analysis. We recalculated the highly variable genes and performed a principal component analysis for dimensionality reduction. To visualize the data, we used UMAP to project cells in 2D space on the basis of the top 10 significant principal components, and labeled cells by their original sample IDs. We then performed pairwise comparison of transcriptomes between the three stages (P1-Sham-d1 vs P1-MI-d1, P1-Sham-d1 vs P1-MI-d3, and P1-MI-d1 vs P1-MI-d3) using the Wilcoxon rank sum test on the scaled data. Differentially expressed genes with p-value < 0.0001 in any of the comparisons were used to generate a consensus DEG list. We next plotted the z-scores of each gene’s expression in all three stages and performed unsupervised hierarchical clustering, which resulted in the identification of three groups of genes showing distinct expression dynamics (Figure 4B).

Analysis of Injury Response in CM5 Cells

To identify which cardiomyocyte population gives rise to CM5 after injury, we performed pseudotime analysis using Monocle 2 package, as described in the tutorials (http://cole-trapnell-lab.github.io/monocle-release/tutorials/) (Qiu et al., 2017; Trapnell et al., 2014). Up to 1000 cells from each major cardiomyocyte cluster in P8 hearts 1-day after MI dataset (P8-MI-d1) were isolated and used as the input expression matrix for trajectory analysis. We excluded CM4 cells from this analysis due to their low abundance. Differentially expressed genes were identified using the differentialGeneTest function with default settings. DDRTree method was used to reduce the dimension of the dataset. Cells were then ordered along pseudotime and colored by their original identities. Genes significantly changed in expression along the injury trajectory of CM5 were identified using the differentialGeneTest function with default settings. Differential gene analysis was performed by comparing CM5 in P8-MI-d3 to CM5 P8-MI-d3 (p-val <0.0001 and FC = 1.5) to identify genes that showed up-regulated and down-regulated expression in CM5 at 3-days post-MI compared to 1-day post-MI.

For comparison, we also performed pseudotime analysis using Palantir, as described in the tutorials (https://nbviewer.iupyter.org/qithub/dpeerlab/Palantir/blob/master/notebooks/Palantir_sample_notebook.ipynb) (Setty et al., 2019). Briefly, raw data of up to 400 cells from each cardiomyocyte cluster in the P8 1-day after MI dataset (P8-MI-d1) were isolated from Seurat object and used as the input expression matrix. The expression matrix was next normalized by total counts and log transformed. Principal component analysis was performed to determine metagenes which were then used to construct diffusion maps and low dimensional embedding. To determine a trajectory, the start cells were set to cells from the CM1 cluster. The computed trajectory was visualized in t-Distributed Stochastic Neighbor Embedding (t-SNE).

Cell-Cycle Analysis

Cell-cycle analysis for individual cardiomyocytes in each cluster was performed using a published approach ((Kowalczyk et al., 2015), https://satijalab.org/seurat/v2.4/cell_cycle_vignette.html). Briefly, a list of genes associated with either S or G2/M cell-cycle phase was used to assign cell-cycle scores, an index of cell-cycle activity, based on the scaled expression of these genes in each cell.

Imputation of snRNAseq Data and Analysis for Gene Expression Correlation

To correct dropout values in our datasets and to increase statistical power calculating gene expression correlation, we used Markov affinity-based graph imputation of cells (MAGIC) which is based on diffusion geometry to denoise cell count matrix and fill in missing values (van Dijk et al., 2018). For gene expression correlation analysis, MAGIC corrected gene expression matrix of CM4 cells was used to calculate expression correlation between each individual gene and Mki67 or Ccnb1 using the Spearman’s rank correlation coefficient test. Cutoffs using Spearman’s rank correlation coefficient > 0.85 and standard deviation of expression across cells greater than 0.1 were used to choose highly correlated genes Table S3. The highly correlated transcription factors were identified by intersecting with a transcription factor gene list (Zhang et al., 2012).

Upstream Regulator Analysis and Gene Regulatory Network Construction

The identified cell-cycle correlated genes in Table S3 were used as input for upstream regulator analysis. Upstream regulators were predicted using iRegulon (Verfaillie et al., 2014), to identify transcription factor (TF) binding motifs overrepresented within 10 kb genomic sequences centered around TSS for the given gene sets, with the following parameters: motif collection - 10K (9713 PWMs), putative regulatory region - 10kb centered around TSS, motif ranking database - 10 kb centered around TSS (7 species), enrichment score threshold - 3.0, ROC threshold for AUC calculation - 0.03, rank threshold - 5000, maximum FDR on motif similarity - 0.001. The TF-target gene interactions derived from this analysis were visualized using Cytoscape 3.5.1 (Shannon et al., 2003) to construct gene regulatory networks.

Cardiomyocyte in vitro Proliferation Assay

NRVMs were isolated from 1- or 2-day-old Sprague-Dawley rats with the Isolation System for Neonatal Rat/Mouse Cardiomyocytes (Cellutron, nc-6031) according to the manufacturer’s instructions. NRVMs were plated at a density of 1.5×105 cells/well to gelatin-coated 12-well plates and were maintained in DMEM/199 medium (3:1, with 3% FBS), and penicillin-streptomycin for 48 h before adenoviral infection. Adenoviruses for SRF, NFYa, NFYb, NFIc, NFE2L1 and NFE2L2 were generated using the Adeno-X Adenoviral System 3 (Clontech, 632267). Packaging of adenovirus and NRVM in vitro proliferation assays were performed as previously described (Wang et al., 2019).

