Abstract
The growth factor Neuregulin‐1 (NRG‐1) regulates myocardial growth and is currently under clinical investigation as a treatment for heart failure. Here, we demonstrate in several in vitro and in vivo models that STAT5b mediates NRG‐1/EBBB4‐stimulated cardiomyocyte growth. Genetic and chemical disruption of the NRG‐1/ERBB4 pathway reduces STAT5b activation and transcription of STAT5b target genes Igf1, Myc, and Cdkn1a in murine cardiomyocytes. Loss of Stat5b also ablates NRG‐1‐induced cardiomyocyte hypertrophy. Dynamin‐2 is shown to control the cell surface localization of ERBB4 and chemical inhibition of Dynamin‐2 downregulates STAT5b activation and cardiomyocyte hypertrophy. In zebrafish embryos, Stat5 is activated during NRG‐1‐induced hyperplastic myocardial growth, and chemical inhibition of the Nrg‐1/Erbb4 pathway or Dynamin‐2 leads to loss of myocardial growth and Stat5 activation. Moreover, CRISPR/Cas9‐mediated knockdown of stat5b results in reduced myocardial growth and cardiac function. Finally, the NRG‐1/ERBB4/STAT5b signaling pathway is differentially regulated at mRNA and protein levels in the myocardium of patients with pathological cardiac hypertrophy as compared to control human subjects, consistent with a role of the NRG‐1/ERBB4/STAT5b pathway in myocardial growth.
Keywords: cardiomyocyte hyperplasia, cardiomyocyte hypertrophy, dynamin, NRG‐1–ErbB pathway, signal transducer and activator of transcription
Subject Categories: Cardiovascular System, Signal Transduction
STAT5b regulates hypertrophic and hyperplastic growth signaling via the NRG‐1/ERBB4 pathway in cardiomyocytes at adult and embryonic stages. Patients with pathological cardiac hypertrophy show perturbed STAT5b signaling.
Introduction
Neuregulin‐1 (NRG‐1) has been implicated as a central regulator of both hypertrophic and hyperplastic cardiomyocyte growth (Zhao et al, 1998; Baliga et al, 1999; Bersell et al, 2009). Neuregulin‐1 is synthesized in cardiac endothelial cells (Lemmens et al, 2006; Hedhli et al, 2011) and released from the cell surface of endothelial cells mainly by the ADAM‐17 protease (Montero et al, 2000). The release of NRG‐1 has been reported to be induced by angiogenic factors through the VEGFR2 receptor (Kivelä et al, 2019). Neuregulin‐1 activates ERBB receptor tyrosine kinases by inducing their dimerization and subsequential transphosphorylation, and is known to signal through the ERBB2/ERBB4 heterodimer in the myocardium (Gassmann et al, 1995; Lee et al, 1995; Meyer & Birchmeier, 1995). Of these two ERBB receptors, only ERBB4 can directly bind NRG‐1. ERBB2 is activated by dimerizing with the NRG‐1‐bound ERBB4 receptor (Plowman et al, 1993). Genetic knockout or chemical inhibition of NRG‐1, ERBB2, or ERBB4 in mouse models has been shown to lead to a distinct heart phenotype during embryogenesis characterized by dilated ventricles, thin ventricular walls and loss of trabeculae (Gassmann et al, 1995; Lee et al, 1995; Meyer & Birchmeier, 1995), and to dilated cardiomyopathy in the adulthood (Crone et al, 2002; Özcelik et al, 2002; García‐Rivello et al, 2005). A similar heart phenotype has also been observed in zebrafish embryos expressing a mutant erbb2 transgene or a protein trap for the Erbb4 ligand Nrg2a, as well as in larvae treated with chemical inhibitor of the Erbb2/Erbb4 heterodimer (Liu et al, 2010; Rasouli & Stainier, 2017). Moreover, overexpression of Nrg‐1 or constitutively active ERBB2 has been reported to lead to cardiomegaly in adult zebrafish and in neonatal and adult mice models, respectively (D'uva et al, 2015; Gemberling et al, 2015). After injury, NRG‐1 promotes regeneration of the heart by inducing proliferation of cardiomyocytes both in adult mice and in adult zebrafish (Bersell et al, 2009; Gemberling et al, 2015). In several injury mouse models, administration of recombinant NRG‐1 has attenuated cardiac injury (Odiete et al, 2012; Mendes‐Ferreira et al, 2013). Therefore, after successful phase II trials (Gao et al, 2010; Jabbour et al, 2011), NRG‐1 is currently tested in phase III clinical trials as a treatment for heart failure (Gao et al, 2018; Zensun Sci. & Tech. Co., Ltd., 2018) .
The NRG‐1/ERBB pathway has been shown to activate PI3K/AKT, MAPK, and SRC/FAK pathways in cardiomyocytes (Baliga et al, 1999; Fukazawa et al, 2003; Kuramochi et al, 2006; Jie et al, 2012; Cai et al, 2016; Pentassuglia et al, 2016), none of which has been demonstrated to directly control NRG‐1/ERBB‐mediated cardiomyocyte growth in in vitro or in vivo settings. Instead, the PI3K/AKT pathway has been affiliated with cardiomyocyte survival, DNA synthesis and glucose uptake (Fukazawa et al, 2003; Bersell et al, 2009; Jie et al, 2012; Pentassuglia et al, 2016), the MAPK pathway with protein synthesis and sarcomere organization (Baliga et al, 1999), and the SRC/FAK pathway with cytoskeletal remodeling (Kuramochi et al, 2006).
In our previous study, we discovered that the transcription factor STAT5b is selectively activated by the JM‐b isoform of ERBB4 (Vaparanta et al, 2022), the ERBB4 isoform preferentially expressed by cardiomyocytes (Fig EV1A) (Elenius et al, 1997; Wang et al, 2021). Of the two STAT5 genes, STAT5b is also the major STAT5 gene expressed in the heart both at mRNA and at protein level (Fig EV1B). Since chemical inhibition of STAT5 has previously been shown to attenuate myocardial growth induced by angiotensin II (Jin et al, 2022) or by pressure overload in mice (Kimura et al, 2018), we set out to explore the role of STAT5b in NRG1/ERBB4‐mediated myocardial growth. As NRG‐1 has been reported to induce both hypertrophic growth in neonatal and adult cardiomyocytes as well as hyperplastic growth in embryonic cardiomyocytes (Zhao et al, 1998), we employed both adult and neonatal murine and embryonic zebrafish models to investigate the role of STAT5b in NRG‐1/ERBB4‐induced cardiac growth. Clinical human samples were examined to investigate whether the hypothesized signaling pathway is active also in human in states of increased myocardium growth. The analyses validated in both in vitro and in vivo models demonstrated that the NRG‐1 receptor ERBB4, the ERBB4‐activated intracellular signaling molecule STAT5b, and the ERBB4 trafficking‐regulator dynamin‐2 mediate both hypertrophic and hyperplastic NRG‐1‐induced cardiomyocyte growth. In addition, we discovered altered signaling of the NRG‐1/ERBB4/STAT5b/dynamin‐2 pathway in the myocardia of patients suffering from pathological cardiac hypertrophy.
Results
NRG‐1/ERBB4 pathway controls STAT5b signaling in vivo in adult murine heart
To explore the role of STAT5b in ERBB4‐mediated hypertrophic myocardial growth, mice expressing the soluble ectodomain (ECD) of ERBB4 in the heart as a result of infection with an adeno‐associated virus of serotype 9 carrying the mouse ERBB4 ECD insert (AAV9‐mERBB4ECD) were utilized. The ERBB4 ECD traps ERBB4 ligands, such as NRG‐1, leading to downregulation of ERBB4 signaling in the cardiomyocytes (Kivelä et al, 2019).
First, the effect of AAV9‐mERBB4ECD treatment on cardiomyocyte size was assessed to learn whether reduction in ERBB4‐sensitive signaling is associated with cardiomyocyte growth. The cardiomyocyte thickness was determined from heart sections (Fig 1A and B). As expected, the average cardiomyocyte thickness was lesser in the AAV9‐mERBB4ECD‐treated mice as compared to the controls (Fig 1A and B).
The activation of STAT5b by phosphorylation in the key tyrosine residue 699 leads to STAT5b dimerization and consequential accumulation into the nucleus at the target gene transcription sites. To explore the role of ErbB4 ligands in STAT5b activation in cardiomyocytes, the nuclear accumulation of STAT5b in the AAV9‐mERBB4ECD and AAV9‐control treated hearts was addressed by immunostaining (Fig 1C and D). The nuclear accumulation of STAT5b was found to be reduced in the AAV9‐mERBB4ECD‐treated hearts as compared to the controls, indicating a role for ERBB4 ligands in STAT5b activation in cardiomyocytes (Fig 1C and D).
The effect of the AAV9‐mERBB4ECD‐treatment on the transcription of known STAT5b target genes was further assessed with real‐time RT‐PCR (Fig 1E). The treatment with AAV9‐mERBB4ECD significantly decreased the levels of Igf1 and Myc transcripts (Fig 1E), both of which are encoded by known STAT5b target genes that have been found to promote cardiac hypertrophy in vivo (Delaughter et al, 1999; Nosaka et al, 1999; Lord et al, 2000; Zhong et al, 2006; Basham et al, 2008; Rotwein, 2012). In addition to Igf1 and Myc, the transcription of a third STAT5b target gene, Cdkn1a (Yu et al, 2010), was similarly decreased.
NRG‐1/ERBB4 pathway controls STAT5b signaling in vitro in neonatal murine cardiomyocytes
To explore the STAT5b‐dependency of NRG‐1/ERBB4‐mediated cardiomyocyte growth, cardiomyocytes from neonatal mice were isolated. As expected, the primary cardiomyocytes expressed ERBB4 JM‐b more than the JM‐a isoform (Fig EV1C) were positive for cardiomyocyte lineage‐specific markers myosin‐heavy‐chain and tropomyosin (Fig EV1D), and exhibited sporadic contractions in the culture plate. To assess the effect of STAT5b knockdown on NRG‐1‐induced cardiomyocyte hypertrophy, the isolated cardiomyocytes were infected with lentiviral particles containing plasmids encoding Stat5b‐targeting shRNAs or control shRNA (Fig EV1E) and stimulated or not with NRG‐1 for 2 days. The cardiomyocytes were stained with an antibody recognizing the heavy chain of a skeletal and cardiac myocyte specific myosin II, analyzed with immunofluorescence, and the cross‐sectional area of the cardiomyocytes was quantified from confocal images taken in plane with the plasma membrane. The Stat5b knockdown significantly reduced the hypertrophic response induced by NRG‐1 (Fig 1F and G), suggesting that NRG‐1‐stimulated hypertrophy is at least partially dependent on STAT5b.
To address the role of ERBB4 as a receptor for NRG‐1 in mediating the hypertrophic response, Erbb4 was knocked down in mouse primary cardiomyocytes by RNA interference (Fig EV1F–H). The treatment with Erbb4‐targeting shRNAs significantly reduced the nuclear accumulation of STAT5b, as well as the cross‐sectional area of cardiomyocytes (Fig 1H and I). To confirm that the transcription of STAT5b target genes in cardiomyocytes was STAT5b‐ and ERBB4‐dependent, expression of Igf1, Myc, and Cdkn1a was analyzed in cells subjected to Stat5b or Erbb4 knockdown after stimulating the cells for 30 min with NRG‐1. Real‐time RT‐PCR analyses demonstrated that both Stat5b‐ and Erbb4‐targeting shRNAs significantly downregulated the expression of all three STAT5b target genes (Fig 1J). Taken together, these results demonstrate that the loss of NRG‐1/ERBB4 signaling correlates with the loss of STAT5b signaling and reduced cardiomyocyte size in murine cardiomyocytes.