Cardiomyocyte in vitro Survival Assay

NRVMs were isolated and cultured as above described. NRVMs were first infected with 6–8 μL adenovirus in 1 mL DMEM/199 medium containing 3% FBS per well in a 12-well plate for 48 h and were then treated with 50 uM H2O2, or 100mM peroxynitrite (Cayman Chemical), or 0.1% Methyl methanesulfonate (MMS, Sigma) for 2 h, 1h and 1h, respectively. NRVMs treated with H2O2 were next washed with PBS and incubated with Calcerin AM dye (Thermo Fisher, L3224) for 20 min following the manufacturer’s protocol. Cell viability was then visualized on a Keyence BZ-X700 microscope.

Bulk RNA Sequencing in NRVMs and Data Analysis

NRVMs were collected at 48 h after infection with adenoviruses for GFP, NFYa, and NFE2L1. RNA was extracted using the RNeasy Mini Kit (QIAGEN, 74104) according to the manufacturer’s protocol. An Agilent 2100 TapeStation was used for RNA quality analysis. Stranded mRNA-seq libraries were generated using the KAPA mRNA HyperPrep Kit (Roche, KK8581) following manufacturer’s protocol. Sequencing was performed on an Illumina NextSeq 500 system using a 75-bp, high-output sequencing kit for single-end sequencing.

For data analysis, quality control of RNA-seq data was performed using FastQC Tool (Version 0.11.4). Sequencing reads were aligned to rat Rnor 6.0reference genome using HiSAT2 (Version 2.0.4) with default settings and --rna-strandness F (Kim et al., 2015). After removal of duplicate reads using Samtools v0.1.18 (Li et al., 2009), aligned reads were counted using featurecount (Version 1.6.0) per gene ID (Liao et al., 2014). Differential gene expression analysis was performed with the R package edgeR (Version 3.20.5) using the GLM approach (Robinson et al., 2010). For each comparison, genes with more than 1 CPM (Count Per Million) in at least three samples were considered as expressed and were used for calculating normalization factor. Cutoff values of absolute fold change greater than 1.5 and false discovery rate less than 0.01 were used to select differentially expressed genes between sample group comparisons. Normalized gene CPM values were used to calculate RPKM (Reads Per Kilobase per Million mapped reads) values, which were then used for heatmap plotting.

Immunohistochemistry and TUNEL Assay

Hearts were fixed in 4% paraformaldehyde in PBS (Sigma, D8537), cryopreserved in 10% sucrose/PBS and 18% sucrose/PBS solution sequentially, embedded in O.C.T. Compound (Fisher Healthcare, 23730571), and cryosectioned at 10 μm intervals. For immunofluorescent staining, cryosections were air-dried for 30 min at room temperature and fixed with 4% PFA for 20 min. Section slides were then washed twice with PBS and permeabilized with 0.3% Triton X-100 (Fisher Scientific, BP151–500) in PBS for 10 min. Sections were then blocked in 5% goat serum (Sigma, G9023)/3% BSA/0.025% Triton X-100/PBS blocking solution for 1 h and stained with the indicated primary antibodies prepared in blocking solution at 4 °C overnight using the following dilutions: cTnT (Invitrogen, MA5–12960, 1:200), Xirp2 (Thermo Fisher Scientific, PA5–57072, 1:50), pH3 (Cell Signaling Technology, 9701S, 1:200), TdTomoto (MyBioSource, MBS448092, 1:500). Sections were subsequently washed with 0.025% Triton X-100/PBS three times (5 min for each wash), and incubated with corresponding secondary antibodies conjugated to Alexa Fluor 488, 555 or 647 (Invitrogen) prepared in blocking solution at room temperature for 1.5 h. After secondary antibody incubation, sections were washed with PBS, incubated with Hoechst (Thermo Scientific, 62249, 1:2000) or DAPI (Invitrogen, D1306, 1:2000)/PBS at room temperature for 10 min, and washed twice with PBS before mounting. Images were obtained using a Zeiss LSM 800 confocal microscope. TUNEL assay was performed using Click-iT Plus TUNEL Assay for In Situ Apoptosis Detection kit (Thermo Fisher Scientific, C10619) following manufacturer’s protocol.

Generation and Injection of AAV9

AAV9 vectors expressing TdTomato, NFYa or NFE2L1 driven by the muscle-specific CK8 promoter were cloned by replacing the Cas9 coding sequence of the AAV9-CK8-Cas9 vector with the corresponding coding sequences of TdTomato, NFYa or NFE2L1, and were packaged by the Harvard Medical School/Boston Children’s Hospital Viral Core, as previously described (Amoasii et al., 2017; Martari et al., 2009). C57BL/6 mice were injected with AAV9 at P4 at a titer of 5E13 vg/kg by intraperitoneal injection.

Transthoracic Echocardiography

Cardiac function was evaluated by two-dimensional transthoracic echocardiography on conscious mice using a VisualSonics Vevo2100 imaging system as described previously (Wang et al., 2019). All measurements were performed by an experienced operator blinded to the study.

Quantification of Cardiomyocyte Size and TUNEL Positive Area

Qualification of cardiomyocyte size was performed on transverse sections using ImageJ software (https://imagej.nih.gov/ij/). Wheat Germ Agglutinin (WGA, Invitrogen, W32466) was co-stained with cTnT to mark cellular membranes of cardiomyocytes. The freehand selection tool was used to trace the cellular boundary of Xirp2+ and Xirp2 cardiomyocytes, and the area was measured (Analysis -> Measure). Only the cardiomyocytes at the transverse view were scored. Animals (n=4) were sectioned and two sections from each animal were imaged at 4–6 non-overlapping view sights.

For quantification of the TUNEL signal intensity, heart cross-sectional area was first traced using the freehand selection tool and then measured. The TUNEL signal was measured as integrated density then normalized by the heart section area.