Dynamin‐2 controls subcellular ERBB4 localization and STAT5b activation in neonatal murine cardiomyocytes
Mass spectrometry‐derived ERBB4 interactome indicated that dynamin‐2 (encoded by DNM2 gene) was one of the prominent interaction partners of ERBB4 (Fig EV2A). Experimentation with proximity ligation assay (PLA) suggested that the heart‐specific ERBB4 JM‐b isoform was more prone to associate with dynamin‐2 than the alternative isoform ERBB4 JM‐a (Fig EV2B and C). The interaction between dynamin‐2 and ERBB4 may have implications for heart biology, since the loss of dnm2 has been, similarly to the loss of Nrg1 and Erbb4, associated with the development of heart failure (Li et al, 2013). Previous research also indicates that the dynamin GTPases control vesicular endocytotic and ER‐to‐golgi trafficking. Of the three genes encoding dynamins, DNM2 is most abundantly expressed in the heart both at mRNA and at protein level (Fig EV2D; Li et al, 2013).
To test whether ERBB4 interacts with Dynamin‐2 in cardiomyocytes, the association between the two proteins was addressed with PLA in primary murine cardiomyocytes. In addition, to analyze whether the interaction was dependent on the Dynamin‐2 GTPase activity, the cardiomyocytes were cultured in the presence or absence of the Dynamin GTPase inhibitor dynasore. ERBB4 and Dynamin‐2 indeed associated with each other in both control‐ and dynasore‐treated cardiomyocytes (Fig 2A and B). Treatment with dynasore, however, significantly affected the subcellular localization of the associations between endogenously expressed ERBB4 and Dynamin‐2. Dynasore selectively reduced the associations at the cell surface and periphery of the cardiomyocytes (Fig 2C and D).
To further explore the effect of Dynamin inhibition on ERBB4 location, ERBB4 isoforms expressed in MCF‐7 cells were analyzed for colocalization with the plasma membrane marker Na‐K ATPase after treatment with dynasore or the control buffer. Dynasore significantly reduced the colocalization of both ERBB4 JM‐a and JM‐b isoforms with Na‐K ATPase at the cell surface, indicating that the cell surface localization of ERBB4 was specifically dependent on the GTPase activity of Dynamin‐2 (Fig EV2E and F).
To explore whether the effect of dynamin‐2 on ERBB4 localization was reflected in the downstream signaling via STAT5b, ERBB4 JM‐b was overexpressed in mammary epithelial cells treated with dynasore or DNM2‐targeting siRNAs. Both treatments abolished ERBB4 overexpression‐promoted phosphorylation of the activating residue of STAT5b (Fig EV2G and H) and dynasore treatment additionally disrupted the co‐precipitation of ERBB4 JM‐b with STAT5b (Fig EV2I). Moreover, nuclear accumulation of STAT5b was reduced in cardiomyocytes treated with dynasore (Fig 2E and F), and both a chemical ERBB inhibitor AG1478 as well as dynasore significantly reduced the NRG‐1‐stimulated expression of STAT5b target genes (Fig 2G).
To control that the inhibition of the GTPase activity of dynamin‐2 also affected the NRG‐1‐stimulated hypertrophic response, dynasore‐treated cardiomyocytes were stimulated with NRG‐1 for 2 days and analyzed for cross‐sectional area. The area was significantly reduced by dynasore (Fig 2H and I). Taken together, the results indicate that Dynamin‐2 regulates subcellular ERBB4 localization and, consequently, NRG‐1‐stimulated STAT5b activation and cardiomyocyte growth.
NRG‐1 activates Stat5 in embryonic zebrafish model of hyperplastic myocardial growth in vivo
In addition to hypertrophic growth, NRG‐1/ERBB signaling has been reported to induce hyperplastic myocardial growth in zebrafish models and in embryonic mice (Zhao et al, 1998; Liu et al, 2010; Gemberling et al, 2015). To explore the role of STAT5b also in the context of NRG‐1‐induced hyperplastic growth, human recombinant NRG‐1 or the control substance (BSA) was injected into the pericardial cavity of zebrafish embryos at 2 days post fertilization (dpf). The embryos were fixed at 4 dpf and stained with a myosin heavy chain antibody to visualize the myocardium. The cross‐sectional ventricular area and ventricle thickness were measured from confocal images taken from anterior cross sections of the ventricles. The NRG‐1 injection induced hyperplastic cardiomyocyte growth as indicated by the increase in cross‐sectional ventricular area but not in ventricular wall thickness (Fig 3A–D). This was further confirmed by assessing the ratio of the amount of nuclei in the ventricle to the ventricular area of the NRG‐1‐injected and BSA‐injected embryos (Fig 3A and B). An increase in the number of nuclei in the myocardium of NRG‐1‐injected embryos was observed indicating proliferative growth.
The effect of NRG‐1 injection to the cardiac function of the zebrafish embryos was further assessed with live imaging (Fig 3E and F, Movies EV1 and EV2). The ventricles of the NRG‐1‐injected and BSA‐injected zebrafish demonstrated similar ejection fractions indicating that the NRG‐1 induced hyperplastic growth was physiological.
To assess the signaling pathways activated by the NRG‐1 injections, western analyses were performed (Fig 3G and H). Unsurprisingly, the Pi3k/Akt and Erk pathways were activated with NRG‐1. In addition, the Stat5 pathway was activated as indicated by the increased phosphorylation in the activating residue of the zebrafish Stat5 (Fig 3G and H). While the phosphospecific antibody does not differentiate between the zebrafish Stat5a and Stat5b, it is of note that both zebrafish Stat5a and Stat5b share more sequence similarity with human STAT5b than with human STAT5a. These data indicate that in a zebrafish model of NRG‐1‐induced hyperplastic myocardial growth, Stat5 is activated by the Nrg‐1/Erbb4 pathway.
Chemical inhibition of the Nrg‐1/Erbb4 pathway results in reduced myocardial growth and Stat5 activation in zebrafish embryos
To assess the effect of the Nrg‐1/Erbb4 pathway inhibition on the hyperplastic myocardial growth and Stat5 activation in vivo, zebrafish embryos were treated at 2 dpf for 2 days with DMSO or the ERBB inhibitors AG1478, lapatinib, or gefitinib. The function and the morphology of the ventricles was examined with live imaging (Fig 4A). The ERBB inhibitor AG1478 has been reported to inhibit ERBB1, ERBB2, and ERBB4 (Egeblad et al, 2001), lapatinib ERBB1 and ERBB2 (Xia et al, 2002), and gefitinib ERBB1 (Anderson et al, 2001). As expected, treatment with both inhibitors reported to inhibit the ERBB2/ERBB4 heterodimer in the myocardium significantly reduced the cross‐sectional area of the ventricles, while treatment with the ERBB1 inhibitor gefitinib had no significant effect (Fig 4A and B). Treatment with AG1478 also reduced the thickness of the ventricular wall. The ejection fraction of all ERBB inhibitor‐treated zebrafish, however, was retained although the AG1478‐ and lapatinib‐treated ventricles displayed abnormal contractions (Movies [Link], [Link]).
Consistent with the role of STAT5b downstream of ERBB4 activation in the mouse models (Fig 1C and D, H and I), treatment with the inhibitors targeting the ERBB2/ERBB4 heterodimer, but not the ERBB1 inhibitor, reduced Stat5 phosphorylation in zebrafish embryos (Fig 4C and D). Phosphorylation of Erk, however, was significantly reduced only by treatment with the ERBB1 inhibitor gefitinib. Signal for phospho‐Akt, surprisingly, was induced by lapatinib and was not significantly affected by treatment with other ERBB inhibitors.
To examine the role of dynamin‐2 in the hyperplastic growth and Stat5 activation, the embryos were treated at 2 dpf for 2 days with or without dynasore. The dynasore treatment resulted in both reduced ventricular area and ventricular wall thickness as compared to the control treatment (Fig 4E and F). The ejection fraction was, however, unaltered in the dynasore‐treated ventricles indicating that the reduced myocardial growth did not lead to heart failure (Fig 4G and H, Movies EV7 and EV8).
Finally, the effect of dynasore on the activation of Stat5, Akt, and Erk signaling was analyzed (Fig 4I and J). Stat5 was significantly less active in the dynasore‐treated zebrafish embryos as compared to the control, while no effect was observed for either Akt or Erk signaling. Taken together, the results indicate that the loss of myocardial growth due to inhibition of the Nrg‐1/Erbb4/Dynamin‐2 pathway correlates with the loss of Stat5, but not of Akt or Erk activation.
CRISPR/Cas9‐mediated knockdown of the zebrafish stat5b gene results in reduced myocardial growth and heart failure in zebrafish embryos
To explore whether knockdown of the stat5b gene was sufficient to reduce myocardial growth, zebrafish embryos were injected with the recombinant CRISPR/Cas9 enzyme and control‐ or stat5b‐targeting guide RNAs (gRNA) at one‐cell stage. The efficacy of the stat5b knockdown was confirmed with genomic sequencing, real‐time RT‐PCR (Fig EV3A and B) and with immunofluorescence staining of whole‐mount embryos (Fig 5A and B). The cross‐sectional area of the ventricles of stat5b gRNA‐injected 4 dpf embryos was significantly smaller when compared to the control gRNA‐injected embryos (Fig 5A and B), indicating reduced myocardial growth.
The ejection fraction of the ventricles of stat5b gRNA‐injected zebrafish embryos was also significantly reduced compared with the control gRNA‐injected embryos (Fig 5C and D; Movies EV9 and EV10) suggesting heart failure. These results indicate that the downregulation of Stat5b alone is sufficient to disrupt the normal growth and function of the myocardium.
The NRG‐1/ERBB4/STAT5b signaling pathway is dysregulated in the myocardium of patients with pathological cardiac hypertrophy
Finally, to explore the role of the NRG‐1/ERBB4/STAT5b signaling pathway in the context of pathological hypertrophic myocardial growth in human, the expression signature of the NRG‐1/ERBB4/STAT5b signaling pathway and activation of STAT5b were investigated in clinical samples representing different types of pathological cardiac hypertrophy or normal heart tissue. First, pre‐existing transcriptome datasets were subjected to principal component analysis to observe whether control subjects and patients suffering from hypertrophic cardiomyopathy could be segregated based on the expression levels of the genes in the NRG‐1/ERBB4/STAT5b signaling pathway (Figs 6A and EV4A). The details of the datasets can be accessed through the Gene Expression Omnibus database (Edgar et al, 2002; Barrett et al, 2013) with the GSE prefix identifiers indicated in the figure legends of Fig 6 and in the Fig EV5. The dimensionality reduction of the expression level of the NRG‐1/ERBB4/STAT5b signaling pathway genes indeed was able to significantly cluster cases with pathological cardiac hypertrophy and those without into separate groups in three independent datasets. The successful segregation indicates that the NRG‐1/ERBB4/STAT5b signaling pathway genes are consistently differentially regulated in hypertrophic vs. normal cardiac tissue. The transcripts of MYC, IGF1 and NRG1 genes were most differentially regulated in the NRG‐1/ERBB4/STAT5b signaling pathway, as indicated by the principal component weights (Fig EV4B).
The role of NRG‐1/ERBB4/STAT5b signaling pathway in pressure overload‐induced pathological cardiac hypertrophy was also investigated from datasets representing cardiac transcriptomes of mice that had undergone transverse aortic constriction surgery (TAC) or sham surgery. The dimensionality reduction in the expression level of the NRG‐1/ERBB4/STAT5b signaling pathway genes led to a statistically significant clustering of the TAC‐ and sham‐treated mice into separate groups in three independent datasets (Figs 6B and EV4C). The successful segregation indicates that NRG‐1/ERBB4/STAT5b signaling pathway genes are also differentially regulated in pressure overload‐induced cardiac hypertrophy. The principal component weights suggested that Igf1 and Dnm2 were most differentially regulated by the TAC vs. sham surgery (Fig EV4D).