Quantitative Real Time PCR Analysis

Total RNA was extracted from NRVMs using Trizol and reverse transcribed using iScript Reverse Transcription Supermix (Bio-Rad) with random primers. The Quantitative Polymerase Chain Reactions (qPCR) were assembled using KAPA SYBR Fast qPCR Master Mix (KAPA, KK4605). Assays were performed using a 7900HT Fast Real-Time PCR machine (Applied Biosystems). Expression values were normalized to 18S mRNA and were represented as fold change. The following oligonucleotides were ordered from Integrated DNA Technologies to measure transcript abundance: 18S Forward: 5′- ACC GCA GCT AGG AAT AAT GGA - 3′; 18S Reverse: 5′- GCC TCA GTT CCG AAA ACC A - 3′; Ccna2 Forward: 5′- CCC GGA GCC AGA AAA CCA CTG GT - 3′; Ccna2 Reverse: 5′- GCT CAC AAG GAT GGC CCG CAT - 3′; Cdc2 Forward: 5′- TTT CGG CCT TGC CAG AGC GTT - 3′; Cdc2 Reverse: 5′- GTG GAG TAG CGA GCC GAG CC - 3′; Aurka Forward: 5′- GGG TGG TCT GTG CAT GCT CCG - 3′; Aurka Reverse: 5′- GCC TCA AAA GGA GGC ATC CCC ACT A - 3′; Aurkb Forward: 5′- ATG AGC AGC GGA CTG CCA CG- 3′; Aurkb Reverse: 5′- GTC CAG GGT GCC GCA CAT GG- 3′; Cdc20 Forward: 5′- GGC TGG GTT CCC CTG CAG ACA T - 3′; Cdc20 Reverse: 5′- TGG GCA AAG CCA TGG CCT GAG A - 3’.

Pathway and Gene Set Enrichment Analysis

Statistical analysis of gene sets was performed using the Metascape (Zhou et al., 2019). Gene Set Enrichment Analysis was used to determine the enrichment of genes that are more highly expressed either in P1 or P14 hearts as well as embryonic genes in cardiomyocytes at different stages (P1 versus P9) and in different clusters (CM4 versus CM1-CM3) (Subramanian et al., 2005). For generating the embryonic enriched gene set, the following dataset was downloaded from ENCODE portal (https://www.encodeproject.org): ENCSR597UZW, and ENCSR526SEX and analyzed using R package edgeR. Gene Set Enrichment Analysis was also performed to determine the enrichment of Hallmark pathway gene sets (Liberzon et al., 2015) in CM4 versus CM1-CM3.

QUANTIFICATION AND STATISTICAL ANALYSIS

Statistical analyses were performed using GraphPad Prism 8 (GraphPad Software Inc) using statistical tests mentioned in each analysis, with p-value < 0.05 considered significant unless otherwise indicated. All data are displayed as mean ± SD unless otherwise indicated.

DATA AND CODE AVAILABILITY

All data, including sequencing reads and single-nucleus expression matrices have been deposited in NCBI’s Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/). The accession number for the snRNA-seq data reported in this study is GSE130699. The accession number for the bulk tissue RNA-seq in NRVMs reported in this study is GSE142364. The accession number for the scATAC-seq data reported in this study is GSE142365. All datasets generated in this study can be accessed under a GEO superseries with accession number GSE142366.

Supplementary Material

2
3

Supplemental Table 1, related to Figure 2. Analysis of injury response in CM5 cells. This table lists differentially expressed genes by comparing CM5 cardiomyocytes to CM1 cardiomyocytes 1-day after P8 injury and comparing CM5 cardiomyocytes at 3-versus 1-day after P8 injury.

4

Supplemental Table 2, related to Figure 4. Analysis of injury response in CM4 cells. This table lists differentially expressed genes by comparing CM4 cardiomyocytes from regenerative hearts in the absence of injury (sham), 1-day, and 3-days after injury. This gene list was used to identify the three groups of genes that were temporally responsive to injury in Figure 4B.

5

Supplemental Table 3, related to Figure 4. Analysis of genes correlated with cell-cycle gene expression. This table lists genes whose expression positively correlates with cell cycle genes Mki67 and Ccnb1 in CM4 cardiomyocytes.

KEY RESOURCES TABLE.