The activation of STAT5b in control subjects and patients suffering from pathological cardiac hypertrophy due to aortic stenosis, alcoholic cardiomyopathy, or idiopathic cardiomyopathy was investigated with immunohistochemistry. Formalin‐fixed paraffin‐embedded (FFPE) sections from cardiac left ventricle were acquired from the Auria Biobank (University Hospital of Turku, Finland). Both heart weight and cardiomyocyte size were greater in the samples representing pathological cardiac hypertrophy (Fig EV4E). The heart sections were stained with an antibody that recognizes the phosphorylated state of the activating residue of STAT5b, and the immunosignal was quantified from cardiomyocytes in representative randomly selected regions. The specificity of the immunosignal was controlled in FFPE samples treated with insulin or control or STAT5 targeting siRNAs (Fig EV5A–D). The signal of the phosphorylated STAT5 in cardiomyocyte nuclei was significantly greater in the cases representing pathological cardiac hypertrophy as compared to the controls (Fig 6C and D; only five cases from both cohorts visualized) indicating that STAT5b activation is upregulated in pathologically hypertrophic myocardium in human. Taken together, these results suggest that the NRG‐1/ERBB4/STAT5b pathway is dysregulated in mammalian pathological cardiac hypertrophy.
Discussion
A recent review identified the effect of NRG‐1 on cardiomyocyte growth as one of the translationally relevant but poorly understood aspects of cardiovascular research, and called for further research on the underlying molecular mechanisms (De Keulenaer et al, 2019). Here, we focused on the signaling pathway of NRG‐1 in primary cardiomyocytes and in the heart tissue of mouse and embryonic zebrafish to elucidate the molecular mechanisms of NRG‐1‐mediated cardiomyocyte growth. The NRG‐1 receptor ERBB4, the ERBB4‐activated signal transducer STAT5b, and dynamin‐2, which was discovered to control the subcellular localization of ErbB4, were identified as key effectors controlling both hyperplastic cardiomyocyte growth during embryogenesis as well as hypertrophic cardiomyocyte growth at neonatal and adult stages (Fig 7). The expression of two transcription factors known as essential regulators of cardiac growth, IGF‐1 and MYC, was controlled by NRG1/ERBB4/STAT5b signaling in neonatal and adult cardiomyocytes. We also found evidence of activation of the NRG1/ERBB4/STAT5b signaling in human samples displaying increased myocardial growth.
In this study, the role of STAT5b in NRG‐1‐induced cardiomyocyte growth was examined across species. While cardiac NRG‐1 signaling has mostly been analyzed in mouse models, the reports of zebrafish embryos have produced similar findings. In both species, the genetic or chemical disruption of genes along the NRG‐1/ERBB pathway leads to the loss of cardiac trabeculation and attenuated growth of the ventricular wall (Gassmann et al, 1995; Lee et al, 1995; Meyer & Birchmeier, 1995; Liu et al, 2010; Rasouli & Stainier, 2017). Few reports have suggested that in zebrafish embryos, cardiac trabeculation is not dependent on the Nrg‐1 ligand, but on another Erbb4 ligand, Nrg‐2a, which has similar expression pattern in endothelial cells as NRG‐1 in mice (Samsa et al, 2016; Rasouli & Stainier, 2017). Overexpression of the zebrafish nrg1 gene, however, has induced cardiomyocyte proliferation in adult zebrafish (Gemberling et al, 2015). Similarly, our findings indicated that the administration of human recombinant NRG‐1 did induce hyperplastic cardiomyocyte growth in zebrafish embryos. It is of note that the molecular mechanisms regulating cardiac function and growth in mice and zebrafish are expected to be different in adulthood when zebrafish cardiomyocytes retain their regenerative capability through cardiomyocyte hyperplasia in much larger quantities than in adult mice (Poss et al, 2002).
The ERK pathway has been established as one of the main pathways regulating hypertrophic cardiomyocyte growth downstream of several stimuli (Gallo et al, 2019). The NRG‐1/ERBB pathway promotes ERK1/2 phosphorylation in cardiomyocytes; therefore, NRG‐1‐induced cardiomyocyte hypertrophy has been attributed to ERK signaling (Baliga et al, 1999; Fukazawa et al, 2003; Kuramochi et al, 2006). A recent report, however, suggests that the activation of ERK signaling alone is not sufficient to confer NRG‐1‐mediated cardiomyocyte hypertrophy (Wang et al, 2021). This is consistent with several reports where treatment with an anti‐ERBB2 antibody or ERBB2‐targeting siRNAs has been unable to reduce the phosphorylation of ERK1/2 in cardiomyocytes (Sawyer et al, 2002; Kuramochi et al, 2006; Wang et al, 2021). Treatment with ERBB4 shRNA has drastically reduced hypertrophic growth but similarly has only led to a slight reduction of ERK1/2 phosphorylation in cardiomyocytes (Wang et al, 2021). The conditional double knockout of Erk1/Erk2 in mice leads to dilated cardiomyopathy resembling the loss of the genes along the NRG‐1/ERBB2/ERBB4 pathway (Kehat et al, 2011). The histology of the dilated ventricles, however, is markedly different between the conditional double Erk1/Erk2 knockout model and the conditional Nrg1/Erbb2/Erbb4 pathway knockout models. The cardiomyocytes in the double Erk1/Erk2 knockout model are elongated and show a loss of concentric hypertrophic response (Kehat et al, 2011), while the cardiomyocytes in the Erbb2/Erbb4 knock‐out models are hypertrophic (Özcelik et al, 2002; García‐Rivello et al, 2005). The asymmetry in the cardiomyocyte morphology of the Erk1/Erk2 vs Erbb2/Erbb4 conditional knockout model hearts and the lack of response in ERK phosphorylation after downregulation of ERBB2 or ERBB4 are consistent with our observations here of NRG‐1/ERBB4 additionally activating STAT5b in murine in vitro and in vivo models of hypertrophic growth. Indeed, the NRG‐1/ERBB4‐mediated hypertrophic cardiomyocyte growth was reduced by STAT5b knock‐down.
The NRG‐1‐mediated hyperplastic growth in turn has been attributed to PI3K/AKT signaling in adult cardiomyocytes based on an experiment, in which the expression of PTEN, the negative regulator of PI3K signaling, reduced DNA synthesis (Bersell et al, 2009). According to the results presented here, the activation of Stat5b was consistently modulated by intervention of the Nrg‐1/Erbb4 signaling pathway, while the phosphorylation of Erk and Akt was significantly modulated by NRG‐1 injections but not by chemical inhibition of the Nrg‐1/Erbb4 pathway in embryonic zebrafish models of hyperplastic myocardium growth. These observations suggest that proliferation of cardiomyocytes is STAT5b dependent during embryogenesis. Whether STAT5b contributes to the proliferation of adult cardiomyocytes remains to be addressed.
The NRG‐1/ERBB pathway is not the only pathway that has been associated with myocardial growth. Several other pathways have been identified as essential for cardiomyocyte proliferation, hypertrophy, and dedifferentiation, which allows for adult cardiomyocytes to re‐enter the cell cycle. These pathways have been extensively reviewed in previous reports and include, the FGF‐1, IGF‐1, Ang‐II, Hippo, and Wnt signaling cascades (Nakamura & Sadoshima, 2018; Bongiovanni et al, 2021). The role of several miRNAs and transcription factors similarly have been established in controlling the expression of genes with central roles in regulating cell cycle and growth of cardiomyocytes (Lock et al, 2018; Bongiovanni et al, 2021). As of yet it is, however, unclear which of these signals are the master regulators of cardiomyocyte growth. In a previous report, the ErbB2‐mediated activation of STAT5 was sufficient to override the Hippo pathway‐induced withdrawal from cell cycle (Hirai et al, 2017), and our findings indicate that the expression of IGF‐1 in neonatal and adult cardiomyocytes is dependent on NRG‐1, ERBB4 and STAT5b. Thus, it is plausible that the NRG‐1/ERBB4/STAT5b pathway is an upstream regulator of the IGF‐1 and Hippo pathways in the myocardium. Any further role of the NRG‐1/ERBB4‐activated STAT5b in the interplay of all these growth and cell cycle signals that converge to induce myocardial growth remains to be elucidated.
Similar to NRG‐1 and ERBB4 (De Keulenaer et al, 2019), STAT5b has previously been proposed to have role in the molecular pathogenesis of heart failure. Indeed, a polymorphism in the exon 1 of the STAT5B gene has been associated with the risk for dilated cardiomyopathy (Peng et al, 2012). Here, we identified differential regulation of the NRG‐1/ERBB4/DNM2/STAT5b signaling in clinical samples of pathological cardiac hypertrophy independent of the etiology. This is consistent with previous observations that chemical inhibition of STAT5 reduces pathological cardiac hypertrophy in in vivo mouse models (Kimura et al, 2018; Jin et al, 2022). Pathological cardiac hypertrophy is a compensatory response that can precede heart failure and ERBB4 expression has been reported to be downregulated in cases in which cardiac hypertrophy has advanced to heart failure (Rohrbach et al, 1999, 2005b). In our zebrafish model, the CRISPR/Cas9‐mediated knockdown of stat5b also led to reduced ejection fraction indicative of heart failure. Further experimentation is, however, needed to understand the role of STAT5b in pathological cardiac hypertrophy and its progression to heart failure.
STAT5b has been suggested to mediate the cardioprotective effect of remote ischemic preconditioning in a murine model of ischemic cardiac injury and in clinical samples (Heusch et al, 2012; Wu et al, 2017; Chen et al, 2018). The results presented here further support the therapeutic potential of STAT5b by introducing STAT5b as one of the downstream effectors of NRG‐1 in the heart. NRG‐1 signaling has been found to attenuate cardiac injury in several animal models including, for example, ischemia, doxorubicin, metabolic stress, and pacing‐induced cardiac injury models (Odiete et al, 2012; Mendes‐Ferreira et al, 2013). The cardioprotective effect of NRG‐1 has been attributed to increased PI3K/AKT signaling (Odiete et al, 2012; Mendes‐Ferreira et al, 2013). Interestingly in the murine model of ischemic cardiac injury and remote ischemic preconditioning the cardioprotective PI3K/AKT signaling was activated downstream of STAT5b (Chen et al, 2018). Consistently, chemical inhibition of STAT5 has been shown to downregulate PI3K/AKT signaling in a mouse model of pressure overload induced cardiac hypertrophy (Kimura et al, 2018). Together these observations putatively suggest that the NRG‐1‐induced cardioprotective PI3K/AKT signaling is dependent on STAT5b. Whether the cardioprotective effect of NRG‐1 truly is dependent on STAT5b and whether, for example, targeting the deactivating phosphatases of STAT5b (Yu et al, 2000; Rigacci et al, 2008; Huang et al, 2012) or repressors of STAT5b‐induced transcription (Nakajima et al, 2001; Sefat‐E‐Khuda et al, 2004; Martens et al, 2005) in the myocardium could serve as an approach with therapeutic potential in heart failure remains to be formally addressed. In accordance with clinical applicability, administration of NRG‐1 and the protein product of the STAT5b target gene IGF1 has demonstrated success in attenuating dilated cardiomyopathy in clinical trials (Welch et al, 2002; Liu et al, 2006; Komamura, 2017).