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
Anti-PCM1 antibody produced in rabbit Sigma-Aldrich Cat#HPA023374; RRID:AB_1855073
Cardiac Troponin T Monoclonal Antibody (13-11) Invitrogen Cat#MA5-12960; RRID:AB_11000742
XIRP2 Polyclonal Antibody Thermo Fisher Scientific Cat#PA5-57072; RRID:AB_2649681
Phospho-Histone H3 (Ser10) Antibody Cell Signaling Technology Cat#9701S; RRID:AB_331535
Aurora B Antibody Abcam Ab2254
Goat tdTomato Polyclonal Antibody MyBioSource Cat#MBS448092
Goat anti-Rabbit IgG (H+L) Cross-Adsorbed Secondary Antibody, Alexa Fluor 647 Invitrogen Cat#A21244; RRID:AB_141663
Goat anti-Mouse IgG (H+L) Cross-Adsorbed Secondary Antibody, Alexa Fluor 488 Invitrogen Cat#A-11001; RRID:AB_2534069
Goat anti-Mouse IgG (H+L) Cross-Adsorbed Secondary Antibody, Alexa Fluor 647 Invitrogen Cat#A-21235; RRID:AB_2535804
Goat anti-Rabbit IgG (H+L) Cross-Adsorbed Secondary Antibody, Alexa Fluor 555 Invitrogen Cat#A-21428; RRID:AB_2535849
Donkey anti-Goat IgG (H+L) Cross-Adsorbed Secondary Antibody, Alexa Fluor 555 Invitrogen Cat#A-21432; RRID:AB_2535853
Bacterial and Virus Strains
Stellar Competent Cells Takara Cat#636766
Adeno-SRF This paper N/A
Adeno-NFYa This paper N/A
Adeno-NFYb This paper N/A
Adeno-NFIc This paper N/A
Adeno-NFE2L1 This paper N/A
Adeno-NEF2L2 This paper N/A
AAV9-TdTomato Wang et al., 2019 N/A
AAV9-NFYa This paper N/A
AAV9-NFE2L1 This paper N/A
Biological Samples
Chemicals, Peptides, and Recombinant Proteins
Dulbecco’s Modified Eagle’s Medium - high glucose Sigma-Aldrich Cat#D5796
Medium 199, Earle’s Salts Gibco Cat#11150-059
Fetal Bovine Serum Gemini Bio Products Cat#100-106
Penicillin-Streptomycin Sigma-Aldrich Cat#P0781
cOmplete Protease Inhibitor Cocktail Roche Cat#11697498001
RNaseOUT Recombinant Ribonuclease Inhibitor Invitrogen Cat#10777019
Hoechst 33342 Invitrogen Cat#H3570
DAPI (4′,6-Diamidino-2-Phenylindole, Dihydrochloride) Invitrogen Cat#D1306
Wheat Germ Agglutinin, Alexa Fluor™ 647 Conjugate Invitrogen Cat#W32466
Peroxynitrite Cayman Chemical Cat#81565
Methyl methanesulfonate Sigma-Aldrich Cat#129925
Hydrogen peroxide Millipore Sigma Cat#H1009
Critical Commercial Assays
Chromium Single Cell 3’ Library & Gel Bead Kit v2 10xGenomics Cat#PN-120237
Chromium Single Cell A Chip Kit 10xGenomics Cat#PN-120236
Chromium i7 Multiplex Kit 10xGenomics Cat#PN-120262
Chromium i7 Multiplex Kit N Set A 10xGenomics Cat# 1000084
Chromium Chip E Single Cell ATAC Kit 10xGenomics Cat#1000086
Chromium Single Cell ATAC Library & Gel Bead Kit V 1.0 10xGenomics Cat# 1000111
D5000 High Sensitivity DNA ScreenTape Agilent Cat#5067-5592
D1000 ScreenTape Agilent Cat#5067-5582
Qubit dsDNA HS Assay Kit Invitrogen Cat#Q32854
Neomyt Kit Cellutron Cat#nc-6031
Adeno-X Adenoviral System 3 Clontech Cat#632267
LIVE/DEAD Viability/Cytotoxicity Kit, for mammalian cells Invitrogen Cat#L3224
RNeasy Mini Kit Qiagen Cat#74104
KAPA mRNA HyperPrep Kit KAPA Cat#KK8581
Click-iT™ Plus TUNEL Assay for In Situ Apoptosis Detection, Alexa Fluor™ 647 dye Invitrogen Cat#C10619
iScript Reverse Transcription Supermix for RT-qPCR Bio-Rad Cat#1708840
KAPA SYBR Fast qPCR Master Mix KAPA Cat#KK4605
Deposited Data
Raw and processed data This paper GEO: GSE130699
Experimental Models: Cell Lines
Adeno-X 293 cells Clontech Cat#632271
Experimental Models: Organisms/Strains
Mouse: ICR/CD-1 Charles River N/A
Mouse: C57BL/6J The Jackson Laboratory N/A
Rat: Sprague Dawley Envigo N/A
Oligonucleotides
Primer: 18S Forward: 5′- ACC GCA GCT AGG AAT AAT GGA - 3′ This paper N/A
Primer: 18S Reverse: 5′- GCC TCA GTT CCG AAA ACC A - 3′ This paper N/A
Primer: Ccna2 Forward: 5′- CCC GGA GCC AGA AAA CCA CTG GT - 3′ This paper N/A
Primer: Ccna2 Reverse: 5′- GCT CAC AAG GAT GGC CCG CAT - 3′ This paper N/A
Primer: Cdc2 Forward: 5′- TTT CGG CCT TGC CAG AGC GTT – 3′ This paper N/A
Primer: Cdc2 Reverse: 5′- GTG GAG TAG CGA GCC GAG CC - 3′ This paper N/A
Primer: Aurka Forward: 5′- GGG TGG TCT GTG CAT GCT CCG - 3′ This paper N/A
Primer: Aurka Reverse: 5′- GCC TCA AAA GGA GGC ATC CCC ACT A - 3′ This paper N/A
Primer: Aurkb Forward: 5′- ATG AGC AGC GGA CTG CCA CG- 3′ This paper N/A
Primer: Aurkb Reverse: 5′- GTC CAG GGT GCC GCA CAT GG- 3′ This paper N/A
Primer: Cdc20 Forward: 5′- GGC TGG GTT CCC CTG CAG ACA T - 3′ This paper N/A
Primer: Cdc20 Reverse: 5′- TGG GCA AAG CCA TGG CCT GAG A - 3′ This paper N/A
Recombinant DNA
Software and Algorithms
Cell Ranger v2.1 10xGenomics https://support.10xgenomics.com/single-cell-gene-expression/software/pipelines/latest/what-is-cell-ranger
Seurat v3.0 Stuart et al., 2019 https://satijalab.org/seurat/
Monocle 2 Qiu et al., 2017; Trapnell et al., 2014 http://cole-trapnell-lab.github.io/monocle-release/
Palantir Setty et al.,2019 https://github.com/dpeerlab/Palantir
MAGIC van Dijk et al., 2018 https://github.com/KrishnaswamyLab/MAGIC
Scrublet Wolock et al., 2019 https://github.com/AllonKleinLab/scrublet
iRegulon Janky et al., 2014 http://iregulon.aertslab.org/index.html
Cytoscape v3.5.1 Shannon et al., 2003 https://cytoscape.org/
FastQC v0.11.4 Babraham Bioinformatics https://www.bioinformatics.babraham.ac.uk/projects/fastqc/
HiSAT2 v2.0.4 Kim et al., 2015 https://ccb.jhu.edu/software/hisat2/index.shtml
Samtools v0.1.18 Li et al., 2009 http://samtools.sourceforge.net/
Featurecount v1.6.0 Liao et al., 2014 http://subread.sourceforge.net/
edgeR v3.20.5 Robinson et al., 2010 https://bioconductor.org/packages/release/bioc/html/edgeR.html
ImageJ NIH https://imagej.nih.gov/ij/index.html
Metascape Zhou et al., 2019 http://metascape.org
GSEA Subramanian et al., 2005 http://software.broadinstitute.org/gsea/index.jsp
GraphPad Prism 8 GraphPad Software Inc
Other

Highlights.

  1. Neonatal cardiomyocytes (CMs) in mice are heterogeneous.

  2. Immature CMs enriched in regenerative hearts enter the cell-cycle upon injury.

  3. Defined transcriptome changes in regenerating CMs in response to injury.

  4. NFYa and NFE2L1 exert proliferative and protective functions in CMs.

ACKNOWLEDGMENTS

We thank Jose Cabrera for graphics, Drs. Jian Xu, Xin Liu and Yoon Jung Kim from the Children’s Research Institute at the University of Texas Southwestern Medical Center for performing the Illumina sequencing, and Dr. Xiang Luo for help with isolating neonatal rat ventricular cardiomyocytes. This work was supported by grants from the NIH (AR-067294, HL-130253, HL-138426, and HD-087351), the Fondation Leducq Transatlantic Networks of Excellence in Cardiovascular Research and the Robert A. Welch Foundation (grant 1-0025 to E.N.O.). Z.W. was supported by a predoctoral fellowship from the American Heart Association and the Harry S. Moss Heart Trust (19PRE34380436).

Footnotes

DECLARATION OF INTERESTS

The authors declare no competing interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCE

  1. Aguirre A, Montserrat N, Zacchigna S, Nivet E, Hishida T, Krause MN, Kurian L, Ocampo A, Vazquez-Ferrer E, Rodriguez-Esteban C et al. (2014). In vivo activation of a conserved microRNA program induces mammalian heart regeneration. Cell stem cell 15, 589–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Alkass K, Panula J, Westman M, Wu TD, Guerquin-Kern JL, and Bergmann O (2015). No Evidence for Cardiomyocyte Number Expansion in Preadolescent Mice. Cell 163, 1026–1036. [DOI] [PubMed] [Google Scholar]
  3. Ames EG, Lawson MJ, Mackey AJ, and Holmes JW (2013). Sequencing of mRNA identifies re-expression of fetal splice variants in cardiac hypertrophy. J Mol Cell Cardiol 62, 99–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Amoasii L, Long C, Li H, Mireault AA, Shelton JM, Sanchez-Ortiz E, McAnally JR, Bhattacharyya S, Schmidt F, Grimm D, et al. (2017). Single-cut genome editing restores dystrophin expression in a new mouse model of muscular dystrophy. Science translational medicine 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Bartelt A, Widenmaier SB, Schlein C, Johann K, Goncalves RLS, Eguchi K, Fischer AW, Parlakgul G, Snyder NA, Nguyen TB, et al. (2018). Brown adipose tissue thermogenic adaptation requires Nrf1-mediated proteasomal activity. Nature medicine 24, 292–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Becht E, McInnes L, Healy J, Dutertre CA, Kwok IWH, Ng LG, Ginhoux F, and Newell EW (2018). Dimensionality reduction for visualizing single-cell data using UMAP. Nature biotechnology. [DOI] [PubMed] [Google Scholar]
  7. Biswas M, and Chan JY (2010). Role of Nrf1 in antioxidant response element-mediated gene expression and beyond. Toxicology and applied pharmacology 244, 16–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bonuccelli G, Peiris-Pages M, Ozsvari B, Martinez-Outschoorn UE, Sotgia F, and Lisanti MP (2017). Targeting cancer stem cell propagation with palbociclib, a CDK4/6 inhibitor: Telomerase drives tumor cell heterogeneity. Oncotarget 8, 9868–9884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bungartz G, Land H, Scadden DT, and Emerson SG (2012). NF-Y is necessary for hematopoietic stem cell proliferation and survival. Blood 119, 1380–1389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Butler A, Hoffman P, Smibert P, Papalexi E, and Satija R (2018). Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nature biotechnology 36, 411–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Chamberlain JS, Jaynes JB, and Hauschka SD (1985). Regulation of creatine kinase induction in differentiating mouse myoblasts. Molecular and cellular biology 5, 484–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Cox EJ, and Marsh SA (2014). A systematic review of fetal genes as biomarkers of cardiac hypertrophy in rodent models of diabetes. PloS one 9, e92903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Cui M, Wang Z, Bassel-Duby R, and Olson EN (2018). Genetic and epigenetic regulation of cardiomyocytes in development, regeneration and disease. Development (Cambridge, England) 145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cusanovich DA, Daza R, Adey A, Pliner HA, Christiansen L, Gunderson KL, Steemers FJ, Trapnell C, and Shendure J (2015). Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science (New York, NY) 348, 910–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Fisher DJ, Heymann MA, and Rudolph AM (1980). Myocardial oxygen and carbohydrate consumption in fetal lambs in utero and in adult sheep. The American journal of physiology 238, H399–405. [DOI] [PubMed] [Google Scholar]
  16. Fleming JD, Pavesi G, Benatti P, Imbriano C, Mantovani R, and Struhl K (2013). NF-Y coassociates with FOS at promoters, enhancers, repetitive elements, and inactive chromatin regions, and is stereo-positioned with growth-controlling transcription factors. Genome research 23, 1195–1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Gabisonia K, Prosdocimo G, Aquaro GD, Carlucci L, Zentilin L, Secco I, Ali H, Braga L, Gorgodze N, Bernini F, et al. (2019). MicroRNA therapy stimulates uncontrolled cardiac repair after myocardial infarction in pigs. Nature 569, 418–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Gemberling M, Karra R, Dickson AL, and Poss KD (2015). Nrg1 is an injury-induced cardiomyocyte mitogen for the endogenous heart regeneration program in zebrafish. eLife 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Habib N, Avraham-Davidi I, Basu A, Burks T, Shekhar K, Hofree M, Choudhury SR, Aguet F, Gelfand E, Ardlie K, et al. (2017). Massively parallel single-nucleus RNA-seq with DroNc-seq. Nature methods 14, 955–958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Hamel FG, Upward JL, Siford GL, and Duckworth WC (2003). Inhibition of proteasome activity by selected amino acids. Metabolism: clinical and experimental 52, 810–814. [DOI] [PubMed] [Google Scholar]
  21. Hu P, Liu J, Zhao J, Wilkins BJ, Lupino K, Wu H, and Pei L (2018). Single-nucleus transcriptomic survey of cell diversity and functional maturation in postnatal mammalian hearts. Genes Dev 32, 1344–1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Iyer H, Collins JJ 3rd, and Newmark PA (2016). NF-YB Regulates Spermatogonial Stem Cell Self-Renewal and Proliferation in the Planarian Schmidtea mediterranea. PLoS genetics 12, e1006109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Jaynes JB, Chamberlain JS, Buskin JN, Johnson JE, and Hauschka SD (1986). Transcriptional regulation of the muscle creatine kinase gene and regulated expression in transfected mouse myoblasts. Molecular and cellular biology 6, 2855–2864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Johnsen O, Skammelsrud N, Luna L, Nishizawa M, Prydz H, and Kolsto AB (1996). Small Maf proteins interact with the human transcription factor TCF11/Nrf1/LCR-F1. Nucleic acids research 24, 4289–4297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Kim D, Langmead B, and Salzberg SL (2015). HISAT: a fast spliced aligner with low memory requirements. Nature methods 12, 357–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Koopman WJ, Nijtmans LG, Dieteren CE, Roestenberg P, Valsecchi F, Smeitink JA, and Willems PH (2010). Mammalian mitochondrial complex I: biogenesis, regulation, and reactive oxygen species generation. Antioxidants & redox signaling 12, 1431–1470. [DOI] [PubMed] [Google Scholar]