In addition to STAT5b, the membrane‐remodeling GTPase dynamin‐2 was identified as a regulator of NRG‐1/ERBB4 signaling promoting cardiomyocyte growth. Inhibition of dynamin GTPase activity reduced ERBB4 at the cell surface as well as STAT5b activation and NRG‐1‐induced cardiomyocyte and myocardial growth in zebrafish embryos. Expression of dynamin‐2 has previously been reported to be decreased in heart failure in isopretenol‐induced rat heart failure model and in clinical samples (Li et al, 2013). It has also been demonstrated that decreased dynamin‐2 expression leads to cardiomyocyte apoptosis by stimulating Ca2+ influx from the sarcolemma (Li et al, 2013). Interestingly, several reports also indicate a role for NRG‐1/ERBB4 signaling in regulating cardiomyocyte apoptosis (Rohrbach et al, 2005a; Jie et al, 2012; Cohen et al, 2014), suggesting that cardiomyocyte apoptosis induced by dynamin‐2 deficiency (Li et al, 2013) could be mediated by the loss of NRG‐1/ERBB4 signaling. Consistently, STAT5b has been shown to regulate the expression of the anti‐apoptotic Bcl2 and Bcl2‐XL in the myocardium (Chen et al, 2018). During induced cardiac hypertrophy, no significant level of apoptosis has been detected in the myocardium (Romano & Ceci, 2020), which is consistent with the observations here of elevated STAT5b activation in states of increased hypertrophic myocardial growth.
Materials and Methods
The details of the reagents, experimental models, instruments, and tools used are provided in the Reagents and Tools table. The sequences for the oligonucleotides utilized in this study are provided in Table EV1.
Reagents and Tools table
Reagent/resource | Reference or source | Identifier or catalog number |
---|---|---|
Experimental models | ||
HEK293T cell line | ATCC | CRL‐3216 |
MDA‐MB‐468 pBABE Puro ErbB4 JM‐a cell line | Described earlier (Sundvall et al, 2012) | |
MDA‐MB‐468 pBABE Puro ErbB4 JM‐b cell line | Described earlier (Sundvall et al, 2012) | |
MDA‐MB‐468 pBABE Puro empty vector cell line | Described earlier (Sundvall et al, 2012) | |
MCF‐7 cell line | ECACC | 86012803 |
HC11 cell line | ATCC | CRL‐3062 |
C57BL/6JOlaHsd (Mus musculus) | Envigo Harlan | C57BL/6JOlaHsd |
Primary cultures from 1 to 3 days old male wildtype NMRI (Mus musculus) | Janvier labs | RjHan:NMRI |
Casper strain (roy, mitfa) (Danio rerio) | Described earlier (White et al, 2008) | |
Patient samples | Auria Biobank | |
Recombinant DNA | ||
pMD2.G (env) | Addgene | 12259 |
pMDLg/pRRE (gag, pol, RRE) | Addgene | 12251 |
pRSV‐Rev (Rev) | Addgene | 12253 |
pc DNA 3.1 ERBB4 JM‐a CYT‐2 –HA Hyg(+) | Described earlier (Maatta et al, 2005) | |
pc DNA 3.1 ERBB4 JM‐b CYT‐2 –HA Hyg(+) | Described earlier (Maatta et al, 2005) | |
pc DNA 3.1 Hyg(+) | Described earlier (Maatta et al, 2005) | |
Control shRNA: sh‐scramble plasmid | Addgene | 1864 |
Antibodies | ||
Mouse monoclonal anti‐β‐actin (AC‐74), 1:10,000 (W) | Sigma | A5441 |
Mouse monoclonal anti‐Akt (pan) (40D4), 1:1,000 (W) | Cell Signaling | 2020S |
Rabbit polyclonal anti‐Phospho‐Akt (Ser473) (D9E) XP®, 1:1,000 (W) | Cell Signaling | 4060 |
Goat polyclonal anti‐Dynamin II (C‐18), 1:1,000 (W), 1:100 (PLA) | Santa Cruz | sc‐6400 |
Rabbit monoclonal anti‐ERBB4 (E200), 1:1,000 (W), 1:100 (PLA), 1 μg/1 mg protein (IP) | Abcam | ab32375 |
Mouse monoclonal anti‐ERBB4 (HFR‐1), 1:50 (IF), 1:100 (PLA), 1 μg/1 mg protein (IP) | Abcam | ab19391 |
Rabbit monoclonal p44/42 MAPK (Erk1/2), 1:1,000 (W) | Cell Signaling | 9102 |
Rabbit polyclonal P‐p44/42 MAPK (T202/Y204), 1:1,000 (W) | Cell Signaling | 9101 |
Mouse monoclonal anti‐myosin heavy chain (MF‐20), 1:1,000 (W), 1:50–200 (IF) | DSHB | MF 20 |
Rabbit polyclonal anti‐STAT5b, 1:500 (W), 1:20 (IF) | R&D | AF1584 |
Rabbit polyclonal anti‐STAT5b, 1:500 (W), 1:50 (IF) | Sigma | SAB2102320 |
Rabbit polyclonal anti‐Phospho‐STAT5 (Tyr694), 1:500 (W) | Cell Signaling | 9351 |
Recombinant Rabbit Anti‐STAT5 (phospho Y694) antibody [E208], 1:1,000 (IHC) | Abcam | ab32364 |
Mouse monoclonal TPM1 antibody, mouse tropomyosin, striated muscle (CH‐1) | DSHB | CH‐1 |
Rabbit anti‐goat IgG‐HRP, 1:10,000 (W) | Santa Cruz | sc‐2768 |
Goat anti‐mouse IgG‐HRP, 1:10,000 (W) | Santa Cruz | sc‐2005 |
Goat anti‐rabbit IgG‐HRP, 1:10,000 (W) | Santa Cruz | sc‐2004 |
IRDye® 680RD Donkey anti‐Mouse IgG (H + L), 1:15,000 (W) | LI‐COR | 925‐68072 |
IRDye® 800CW Donkey anti‐Mouse IgG (H + L), 1:15,000 (W) | LI‐COR | 925‐32212 |
IRDye® 680RD Donkey anti‐Rabbit IgG (H + L), 1:15,000 (W) | LI‐COR | 926‐68073 |
IRDye® 800CW Donkey anti‐Rabbit IgG (H + L), 1:15,000 (W) | LI‐COR | 925‐32213 |
Goat anti‐Mouse IgG (H+L) Highly Cross‐Adsorbed Secondary Antibody, Alexa Fluor 488, 1:100–500 (IF) | Invitrogen | A‐11029 |
Goat anti‐Rabbit IgG (H+L) Highly Cross‐Adsorbed Secondary Antibody, Alexa Fluor 555 | Invitrogen | A‐21429 |
Oligonucleotides and other sequence‐based reagents | ||
PCR primers | This study | Table EV1 |
Universal probe #21 (Cdkn1a) | Roche | 4686942001 |
Universal probe #34 (Igf1) | Roche | 4687671001 |
Universal probe #77 (Myc) | Roche | 4689003001 |
Universal probe #77 (Stat5b) | Roche | 4689003001 |
Universal probe #66 (Tpt1) | Roche | 4688651001 |
The Universal Probe Library Mouse GAPDH Gene Assay | Sigma | 5046211001 |
Prime Time Std qPCR Assay, B2m | IDT | Mm.PT.39a.22214835 |
Prime Time Std qPCR Assay, Ppia | IDT | Mm.PT.39a.2.gs |
Universal probe #147 (rlp13a) | Roche | 4694333001 |
siRNA control: AllStars Negative Control siRNA | Qiagen | SI03650318 |
siRNA DNM2#1: Silencer® Select DNM2 siRNA | Thermo Fisher Scientific | s4212 |
siRNA DNM2#2: Silencer® Select DNM2 siRNA | Thermo Fisher Scientific | s4213 |
Hs_STAT5A_2 FlexiTube siRNA | Qiagen | SI00048426 |
Hs_STAT5B_3 FlexiTube siRNA | Qiagen | SI00100408 |
shRNAs | This study | Table EV1 |
Alt‐R® CRISPR‐Cas9 Negative Control crRNA #1 | IDT | 1072544 |
crRNAs | This study | Table EV1 |
Alt‐R® CRISPR‐Cas9 tracrRNA | IDT | 1072532 |
Chemicals, enzymes and other reagents | ||
Tyrphostin AG‐1478 | Cayman Chemicals | 10010244 |
BrightVision Goat Anti‐ Mouse/Rabbit IgG HRP two step detection system | Immunologic | DPVB55HRP |
BrightDiluent, normal antibody diluent | Immunologic | Bd09‐125 |
BS3 (bis(sulfosuccinimidyl)suberate) | Thermo Fisher Scientific | 21580 |
Cardiomyocyte Isolation Enzyme 1 (with papain) | Thermo Fisher Scientific | 88288 |
Cardiomyocyte Isolation Enzyme 2 (with thermolysin) | Thermo Fisher Scientific | 88289 |
Carnitine | Santa Cruz | sc‐205727A |
Creatine | Santa Cruz | sc‐214774A |
DAPI (4′,6‐diamidino‐2‐phenylindole) | Sigma | D9542 |
DMEM | Lonza | 12‐614F |
DMSO, Dimethyl Sulfoxide, Fisher BioReagents™ | Thermo Fisher Scientific | BP231‐100 |
DSP (dithiobis(succinimidyl propionate)), Lomant's Reagent | Thermo Fisher Scientific | 22585 |
DTBP (Wang and Richard's Reagent) | Thermo Fisher Scientific | 20665 |
Dynasore | Calbiochem | 324410 |
Duolink® In Situ PLA® Probe Anti‐Rabbit PLUS Affinity purified Donkey anti‐Rabbit IgG (H + L) | Sigma | DUO92002 |
Duolink® In Situ PLA® Probe Anti‐Goat MINUS Affinity purified Donkey anti‐Goat IgG (H + L) | Sigma | DUO92006 |
Duolink® In Situ Detection Reagents Orange | Sigma | DUO92007 |
Estrogen | Sigma | E2758 |
Fugene 6 | Promega | E2691 |
PureProteome™ Protein G Magnetic Bead System | Millipore | LSKMAGG10 |
Protein G‐sepharose™ 4 Fast Flow | GE Healthcare | 17‐0618‐01 |
EGF human, recombinant | Sigma | E9644 |
Fetal bovine serum | Biowest | S1810‐500 |
Gefitinib | Cayman Chemicals | 13166 |
Gelatin | Sigma | G‐1890 |
HBSS | Thermo Fisher Scientific | 88284 |
Hilymax | Dojindo | H357 |
Horse serum | GIBCO | 16050‐122 |
Mayer's hematoxylin | Sigma | 03971 |
Matrigel® Growth Factor Reduced | Corning | 356231 |
Medium 199 | Sigma | M2520 |
EnGen Cas9 NLS | New England Biolabs | M0646T |
Lapatinib | Selleckchem | S2111 |
Laemmli SDS sample buffer, reducing (6×) | Alfa aesar | J61337 |
Lipofectamine 2000 | Thermo Fisher Scientific | 11668019 |
Pronase E | Serva | 33635.02 |
Recombinant Human NRG‐1‐beta 1 | R&D | 396‐HB |
Matrigel (growth factor reduced) | Corning | 356231 |
Mowiol® 4‐88 | Sigma | 475904 |
Nuclease‐free duplex buffer | IDT | 11‐01‐03‐01 |
Penicillin‐Streptomycin Mixture | Lonza | DE17‐603E |
Percoll Plus | GE Healthcare | 17‐5445‐02 |
Pertex® Mounting Medium | Histolab | 00811‐EX |
Phenol red solution | Sigma | P0290 |
Protease inhibitor cocktail | Thermo Fisher Scientific | A32957 |
ReproSil‐Pur 5 μm 200 Å C18‐AQ beads | Dr. Maisch | r15.aq. |
RPMI 1640 | Lonza | BE04‐558F |
SensiFAST cDNA Synthesis Kit | Bioline | BIO‐65054 |
TaqMan universal Master Mix II | Applied Biosystems | 4440038 |
Taq I polymerase | Thermo Fisher Scientific | P0401 |
Taurine | Santa Cruz | sc‐202354 |
tC18 SepPak 96‐well plate | Waters | 86002321 |
Tricaine methane‐sulfonate | Finquel, Argent Laboratories | MS‐222 |
NucleoSpin TriPrep | Macherey‐Nagel | 740966.50 |
Trisure | Bioline | BIO‐38033 |
Tris‐EDTA pH 9,0 | Biosite | BSC‐20B1M |
Trypsin/EDTA | Lonza | CC‐5012 |
Trypsin Protease, MS Grade | Thermo Fisher Scientific | 90057 |
UltraGlutamine | Lonza | BE17‐605E/U1 |
Software | ||
GraphPad Prism 8.4.2 | GraphPad Software (https://www.graphpad.com/) | |
Matlab 2016a | MathWorks (https://mathworks.com/products/matlab.html) | |
FiJi v1.53c | Described earlier (Schindelin et al, 2012; https://imagej.net/software/fiji/) | |
Image Studio Lite v5.2 | Li‐COR (https://www.licor.com/bio/image‐studio‐lite/) | |
CaseViewer v2.3 | 3DHISTECH (https://www.3dhistech.com/solutions/caseviewer/) | |
QuantStudio 12K Software v1.4 | Thermo Fisher Scientific | |
MaxQuant v1.3 | Described earlier (Cox & Mann, 2008) | |
Metamorpheus (version 0.0.312) | Described earlier (Solntsev et al, 2018) | |
FlashLFQ | Described earlier (Millikin et al, 2018) | |
SpatTrack v2 | Described earlier (Lund et al, 2014) | |
colocalization algorithm of Villalta et al | Described earlier (Villalta et al, 2011) | |
R v3.6.1 | R Core Team (R Core Team, 2017; https://www.r‐project.org/) | |
RStudio v1.2.1335 | RStudio team (RStudio team, 2019; https://www.posit.co) | |
BioRender | https://www.biorender.com | |
Other | ||