  27. Kowalczyk MS, Tirosh I, Heckl D, Rao TN, Dixit A, Haas BJ, Schneider RK, Wagers AJ, Ebert BL, and Regev A (2015). Single-cell RNA-seq reveals changes in cell cycle and differentiation programs upon aging of hematopoietic stem cells. Genome research 25, 1860–1872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Lake BB, Codeluppi S, Yung YC, Gao D, Chun J, Kharchenko PV, Linnarsson S, and Zhang K (2017). A comparative strategy for single-nucleus and single-cell transcriptomes confirms accuracy in predicted cell-type expression from nuclear RNA. Sci Rep 7, 6031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, and Durbin R (2009). The Sequence Alignment/Map format and SAMtools. Bioinformatics (Oxford, England) 25, 2078–2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Li Y, He L, Huang X, Bhaloo SI, Zhao H, Zhang S, Pu W, Tian X, Li Y, Liu Q, et al. (2018). Genetic Lineage Tracing of Nonmyocyte Population by Dual Recombinases. Circulation 138, 793–805. [DOI] [PubMed] [Google Scholar]
  31. Liao Y, Smyth GK, and Shi W (2014). featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics (Oxford, England) 30, 923–930. [DOI] [PubMed] [Google Scholar]
  32. Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, and Tamayo P (2015). The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell systems 1, 417–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Lopaschuk GD, Collins-Nakai RL, and Itoi T (1992). Developmental changes in energy substrate use by the heart. Cardiovascular research 26, 1172–1180. [DOI] [PubMed] [Google Scholar]
  34. Lundin C, North M, Erixon K, Walters K, Jenssen D, Goldman AS, and Helleday T (2005). Methyl methanesulfonate (MMS) produces heat-labile DNA damage but no detectable in vivo DNA double-strand breaks. Nucleic acids research 33, 3799–3811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Mahmoud AI, Porrello ER, Kimura W, Olson EN, and Sadek HA (2014). Surgical models for cardiac regeneration in neonatal mice. Nature protocols 9, 305–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Maity SN, and de Crombrugghe B (1998). Role of the CCAAT-binding protein CBF/NF-Y in transcription. Trends in biochemical sciences 23, 174–178. [DOI] [PubMed] [Google Scholar]
  37. Maliken BD, and Molkentin JD (2018). Undeniable Evidence That the Adult Mammalian Heart Lacks an Endogenous Regenerative Stem Cell. Circulation 138, 806–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Martari M, Sagazio A, Mohamadi A, Nguyen Q, Hauschka SD, Kim E, and Salvatori R (2009). Partial rescue of growth failure in growth hormone (GH)-deficient mice by a single injection of a double-stranded adeno-associated viral vector expressing the GH gene driven by a muscle-specific regulatory cassette. Human gene therapy 20, 759–766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Martinez-Outschoorn UE, Peiris-Pages M, Pestell RG, Sotgia F, and Lisanti MP (2017). Cancer metabolism: a therapeutic perspective. Nature reviews Clinical oncology 14, 11–31. [DOI] [PubMed] [Google Scholar]
  40. Menheniott TR, Woodfine K, Schulz R, Wood AJ, Monk D, Giraud AS, Baldwin GS, Moore GE, and Oakey RJ (2008). Genomic imprinting of Dopa decarboxylase in heart and reciprocal allelic expression with neighboring Grb10. Molecular and cellular biology 28, 386–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Mohamed TMA, Ang YS, Radzinsky E, Zhou P, Huang Y, Elfenbein A, Foley A, Magnitsky S, and Srivastava D (2018). Regulation of Cell Cycle to Stimulate Adult Cardiomyocyte Proliferation and Cardiac Regeneration. Cell 173, 104–116.e112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Monroe TO, Hill MC, Morikawa Y, Leach JP, Heallen T, Cao S, Krijger PHL, de Laat W, Wehrens XHT, Rodney GG, et al. (2019). YAP Partially Reprograms Chromatin Accessibility to Directly Induce Adult Cardiogenesis In Vivo. Dev Cell 48, 765–779.e767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Morikawa Y, Zhang M, Heallen T, Leach J, Tao G, Xiao Y, Bai Y, Li W, Willerson JT, and Martin JF (2015). Actin cytoskeletal remodeling with protrusion formation is essential for heart regeneration in Hippo-deficient mice. Science signaling 8, ra41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Nakada Y, Canseco DC, Thet S, Abdisalaam S, Asaithamby A, Santos CX, Shah AM, Zhang H, Faber JE, Kinter MT, et al. (2017). Hypoxia induces heart regeneration in adult mice. Nature 541, 222–227. [DOI] [PubMed] [Google Scholar]
  45. Ohtsuji M, Katsuoka F, Kobayashi A, Aburatani H, Hayes JD, and Yamamoto M (2008). Nrf1 and Nrf2 play distinct roles in activation of antioxidant response element-dependent genes. The Journal of biological chemistry 283, 33554–33562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Pacher P, Beckman JS, and Liaudet L (2007). Nitric oxide and peroxynitrite in health and disease. Physiol Rev 87, 315–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Pangonyte D, Stalioraityte E, Ziuraitiene R, Kazlauskaite D, Palubinskiene J, and Balnyte I (2008). Cardiomyocyte remodeling in ischemic heart disease. Medicina (Kaunas, Lithuania) 44, 848–854. [PubMed] [Google Scholar]
  48. Piquereau J, Caffin F, Novotova M, Lemaire C, Veksler V, Garnier A, Ventura-Clapier R, and Joubert F (2013). Mitochondrial dynamics in the adult cardiomyocytes: which roles for a highly specialized cell? Front Physiol 4, 102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Pjanic M, Schmid CD, Gaussin A, Ambrosini G, Adamcik J, Pjanic P, Plasari G, Kerschgens J, Dietler G, Bucher P, et al. (2013). Nuclear Factor I genomic binding associates with chromatin boundaries. BMC genomics 14, 99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA, Olson EN, and Sadek HA (2011). Transient regenerative potential of the neonatal mouse heart. Science (New York, NY) 331, 1078–1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Porrello ER, Mahmoud AI, Simpson E, Johnson BA, Grinsfelder D, Canseco D, Mammen PP, Rothermel BA, Olson EN, and Sadek HA (2013). Regulation of neonatal and adult mammalian heart regeneration by the miR-15 family. Proceedings of the National Academy of Sciences of the United States of America 110, 187–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Puente BN, Kimura W, Muralidhar SA, Moon J, Amatruda JF, Phelps KL, Grinsfelder D, Rothermel BA, Chen R, Garcia JA, et al. (2014). The oxygen-rich postnatal environment induces cardiomyocyte cell-cycle arrest through DNA damage response. Cell 157, 565–579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Qiu X, Mao Q, Tang Y, Wang L, Chawla R, Pliner HA, and Trapnell C (2017). Reversed graph embedding resolves complex single-cell trajectories. Nature methods 14, 979–982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Quaife-Ryan GA, Sim CB, Ziemann M, Kaspi A, Rafehi H, Ramialison M, El-Osta A, Hudson JE, and Porrello ER (2017). Multi-Cellular Transcriptional Analysis of Mammalian Heart Regeneration. Circulation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Rickner HD, Niu SY, and Cheng CS (2019). ATAC-seq Assay with Low Mitochondrial DNA Contamination from Primary Human CD4+ T Lymphocytes. J Vis Exp. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Robinson MD, McCarthy DJ, and Smyth GK (2010). edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics (Oxford, England) 26, 139–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Sancho P, Barneda D, and Heeschen C (2016). Hallmarks of cancer stem cell metabolism. British journal of cancer 114, 1305–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Setty M, Kiseliovas V, Levine J, Gayoso A, Mazutis L, and Pe’er D (2019). Characterization of cell fate probabilities in single-cell data with Palantir. Nature biotechnology 37, 451–460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, and Ideker T.J.G.r. (2003). Cytoscape: a software environment for integrated models of biomolecular interaction networks. 13, 2498–2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Singh BN, Koyano-Nakagawa N, Gong W, Moskowitz IP, Weaver CV, Braunlin E, Das S, van Berlo JH, Garry MG, and Garry DJ (2018). A conserved HH-Gli1-Mycn network regulates heart regeneration from newt to human. Nature communications 9, 4237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM 3rd, Hao Y, Stoeckius M, Smibert P, and Satija R (2019). Comprehensive Integration of Single-Cell Data. Cell 177, 1888–1902.e1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, et al. (2005). Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proceedings of the National Academy of Sciences of the United States of America 102, 15545–15550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Sun X, and Nunes SS (2017). Bioengineering Approaches to Mature Human Pluripotent Stem Cell-Derived Cardiomyocytes. Front Cell Dev Biol 5, 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Suraweera A, Munch C, Hanssum A, and Bertolotti A (2012). Failure of amino acid homeostasis causes cell death following proteasome inhibition. Molecular cell 48, 242–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Taegtmeyer H, Sen S, and Vela D (2010). Return to the fetal gene program: a suggested metabolic link to gene expression in the heart. Annals of the New York Academy of Sciences 1188, 191–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Tao G, Kahr PC, Morikawa Y, Zhang M, Rahmani M, Heallen TR, Li L, Sun Z, Olson EN, Amendt BA, et al. ((2016). Pitx2 promotes heart repair by activating the antioxidant response after cardiac injury. Nature 534, 119–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Tian Y, Liu Y, Wang T, Zhou N, Kong J, Chen L, Snitow M, Morley M, Li D, Petrenko N, et al. (2015). A microRNA-Hippo pathway that promotes cardiomyocyte proliferation and cardiac regeneration in mice. Science translational medicine 7, 279ra238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, Lennon NJ, Livak KJ, Mikkelsen TS, and Rinn JL (2014). The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nature biotechnology 32, 381–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. van Dijk D, Sharma R, Nainys J, Yim K, Kathail P, Carr AJ, Burdziak C, Moon KR, Chaffer CL, Pattabiraman D et al. (2018). Recovering Gene Interactions from Single-Cell Data Using Data Diffusion. Cell 174, 716–729.e727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Verfaillie A, Imrichová H, Van de Sande B, Standaert L, Christiaens V, Hulselmans G, Herten K, Sanchez MN, Potier D, and Svetlichnyy D.J.P.c.b. (2014). iRegulon: from a gene list to a gene regulatory network using large motif and track collections. 10, e1003731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. von Gise A, Lin Z, Schlegelmilch K, Honor LB, Pan GM, Buck JN, Ma Q, Ishiwata T, Zhou B, Camargo FD, et al. (2012). YAP1, the nuclear target of Hippo signaling, stimulates heart growth through cardiomyocyte proliferation but not hypertrophy. Proceedings of the National Academy of Sciences of the United States of America 109, 2394–2399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Wang Z, Cui M, Shah AM, Ye W, Tan W, Min YL, Botten GA, Shelton JM, Liu N, Bassel-Duby R, et al. (2019). Mechanistic basis of neonatal heart regeneration revealed by transcriptome and histone modification profiling. Proceedings of the National Academy of Sciences of the United States of America 116, 18455–18465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Widenmaier SB, Snyder NA, Nguyen TB, Arduini A, Lee GY, Arruda AP, Saksi J, Bartelt A, and Hotamisligil GS (2017). NRF1 Is an ER Membrane Sensor that Is Central to Cholesterol Homeostasis. Cell 171, 1094–1109.e1015. [DOI] [PubMed] [Google Scholar]
  74. Wisneski JA, Gertz EW, Neese RA, Gruenke LD, Morris DL, and Craig JC (1985). Metabolic fate of extracted glucose in normal human myocardium. The Journal of clinical investigation 76, 1819–1827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Wolock SL, Lopez R, and Klein AM (2019). Scrublet: Computational Identification of Cell Doublets in Single-Cell Transcriptomic Data. Cell systems 8, 281–291.e289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Wu H, Kirita Y, Donnelly EL, and Humphreys BD (2019). Advantages of Single-Nucleus over Single-Cell RNA Sequencing of Adult Kidney: Rare Cell Types and Novel Cell States Revealed in Fibrosis. Journal of the American Society of Nephrology : JASN 30, 23–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Xin M, Kim Y, Sutherland LB, Murakami M, Qi X, McAnally J, Porrello ER, Mahmoud AI, Tan W, Shelton JM, et al. (2013a). Hippo pathway effector Yap promotes cardiac regeneration. Proceedings of the National Academy of Sciences of the United States of America 110, 13839–13844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Xin M, Olson EN, and Bassel-Duby R (2013b). Mending broken hearts: cardiac development as a basis for adult heart regeneration and repair. Nature reviews Molecular cell biology 14, 529–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Yasmin W, Strynadka KD, and Schulz R (1997). Generation of peroxynitrite contributes to ischemia-reperfusion injury in isolated rat hearts. Cardiovascular research 33, 422–432. [DOI] [PubMed] [Google Scholar]
  80. Zhang HM, Chen H, Liu W, Liu H, Gong J, Wang H, and Guo AY (2012). AnimalTFDB: a comprehensive animal transcription factor database. Nucleic acids research 40, D144–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Zhou X, Liao WJ, Liao JM, Liao P, and Lu H (2015). Ribosomal proteins: functions beyond the ribosome. Journal of molecular cell biology 7, 92–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, Benner C, and Chanda SK (2019). Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nature communications 10, 1523. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