TC20 Automated Cell Counter | Bio‐Rad | 1450102 |
Motorized pellet pestle | Sigma | Z359971 |
Zeiss LSM 780 confocal microscope | Zeiss (https://www.zeiss.com/microscopy/en/home.html) | |
Zeiss LSM 880 confocal microscope | Zeiss (https://www.zeiss.com/microscopy/en/home.html) | |
Zeiss AxioZOOM.V16 | Zeiss (https://www.zeiss.com/microscopy/en/home.html) | |
Labvision autostainer | Thermo Fisher Scientific | A80500024 |
Pannoramic P1000 slide scanner | 3DHISTECH | |
QuantStudio 12K Flex Real‐Time PCR System | Thermo Fisher Scientific | |
Odyssey DLx | Li‐COR | |
Nanoject II microinjector | Drummond Scientific | |
Nanodrop spectrophotometer | Thermo Fisher Scientific | |
Thermo Fisher Scientific LTQ Orbitrap XL mass‐spectrometer | Thermo Fisher Scientific | |
Easy‐nLC 1200 liquid chromatography system coupled to an Orbitrap Q‐Exactive HF instrument | Thermo Fisher Scientific | |
Vacuum centrifuge | Heto Lab |
Methods and Protocols
Isolation and culture of primary murine neonatal cardiomyocytes
NMRI male mice (Janvier Labs) were housed in the Central Animal Laboratory of University of Turku. The housing, upkeep, and euthanasia of animals were conducted according to Directive 2010/63/EU and under license from The Ministry of Agriculture and Forestry of Finland. The neonatal mice were sacrificed by decapitation. Freshly dissected neonatal (≤3 days of postpartum) hearts were minced and washed twice with ice‐cold HBSS (Hank's balanced salt solution). The hearts were enzymatically digested with Cardiomyocyte Isolation Enzymes 1 and 2, papain, and thermolysin, according to the manufacturer's instructions, except that thermolysin was used 1:5 of its suggested volume. The samples were washed twice with ice‐cold ADS buffer (Louch et al, 2011), mechanically agitated, and centrifuged at 900 g for 6 min in ADS buffer. The isolated cells were purified with a Percoll gradient, as previously described (Louch et al, 2011). Cell concentration and viability of cardiomyocytes were determined with TC20 Automated Cell Counter. Cells were plated on 1% gelatin‐coated plates and maintained in plating medium (68% DMEM, 17% 199, 10% horse serum, 5% fetal bovine serum, pH 7.2; Louch et al, 2011). After 2 days, the medium was changed to serum‐free maintenance medium (80% DMEM, 20% 199, pH 7.2, 5 mM Creatine, 2 mM Carnitine, 5 mM Taurine; Louch et al, 2011). For immunofluorescence analyses, the primary neonatal cardiomyocytes were plated in coverslips coated with 2% growth factor reduced Matrigel and cultured in plating medium. The primary cultures were sporadically tested for mycoplasma infections and infected cells were disposed.
Zebrafish husbandry
Zebrafish (Danio rerio) of casper strain (roy, mitfa) were housed at Zebrafish Core of Turku Bioscience Center, University of Turku and Åbo Akademi according to standard procedures (Nüsslein‐Volhard & Dahm, 2002). Embryos were obtained through natural spawning in dedicated breeding tanks. After collection, the embryos were flushed and incubated in E3‐medium at +28.5°C in a humidified incubator. The zebrafish embryos were anesthetized by administrating a single dose of tricaine (Ethyl 3‐aminobenzoate methanesulfonate) into the culture medium at end concentration of 200 mg/l. The zebrafish embryos were euthanized under terminal anesthesia by fixation in 4% paraformaldehyde or direct homogenization using a motorized pellet pestle. The zebrafish embryos at the age (< 5dpf) used in experiments are not considered protected animals as defined by Directive 2010/63/EU. The adult zebrafish were housed and used in mating according to Directive 2010/63/EU and under license MMM/465/712‐93 (Ministry of Forestry and Agriculture). The ARRIVE 2.0 reporting guideline was utilized. No statistical method was used for sample size estimation due to the high yield of embryos per spawning. The embryos were randomly allocated from one plate to different experimental groups. The group allocation was executed by a separate investigator independent of the investigators who analyzed the embryos. Underdeveloped embryos were excluded before treatment. Dead embryos were excluded before and after treatment. These exclusion criteria were set a priori.
Patient sample characteristics
The formalin‐fixed paraffin‐embedded myocardial tissue sections were acquired from Auria biobank though an application process (application number: AB20‐3971). The Finnish Biobank Act (Finlex 688/2012) enables the use of biobank resources in research. The patients whose samples have been used in the study have given their consent to use their samples in research to Auria biobank. The research conforms to the principles outlined in the Declaration of Helsinki. The cohorts included myocardial samples obtained from 19 patients who underwent medical autopsy at Turku University Hospital between 2008 and 2017. The hypertrophy cohort (median age 67; 23% female) included six samples from patients with aortic valve stenosis, four samples from patients with alcoholic cardiomyopathy and three from patients with idiopathic cardiomyopathy. The control cohort (median age 48; 50% female) included six cardiac samples taken from autopsied patients with noncardiac diseases (two cases of cirrhosis, one case of metastatic pancreatic carcinoid, one case of pulmonary embolism, one case of sepsis due to Streptococcus pneumoniae, and one case of cholangiocarcinoma). All the cases in the control cohort were negative for cardiomyocyte hypertrophy based on a histopathological analysis. One hypertrophy sample where the mean cardiomyocyte thickness was equal to the normal controls was excluded from the analysis.
Adeno‐associated virus‐treated mouse samples
The adeno‐associated virus (AAV)‐treated mouse samples were acquired from the experiments described previously (Kivelä et al, 2019). Before tissue collection, mice were injected intraperitoneally with 80 mg/kg of ketamine and 10 mg/kg of xylazine and euthanized by cervical dislocation. The local committee appointed by the district of southern Finland approved the animal experiments. FELASA (Federation of European Laboratory Animal Science Associations) guidelines and recommendations were followed. All the animal procedures and experiments were performed under the EU directive 2010/63/EU guidelines of the European parliament on the protection of animals used for scientific purposes.
Viral transduction
Lentiviral vectors were produced in HEK293T cells using a third‐generation lentiviral packaging system (Dull et al, 1998). The HEK293T cells were cultured in DMEM supplemented with 10% FBS, 5 mM UltraGlutamine, and 1 U of penicillin and streptomycin. Cells were co‐transfected overnight with pRSV‐Rev, pMDLg/pRRE, pMD2.G, and pLKO.1‐puromycin encoding one of the shRNA constructs. Medium was changed 24 h after transfection, and virus‐containing medium was collected at 48 and 72 h and filtered. Primary cardiomyocytes were infected with the lentiviruses at a multiplicity of infection (MOI) of 1 in the presence of 8 μg/ml polybrene. The medium was changed to serum‐free maintenance medium (80% DMEM, 20% 199, pH 7.2, 5 mM Creatine, 2 mM Carnitine, 5 mM Taurine; Louch et al, 2011) after 5 h. The cells were analyzed 24 h after infection.
Neuregulin stimulation and chemical inhibition
For western analyses, cells were stimulated with 50 ng/ml of human recombinant NRG‐1β for 2 min where indicated. For real‐time RT‐PCR analyses, the cells were incubated with 50 ng/ml NRG‐1β for 30 min. For immunofluorescence analyses of hypertrophic growth, the isolated cardiomyocytes were incubated with 50 ng/ml NRG‐1β for 2 days. To inhibit signaling, isolated cardiomyocytes were treated with the ERBB inhibitor 10 μM AG 1478 for 1 h or 50 μM dynasore for 3–6 h. To analyze the effect of ERBB or dynamin inhibition to the morphology and function of the heart of embryonic zebrafish, zebrafish embryos were immersed in E3‐medium supplemented with 10 μM AG 1478, 30 μM lapatinib, 30 μM gefitinib or 50 μM dynasore at 2 dpf for 2–3 days. To analyze the effect of ERBB or dynamin inhibition on downstream signaling with western analyses, zebrafish embryos at 4–5 dpf were immersed in E3‐medium supplemented with 10 μM AG 1478, 30 μM lapatinib, 30 μM gefitinib or 50 μM dynasore for 1–3 h. To analyze the effect of NRG‐1 on the morphology and function of the heart of embryonic zebrafish, 100 pg of recombinant human NRG‐1β or control substance (0.2% BSA in PBS) was microinjected to the pericardial sac of 2 dpf zebrafish embryos and the embryos were analyzed at 4–5 dpf. To analyze the effect of NRG‐1 to downstream signaling with western analysis, 100 pg of recombinant human NRG‐1β with 100 mM Na3VO4 or control substance with 100 mM Na3VO4 was microinjected to the pericardial sac of 4–5 dpf zebrafish embryos and lysed after 20 min.
Immunofluorescence and immunohistochemistry
For immunofluorescence analysis of isolated cardiomyocytes, samples were fixed and permeabilized with methanol for 10–15 min at −20°C and stained with the indicated primary antibodies. To remove nonspecific binding, 3% BSA in PBS was used as a blocking agent. Primary antibodies were detected with Alexa‐conjugated secondary antibodies and nuclei were visualized with DAPI. Mowiol was used for mounting. The samples were imaged with Zeiss LSM 780 confocal microscope with the 40× and 63× Zeiss C‐Apochromat objectives (Numerical Aperture: 1.2).
For immunofluorescence analysis of murine cardiac cryosections, the cryosections were allowed to reach room temperature and were rehydrated in PBS for 30 min. Sections were blocked by 3% BSA in PBS and stained with anti‐STAT5b for 3 h. The sections were washed three times with PBS and incubated with the secondary Alexa Fluor 488 goat anti‐rabbit antibody. Actin and nuclei were visualized with Alexa Fluor 555‐conjugated phalloidin and DAPI (4′,6‐diamidino‐2‐phenylindole), respectively. Coverslips were mounted with Mowiol. The samples were imaged with Zeiss LSM 780 and 880 confocal microscopes with the 40× Zeiss C‐Apochromat and the 40x Zeiss LD LCI Plan‐Apochromat objectives (Numerical Aperture: 1.2).