2
3

Supplemental Table 1, related to Figure 2. Analysis of injury response in CM5 cells. This table lists differentially expressed genes by comparing CM5 cardiomyocytes to CM1 cardiomyocytes 1-day after P8 injury and comparing CM5 cardiomyocytes at 3-versus 1-day after P8 injury.

4

Supplemental Table 2, related to Figure 4. Analysis of injury response in CM4 cells. This table lists differentially expressed genes by comparing CM4 cardiomyocytes from regenerative hearts in the absence of injury (sham), 1-day, and 3-days after injury. This gene list was used to identify the three groups of genes that were temporally responsive to injury in Figure 4B.

5

Supplemental Table 3, related to Figure 4. Analysis of genes correlated with cell-cycle gene expression. This table lists genes whose expression positively correlates with cell cycle genes Mki67 and Ccnb1 in CM4 cardiomyocytes.

Data Availability Statement

All data, including sequencing reads and single-nucleus expression matrices have been deposited in NCBI’s Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/). The accession number for the snRNA-seq data reported in this study is GSE130699. The accession number for the bulk tissue RNA-seq in NRVMs reported in this study is GSE142364. The accession number for the scATAC-seq data reported in this study is GSE142365. All datasets generated in this study can be accessed under a GEO superseries with accession number GSE142366.

RESOURCES