For immunofluorescence analysis of zebrafish embryos, the embryos were fixed and permeabilized with 4% PFA and 0.2% Triton X‐100 in PBS for 30 min. The embryos were washed four times with 0.2% Tween 20 in PBS and blocked with 3% BSA and 0.2% Tween 20 in PBS for 1 h. The primary and Alexa 555‐ or 488‐conjugated secondary antibodies were incubated overnight in +4°C under gentle agitation. The embryos were washed 4 times with 0.2% Tween 20 in PBS for 30 min under gentle agitation after primary and secondary antibody incubation. The stained embryos were imaged with Zeiss LSM 780 confocal microscope with the 40× Zeiss C‐Apochromat objective. The images were captured and analyzed independently by two separate investigators.
Human heart tissue sections were analyzed by immunohistochemistry in the Histology Core Facility of the Institute of Biomedicine, University of Turku. After deparaffinization and rehydration, the sections were incubated in Tris‐EDTA pH 9.0 in microwave oven for 7 min with 600 W followed by 7 min with 450 W, for antigen retrieval. The sections were incubated in the presence of hydrogen peroxide to inhibit endogenous enzyme function and in normal antibody diluent to prevent nonspecific binding of proteins. The sections were stained with an automated Labvision autostainer with anti‐pSTAT5 for 60 min in room temperature. Primary antibody was detected with BrightVision Goat Anti‐ Mouse/Rabbit IgG HRP two step detection system with horseradish peroxidase (HRP)‐conjugated secondary antibodies. Slides were counterstained with Mayer's hematoxylin for 1 min at room temperature, dehydrated and mounted with Pertex. The stained section were imaged with Panoramic P1000 slide scanner. The samples were analyzed blind.
Proximity ligation assay
The proximity ligation assay of dynamin‐2/ERBB4 association was performed according to the manufacturer's protocol with anti‐mouse minus and anti‐goat plus probes. A 1:100 dilution of primary dynamin‐2 and ERBB4 antibodies was used. The primary ERBB4 antibody was omitted from the technical control.
Western analysis
The zebrafish embryos were ground with an electronic pestle into 6× Laemmli buffer and incubated at 100°C for 10 min. The primary cultures were lysed with Triton‐X based lysis buffer (0.1% Triton X‐100, 1 mM EDTA, 5 mM NaF, 10 mM Tris–HCl, pH 7.4, and a protease inhibitor cocktail). The protein concentration was measured with Bradford assay and adjusted, and 6× Laemmli buffer was added to the lysed samples to obtain a 1× Laemmli solution. The samples were incubated at 100°C for 5 min. After incubation, the samples were centrifuged and loaded onto SDS–PAGE gels, run, and transferred to nitrocellulose membranes. The membranes were probed with the indicated primary antibodies and IR‐conjugated secondary antibodies and imaged with the Li‐COR Odyssey system. The densitometric quantitation was performed with Image Studio Lite.
RNA extraction and real‐time RT‐PCR
Total RNA was extracted using TRIsure RNA isolation reagent according to the manufacturer's instructions. RNA concentrations were determined using a Nanodrop spectrophotometer, and complementary DNA was synthesized with Sensifast cDNA Synthesis Kit from 1 μg of total RNA. Real‐time RT–PCR analyses of gene expression were performed with 500 nM of primers, 100 nM of 5′ 6‐FAM‐labeled probe and TaqMan universal Master Mix II according to the manufacturer's protocol. Thermal cycling was performed with QuantStudio 12K Flex Real‐Time PCR System. The relative real‐time RT PCR values were determined by normalizing the C t values of the gene of interest with the C t values of the housekeeping genes as follows: . The C t values of the genes of interest in isolated cardiomyocytes were normalized to the average C t values of B2m and Ppia housekeeping genes. The C t values of the genes of interest in cardiac tissue samples were normalized to the C t value of the Tpt1 gene. Some replicates were excluded due to technical concerns.
In vivo imaging of zebrafish embryos
The embryos were anesthetized with Tricaine methane‐sulfonate and molded into low‐melting point agarose. The fast time laps videos of embryos were taken with HC Image Software of Zeiss AxioZOOM.V16 with 1.0× PlanApo Z objective with 80× magnification. The videos were imaged and analyzed independently by two separate investigators.
CRISPR/Cas9 mediated knockdown of STAT5b in zebrafish embryos
The CRISPR/Cas9 mediated knockdown was carried out largely as described in a previous report (Hoshijima et al, 2019). Guide RNAs were generated by combining 5 μl of crRNAs (100 μM) and tracRNA (100 μM). Duplexes were annealed by heating to 95°C for 5 min, cooled down by 0.1°C/s to 25°C, and incubated at 25°C for 5 min. Ten microlitre of nuclease‐free duplex buffer (IDT) was added to reach a final concentration of 25 μM and duplexes were stored at −20°C.
To generate crRNA:tracRNA:Cas9 complexes, 1 μl of duplex, 1.25 μl of EnGen Cas9 NLS solution, 1.75 μl H2O, and 1 μl of phenol red solution were mixed together and incubated for 5 min at 37°C prior to injection. Next, 2.3 nl of the solution was injected into 1–4 cell stage zebrafish embryos of casper line using Nanoject II microinjector. After injection, the embryos were placed in a +28.5°C incubator in E3‐medium supplemented with 1:100 Pencillin‐Streptomycin mixture. At 1 dpf, dead embryos were removed and 15 μl of pronase E was added to facilitate hatching. The embryos were analyzed at 4 dpf.
Cell culture
The MDA‐MB‐468 and HC11 cell lines stably expressing ErbB4 JM‐a and JM‐b have been described before (Sundvall et al, 2012). The HC11 cells were cultured in RPMI supplemented with 10% FBS, 2 mM UltraGlutamine, 1 U penicillin and streptomycin, 100 ng/ml EGF, 300 ng/ml puromycin and 1:2000 insulin solution. The MCF‐7 cells were cultured in RPMI supplemented with 10% FBS, 5 mM UltraGlutamine, 1 U penicillin and streptomycin, 10 nM estrogen and 1:2000 insulin solution. The MDA‐MB‐468 cells were cultured in DMEM supplemented with 10% FBS, 5 mM Glutamine, 1 U penicillin and streptomycin and 300 ng/ml puromycin. All cell lines were maintained at 37°C in 5% CO2 in media. The cell lines were sporadically tested for mycoplasma infections and infected cells were disposed. No authentications were recently performed for the cell lines.
Transfection
Fugene 6 or Hilymax was used for DNA plasmid transfection in MCF‐7, and Lipofectamine 2000 for siRNA transfection in MDA‐MB‐468 cells, as recommended by the manufacturer's protocol. DNM2 siRNAs were used at 30 nM concentration and incubated with the cells for 48 h. STAT5 siRNAs were used at 50 nM concentration and incubated with the cells for 48 h.
Immunoprecipitation
The cells were serum starved and treated where indicated with 50 ng/ml NRG‐1β for 2 min before lysis with Triton‐X based lysis buffer (0.1% Triton X‐100, 1 mM EDTA, 5 mM NaF, 10 mM Tris–HCl, pH 7.4, and a protease inhibitor cocktail). The lysates were precleared for 1 h at 4°C with G‐sepharose beads before immunoprecipitation for 3–12 h at 4°C with the indicated antibodies. The immunoprecipitates were washed seven times with 0.2% Tween 20 in PBS and eluted and denatured with incubation with 6× Laemmli buffer for 5 min at 100°C. The samples were loaded onto SDS–PAGE gels, run, and transferred to nitrocellulose membranes. The membranes were probed with the the indicated primary antibodies and HRP (horseradish peroxidase)‐linked secondary antibodies. The membranes were incubated with a chemiluminescence substrate for 5 min and the light signal was captured with X‐ray film.
Mass spectrometry
The cells were cross‐linked with 1 mM DSP or DTPB for 2 min and quenched with 50 mM Tris–HCl pH 7.4 for 15 min at room temperature. The cross‐linked cells were lysed with Triton‐X based lysis buffer and precleared with 50 μl of preclear beads for 1 h at 4°C under gentle agitation. To prepare the beads for the preclearing, 160 μl of magnetic G‐protein beads were blocked with 5% BSA, incubated with 8 μg mouse IgG and 8 μg of rabbit IgG for 3 h at 4°C and cross‐linked with 3 mM BS3 crosslinker for 30 min at room temperature. The mixture was quenched with 50 mM Tris pH 7.5. The precleared lysate was incubated with the ErbB4 antibodies cross‐linked to magnetic G‐protein beads for 3 h at 4°C under gentle agitation. To prepare the ErbB4 G‐protein beads, 160 μl of magnetic G‐protein beads were blocked with 5% BSA, incubated with 8 μg anti‐ErbB4 (HFR‐1) and 8 μg anti‐ErbB4 (E200) for 3 h at 4°C and cross‐linked with 3 mM BS3 crosslinker for 30 min at room temperature. The mixture was quenched with 50 mM Tris pH 7.5 for 15 min and washed 3 times with 0.1% Tween 20 in PBS. The immunoprecipitates were washed 7 times with 0.2% Tween 20 in PBS. In the first experiment, the MDA‐MB‐468 ErbB4 JM‐a and JM‐b immunoprecipitates were eluted with 0.2 M glycine (pH 2.5), methanol‐cholorform precipitated and sent to Proteomics Core Laboratory in the University of Tartu for LC/MS/MS analysis with Thermo Fisher Scientific LTQ Orbitrap XL mass‐spectrometer. The mass spectrometry files were analyzed with MaxQuant software. In the second experiment, the MDA‐MB‐468 ErbB4 JM‐a and JM‐b immunoprecipitates were eluted with 6 M guanidine hydrochloride, 5 mM tris(2‐carboxyethyl)phosphine, 10 mM chloroacetamide and 100 mM Tris, pH 8.5 in 95°C for 10 min. Guanidine hydrochloride was diluted to 2 M and eluted samples were digested with 1:50 (w/v) trypsin overnight at 37°C. Following digestion, the reactions were quenched with 10% trifluoroacetic acid at a final concentration of approximately 0.5% (pH ~ 2), desalted on tC18 SepPak 96‐well plate, and dried by vacuum centrifugation. The mass spectrometry analyses were performed in the Turku Proteomics Facility at Turku Bioscience (University of Turku and Åbo Akademi University). The dried peptides samples were resuspended in 0.1% formic acid before analysis with mass spectrometers. The peptides were analyzed on an Easy‐nLC 1200 liquid chromatography system coupled to an Orbitrap Q‐Exactive HF instrument equipped with a nanoelectrospray source. Peptides were loaded on an in‐house packed 100 μm × 2 cm precolumn packed with ReproSil‐Pur 5 μm 200 Å C18‐AQ beads using 0.1% formic acid in water (buffer A) and separated by reverse phase chromatography on a 75 μm × 15 cm analytical column packed with ReproSil‐Pur 5 μm 200 Å C18‐AQ beads. All separations were performed using a 40 min gradient at a flow rate of 300 nl/min. The gradient ranged from 6% buffer B (80% acetonitrile in 0.1% formic acid) to 36% buffer B in 30 min. Buffer B concentration was ramped up to 100% in 5 min, and 100% buffer B was run 5 min for washout. The DDA method consisted of a full MS scan (120,000 resolution, 3E6 automatic gain control [AGC] target, 100 ms maximum injection time, 300–1,750 m/z, profile mode), followed by data‐dependent MS/MS acquisitions on the top 20 most intense precursor ions (15,000 resolution, 5E4 AGC target, 100 ms maximum injection time, 2.0 m/z isolation window, normalized collision energy = 27, centroid mode). MS/MS spectra were searched with Metamorpheus against human proteome. Proteomes contained known post‐translational modifications (downloaded from Uniprot at 10.1.2021). Mass spectrometry files were calibrated, possible post‐translational modifications were searched, and peptides and proteins were identified, and quantified using FlashLFQ algorithm. Cysteine carbamidomethylation (57.021463) was set as a constant modification and methionine oxidation was set as a variable modification. All other possible modifications were set using G‐DPM search in Metamorpheus. Search results were filtered to a 1% FDR at PSM level. Peptides were accepted with search engine score above 5. The analyzed mass spectrometry data is provided as Dataset EV1 and EV2.
Genomic DNA and RNA isolation from zebrafish embryos
To isolate genomic DNA and RNA from zebrafish embryos, the embryos were lysed with an electronic pestle to the lysis buffer of the NucleoSpin TriPrep. The genomic DNA and RNA isolation was conducted with the NucleoSpin TriPrep kit according to the manufacturer's protocol.
Nested PCR and sequencing
For sequencing CRISPR/Cas9 edited zebrafish, the genomic DNA of the zebrafish embryos were amplified by PCR using 10 nM outer Dr. Cas9.STAT5B primers with Taq I polymerase. The PCR products from the first PCR amplification were used as a template for amplification with 10 nM inner Dr.Cas9.STAT5B primers and the PCR reactions were supplemented with 2.5 mM Mg2Cl2. The amplified product was Sanger sequenced.
Preparation of formalin fixed paraffin embedded samples
To prepare formalin fixed paraffin embedded (FFPE) samples to control the specificity of the pSTAT5 immunostaining, MCF‐7 cells were serum starved for 12 h and treated or not with 1:2000 insulin solution for 10 min, and MDA‐MB‐468 cells were transfected with control or STAT5 siRNAs and serum starved for 12 h. The cells were incubated with a trypsin–EDTA solution at 37°C for 5 min to detach the cells, centrifuged for 5 min with 500 g in +4°C and washed twice with ice‐cold PBS. The cells were fixed with 10% neutral buffered formalin in PBS for 20 min in room temperature and washed twice with PBS. The cells were suspended into 1.5% agarose in PBS warmed to 50°C and allowed to cool down to room temperature for 15 min. The agarose cell pellets were transferred to 70% ethanol and embedded into paraffin blocks with a standard protocol by the Histology Core Facility of the Institute of Biomedicine, University of Turku. Five μm sections from the blocks were stained as the FFPE myocardial tissue sections from patients.
Image analysis
The area of isolated cardiomyocytes and zebrafish embryo ventricles, and the thickness of ventricles was measured with Fiji. The ejection fraction of zebrafish ventricle was determined by measuring the average width of the ventricle at systole and diastole with Fiji and fitting the values to the simplified Quinones equation: The ventricular width, measured from outer margins of the myocardium, at diastole and at systole was used as the end‐diastolic dimension (EDD) and end‐systolic dimension (ESD), respectively. The amount of PLA signals was measured with the particle analyzer function of Fiji and the location of PLA signals was analyzed with SpatTrack. The colocalization of immunofluorescence signals was measured with the algorithm of Villalta et al, 2011. Four colocalization measures (Pearson's correlation, Manders overlap, M1/M2, k1/k2) were produced of which k1/k2 ( and , where S 1 and S 2 are the intensity of the signal from channel 1 and 2, respectively) is visualized in the images. The relative intensity of the DAB staining was measured from color deconvoluted images with Fiji. The average intensity in the cardiomyocyte nuclei to cardiomyocyte cytosol (background) was used. The intensity of immunofluorescence signal was measured with Fiji.
Data and statistical analyses
Matlab R2016a and Graph Pad Prism 8.4.2 were used for statistical analysis. The normality and homoscedasticity of each dataset was tested with Shapiro–Wilkis, Kolmogorov–Smirnov, Bartlett's and Brown–Forsythe test and parametric, parametric testing with variance correction or nonparametric testing was chosen accordingly. All data combined from independent experiments were median normalized except for the data in Fig 1J. The data in 1J were normalized to the range to fit the combined data into a normal distribution. The transcriptome datasets of subjects with pathological hypertrophy and control subjects were downloaded from the Gene Expression Omnibus database (Edgar et al, 2002; Barrett et al, 2013). The principle component analysis of the pre‐existing datasets was performed with R in RStudio. The statistical significance of the categorization of the dimensionality‐reduced values was calculated by estimating an empirical probability distribution for the relative distance and drawing the cumulative probability from the corresponding cumulative probability function. The relative distance was determined by the sum of all Euclidian distances within the category divided by the sum of all Euclidian distances between categories. The empirical probability distribution was simulated by drawing 10,000 random gene sets of same size from the dataset, subjecting the gene sets to principal component analysis and calculating the relative distance between the categories of the dimensionality reduced values. The schematic figures were created with biorender.com.
Author contributions
Katri Vaparanta: Conceptualization; resources; data curation; formal analysis; funding acquisition; validation; investigation; visualization; methodology; writing – original draft; project administration; writing – review and editing. Anne Jokilammi: Validation; investigation; methodology; writing – review and editing. Ilkka Paatero: Resources; investigation; methodology; writing – review and editing. Johannes A Merilahti: Data curation; investigation; writing – review and editing. Juho Heliste: Methodology; writing – review and editing. Karthik Amudhala Hemanthakumar: Resources. Riikka Kivelä: Resources; writing – review and editing. Kari Alitalo: Resources. Pekka Taimen: Resources; investigation; methodology; writing – review and editing. Klaus Elenius: Conceptualization; supervision; funding acquisition; visualization; writing – original draft; writing – review and editing.
Disclosure and competing interests statement
KE has ownership interest in Abomics, Novo Nordisk, Orion, and Roche. JH declares competing financial interests, including direct investments and a role as a medical advisor in Abomics Oy. PT reports personal fees from Roche, AstraZeneca, Bayer and MSD and non‐financial support from MSD, all outside the described work. The other authors declare no conflict of interest.
Supporting information
Acknowledgments
We thank Maria Tuominen, Mika Savisalo, Nea Konttinen, Merja Lakkisto, Minna Santanen, Julia Palosara and Sinikka Kollanus for skillful technical assistance, and Riku Kiviranta and his laboratory for providing research material. Turku Doctoral Program of Molecular Medicine is acknowledged for excellent doctoral training and funding. The Auria biobank is acknowledged for providing patient samples. We acknowledge the Cell Imaging Core, Proteomics Core, Zebrafish Core and Finnish Functional Genomics Centre (Turku Bioscience Centre, University of Turku and Åbo Akademi University) all supported by Biocenter Finland and Histology Core Facility of the Institute of Biomedicine, University of Turku for their excellent service. The Proteomics Core Laboratory in the University of Tartu is acknowledged for their proteomics services. This work was financially supported by the Academy of Finland, Cancer Foundation Finland, the Sigrid Juselius Foundation, the Turku University Central Hospital, the Turku University Foundation, Orion Research Foundation, K. Albin Johansson's Foundation, the Cancer Society of Southwestern Finland, the Maud Kuistila memorial Foundation, Aarne Koskelo Foundation, Paulo Foundation, Emil Aaltonen Foundation, the Finnish Foundation for Cardiovascular Research, Paavo Nurmi Foundation, Finnish Cultural Foundation and the Varsinais‐Suomi Regional Fund of the Finnish Cultural Foundation.
EMBO reports (2023) 24: e56689
Data availability
The datasets produced in this study are available in the following databases:
The Mass spectrometry derived interactome data: ProteomeXchange Consortium (Deutsch et al, 2023) via the PRIDE (Perez‐Riverol et al, 2022) partner repository PXD040166 (https://www.ebi.ac.uk/pride/archive/projects/PXD040166).
References
- Anderson NG, Ahmad T, Chan K, Dobson R, Bundred NJ (2001) ZD1839 (Iressa), a novel epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, potently inhibits the growth of EGFR‐positive cancer cell lines with or without erbB2 overexpression. Int J Cancer 94: 774–782 [DOI] [PubMed] [Google Scholar]
- Baliga RR, Pimental DR, Zhao YY, Simmons WW, Marchionni MA, Sawyer DB, Kelly RA (1999) NRG‐1‐induced cardiomyocyte hypertrophy. Role of PI‐3‐kinase, p70(S6K), and MEK‐MAPK‐RSK. Am J Physiol 277: H2026–H2037 [DOI] [PubMed] [Google Scholar]
- Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M et al (2013) NCBI GENE EXPRESSION OMNIBUS: archive for functional genomics data sets–update. Nucleic Acids Res 41: D991–D995 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Basham B, Sathe M, Grein J, McClanahan T, D'Andrea A, Lees E, Rascle A (2008) In vivo identification of novel STAT5 target genes. Nucleic Acids Res 36: 3802–3818 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bersell K, Arab S, Haring B, Kühn B (2009) Neuregulin1/ErbB4 signaling induces cardiomyocyte proliferation and repair of heart injury. Cell 138: 257–270 [DOI] [PubMed] [Google Scholar]
- Bisping E, Ikeda S, Kong S, Tarnavski O, Bodyak N, McMullen JR, Rajagopal S, Son JK, Ma Q, Springer Z et al (2006) Gene Expression Omnibus GSE5500 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE5500). [DATASET] [DOI] [PMC free article] [PubMed]
- Bongiovanni C, Sacchi F, Da PS, Pantano E, Miano C, Morelli MB, D'uva G (2021) Reawakening the intrinsic cardiac regenerative potential: molecular strategies to boost dedifferentiation and proliferation of endogenous cardiomyocytes. Front Cardiovasc Med 8: 750604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai MX, Shi XC, Chen T, Tan ZN, Lin QQ, Du SJ, Tian ZJ (2016) Exercise training activates neuregulin 1/ErbB signaling and promotes cardiac repair in a rat myocardial infarction model. Life Sci 149: 1–9 [DOI] [PubMed] [Google Scholar]
- Chen H, Jing XY, Shen YJ, Wang TL, Ou C, Lu SF, Cai Y, Li Q, Chen X, Ding YJ et al (2018) Stat5‐dependent cardioprotection in late remote ischaemia preconditioning. Cardiovasc Res 114: 679–689 [DOI] [PubMed] [Google Scholar]
- Cohen JE, Purcell BP, MacArthur JW, Mu A, Shudo Y, Patel JB, Brusalis CM, Trubelja A, Fairman AS, Edwards BB et al (2014) A bioengineered hydrogel system enables targeted and sustained intramyocardial delivery of neuregulin, activating the cardiomyocyte cell cycle and enhancing ventricular function in a murine model of ischemic cardiomyopathy. Circ Hear Fail 7: 619–626 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cox J, Mann M (2008) MaxQuant enables high peptide identification rates, individualized p.p.b.‐range mass accuracies and proteome‐wide protein quantification. Nat Biotechnol 26: 1367–1372 [DOI] [PubMed] [Google Scholar]
- Crone SA, Zhao YY, Fan L, Gu Y, Minamisawa S, Liu Y, Peterson KL, Chen J, Kahn R, Condorelli G et al (2002) ErbB2 is essential in the prevention of dilated cardiomyopathy. Nat Med 8: 459–465 [DOI] [PubMed] [Google Scholar]
- De Keulenaer GW, Feyen E, Dugaucquier L, Shakeri H, Shchendrygina A, Belenkov YN, Brink M, Vermeulen Z, Segers VFM (2019) Mechanisms of the multitasking endothelial protein NRG‐1 as a compensatory factor during chronic heart failure. Circ Heart Fail 12: 6288 [DOI] [PubMed] [Google Scholar]
- Delaughter MC, Taffet GE, Fiorotto ML, Entman ML, Schwartz RJ (1999) Local insulin‐like growth factor I expression induces physiologic, then pathologic, cardiac hypertrophy in transgenic mice. FASEB J 13: 1923–1929 [DOI] [PubMed] [Google Scholar]
- Deutsch EW, Bandeira N, Perez‐Riverol Y, Sharma V, Carver JJ, Mendoza L, Kundu DJ, Wang S, Bandla C, Kamatchinathan S et al (2023) The ProteomeXchange consortium at 10 years: 2023 update. Nucleic Acids Res 51: D1539–D1548 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dull T, Zufferey R, Kelly M, Mandel RJ, Nguyen M, Trono D, Naldini L (1998) A third‐generation lentivirus vector with a conditional packaging system. J Virol 72: 8463–8471 [DOI] [PMC free article] [PubMed] [Google Scholar]
- D'uva G, Aharonov A, Lauriola M, Kain D, Yahalom‐Ronen Y, Carvalho S, Weisinger K, Bassat E, Rajchman D, Yifa O et al (2015) ERBB2 triggers mammalian heart regeneration by promoting cardiomyocyte dedifferentiation and proliferation. Nat Cell Biol 17: 627–638 [DOI] [PubMed] [Google Scholar]
- Edgar R, Domrachev M, Lash AE (2002) Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res 30: 207–210 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Egeblad M, Mortensen OH, Van Kempen LCLT, Jäättelä M (2001) BIBX1382BS, but not AG1478 or PD153035, inhibits the ErbB kinases at different concentrations in intact cells. Biochem Biophys Res Commun 281: 25–31 [DOI] [PubMed] [Google Scholar]
- Elenius K, Corfas G, Paul S, Choi CJ, Rio C, Plowman GD, Klagsbrun M (1997) A novel Juxtamembrane domain isoform of HER4/ErbB4: isoform‐specific tissue distribution and differential processing in response to phorbol ester. J Biol Chem 272: 26761–26768 [DOI] [PubMed] [Google Scholar]
- Fukazawa R, Miller TA, Kuramochi Y, Frantz S, Kim Y‐D, Marchionni MA, Kelly RA, Sawyer DB (2003) Neuregulin‐1 protects ventricular myocytes from anthracycline‐induced apoptosis via erbB4‐dependent activation of PI3‐kinase/Akt. J Mol Cell Cardiol 35: 1473–1479 [DOI] [PubMed] [Google Scholar]
- Gallo S, Vitacolonna A, Bonzano A, Comoglio P, Crepaldi T (2019) ERK: a key player in the pathophysiology of cardiac hypertrophy. Int J Mol Sci 20: 2164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao R, Zhang J, Cheng L, Wu X, Dong W, Yang X, Li T, Liu X, Xu Y, Li X et al (2010) A phase II, randomized, double‐blind, multicenter, based on standard therapy, placebo‐controlled study of the efficacy and safety of recombinant human neuregulin‐1 in patients with chronic heart failure. J Am Coll Cardiol 55: 1907–1914 [DOI] [PubMed] [Google Scholar]
- Gao R, Zhang J, Liu H, Wang L, Pang X, Fu L, Meng F, Zhou M (2018) A Phase III, randomized, double-blind, multicenter, placebo-controlled study of the efficacy and safety of neucardin in patients with chronic heart failure. J Am Coll Cardiol 71: A668 [DOI] [PubMed] [Google Scholar]
- García‐Rivello H, Taranda J, Said M, Cabeza‐Meckert P, Vila‐Petroff M, Scaglione J, Ghio S, Chen J, Lai C, Laguens RP et al (2005) Dilated cardiomyopathy in Erb‐b4‐deficient ventricular muscle. Am J Physiol 289: H1153–H1160 [DOI] [PubMed] [Google Scholar]
- Gassmann M, Casagranda F, Orioli D, Simon H, Lai C, Klein R, Lemke G (1995) Aberrant neural and cardiac development in mice lacking the ErbB4 neuregulin receptor. Nature 378: 390–394 [DOI] [PubMed] [Google Scholar]
- Gemberling M, Karra R, Dickson AL, Poss KD (2015) Nrg1 is an injury‐induced cardiomyocyte mitogen for the endogenous heart regeneration program in zebrafish. Elife 4: e05871 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hebl VB, Bos JM, Oberg AL, Sun Z, Herman DS, Teekakirikul P, Seidman JG, Seidman CE, dos Remedios CG, Schaff HV et al (2012) Gene Expression Omnibus GSE36961 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE36961). [DATASET]
- Hedhli N, Huang Q, Kalinowski A, Palmeri M, Hu X, Russell RR, Russell KS (2011) Endothelium‐derived neuregulin protects the heart against ischemic injury. Circulation 123: 2254–2262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heusch G, Musiolik J, Kottenberg E, Peters J, Jakob H, Thielmann M (2012) STAT5 activation and cardioprotection by remote ischemic preconditioning in HumansNovelty and significance. Circ Res 110: 111–115 [DOI] [PubMed] [Google Scholar]
- Hirai M, Arita Y, McGlade CJ, Lee KF, Chen J, Evans SM (2017) Adaptor proteins NUMB and NUMBL promote cell cycle withdrawal by targeting ERBB2 for degradation. J Clin Invest 127: 569–582 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoshijima K, Jurynec MJ, Klatt Shaw D, Jacobi AM, Behlke MA, Grunwald DJ (2019) Highly efficient CRISPR‐Cas9‐based methods for generating deletion mutations and F0 embryos that lack Gene function in zebrafish. Dev Cell 51: 645–657.e4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang CY, Lin YC, Hsiao WY, Liao FH, Huang PY, Tan TH (2012) DUSP4 deficiency enhances CD25 expression and CD4+ T‐cell proliferation without impeding T‐cell development. Eur J Immunol 42: 476–488 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jabbour A, Hayward CS, Keogh AM, Kotlyar E, McCrohon JA, England JF, Amor R, Liu X, Li XY, Zhou MD et al (2011) Parenteral administration of recombinant human neuregulin‐1 to patients with stable chronic heart failure produces favourable acute and chronic haemodynamic responses. Eur J Heart Fail 13: 83–92 [DOI] [PubMed] [Google Scholar]
- Jie B, Zhang X, Wu X, Xin Y, Liu Y, Guo Y (2012) Neuregulin‐1 suppresses cardiomyocyte apoptosis by activating PI3K/Akt and inhibiting mitochondrial permeability transition pore. Mol Cell Biochem 370: 35–43 [DOI] [PubMed] [Google Scholar]
- Jin G, Wang L, Ma J (2022) Inhibiting STAT5 significantly attenuated ang II‐induced cardiac dysfunction and inflammation. Eur J Pharmacol 915: 174689 [DOI] [PubMed] [Google Scholar]
- Kehat I, Davis J, Tiburcy M, Accornero F, Saba‐El‐Leil MK, Maillet M, York AJ, Lorenz JN, Zimmermann WH, Meloche S et al (2011) ERK1/2 regulate the balance between eccentric and concentric cardiac growth. Circ Res 108: 176–183 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kimura A, Ishida Y, Furuta M, Nosaka M, Kuninaka Y, Taruya A, Mukaida N, Kondo T (2018) Protective roles of interferon‐γ in cardiac hypertrophy induced by sustained pressure overload. J Am Hear Assoc Cardiovasc Cerebrovasc Dis 7: e008145 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kivelä R, Hemanthakumar KA, Vaparanta K, Robciuc M, Izumiya Y, Kidoya H, Takakura N, Peng X, Sawyer DB, Elenius K et al (2019) Endothelial cells regulate physiological cardiomyocyte growth via VEGFR2‐mediated paracrine signaling. Circulation 139: 2570–2584 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Komamura K (2017) Recombinant insulin‐like growth factor‐1 improves cardiac function and symptoms in the patients on the waiting list for heart transplantation with end‐stage dilated cardiomyopathy. Eur Heart J 38: ehx502.P1105 [Google Scholar]
- Kuramochi Y, Guo X, Sawyer DB (2006) Neuregulin activates erbB2‐dependent src/FAK signaling and cytoskeletal remodeling in isolated adult rat cardiac myocytes. J Mol Cell Cardiol 41: 228–235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lai L, Martin O, Leone TC, Kelly DP (2014) Gene Expression Omnibus GSE56348 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE56348). [DATASET]
- Lee KF, Simon H, Chen H, Bates B, Hung MC, Hauser C (1995) Requirement for neuregulin receptor erbB2 in neural and cardiac development. Nature 378: 394–398 [DOI] [PubMed] [Google Scholar]
- Lemmens K, Segers VFM, Demolder M, De Keulenaer GW (2006) Role of Neuregulin‐1/ErbB2 signaling in endothelium‐cardiomyocyte cross‐talk. J Biol Chem 281: 19469–19477 [DOI] [PubMed] [Google Scholar]
- Li J, Zhang DS, Ye JC, Li CM, Qi M, Liang DD, Xu XR, Xu L, Liu Y, Zhang H et al (2013) Dynamin‐2 mediates heart failure by modulating Ca2+−dependent cardiomyocyte apoptosis. Int J Cardiol 168: 2109–2119 [DOI] [PubMed] [Google Scholar]
- Liu X, Gu X, Li Z, Li X, Li H, Chang J, Chen P, Jin J, Xi B, Chen D et al (2006) Neuregulin‐1/erbB‐activation improves cardiac function and survival in models of ischemic, dilated, and viral cardiomyopathy. J Am Coll Cardiol 48: 1438–1447 [DOI] [PubMed] [Google Scholar]
- Liu J, Bressan M, Hassel D, Huisken J, Staudt D, Kikuchi K, Poss KD, Mikawa T, Stainier DYR (2010) A dual role for ErbB2 signaling in cardiac trabeculation. Development 137: 3867–3875 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lock MC, Tellam RL, Botting KJ, Wang KCW, Selvanayagam JB, Brooks DA, Seed M, Morrison JL (2018) The role of miRNA regulation in fetal cardiomyocytes, cardiac maturation and the risk of heart disease in adults. J Physiol 596: 5625–5640 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lord JD, McIntosh BC, Greenberg PD, Nelson BH (2000) The IL‐2 receptor promotes lymphocyte proliferation and induction of the c‐myc, bcl‐2, and bcl‐x genes through the trans‐activation domain of Stat5. J Immunol 164: 2533–2541 [DOI] [PubMed] [Google Scholar]
- Louch WE, Sheehan KA, Wolska BM (2011) Methods in cardiomyocyte isolation, culture, and gene transfer. J Mol Cell Cardiol 51: 288–298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lund FW, Jensen MLV, Christensen T, Nielsen GK, Heegaard CW, Wüstner D (2014) SpatTrack: an imaging toolbox for analysis of vesicle motility and distribution in living cells. Traffic 15: 1406–1429 [DOI] [PubMed] [Google Scholar]
- Maatta JA, Sundvall M, Junttila TT, Peri L, Laine VJO, Isola J, Egeblad M, Elenius K (2005) Proteolytic cleavage and phosphorylation of a tumor‐associated ErbB4 isoform promote ligand‐independent survival and cancer cell growth. Mol Biol Cell 17: 67–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Margulies KB, Cappola TP (2019) Gene Expression Omnibus GSE141910 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE141910). [DATASET]
- Martens N, Uzan G, Wery M, Hooghe R, Hooghe‐Peters EL, Gertler A (2005) Suppressor of cytokine signaling 7 inhibits prolactin, growth hormone, and leptin signaling by interacting with STAT5 or STAT3 and attenuating their nuclear translocation. J Biol Chem 280: 13817–13823 [DOI] [PubMed] [Google Scholar]
- Mendes‐Ferreira P, De Keulenaer GW, Leite‐Moreira AF, Brás‐Silva C (2013) Therapeutic potential of neuregulin‐1 in cardiovascular disease. Drug Discov Today 18: 836–842 [DOI] [PubMed] [Google Scholar]
- Meyer D, Birchmeier C (1995) Multiple essential functions of neuregulin in development. Nature 378: 386–390 [DOI] [PubMed] [Google Scholar]
- Millikin RJ, Solntsev SK, Shortreed MR, Smith LM (2018) Ultrafast peptide label‐free quantification with FlashLFQ. J Proteome Res 17: 386–391 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Montero JC, Yuste L, Díaz‐Rodríguez E, Esparís‐Ogando A, Pandiella A (2000) Differential shedding of transmembrane neuregulin isoforms by the tumor necrosis factor‐α‐converting enzyme. Mol Cell Neurosci 16: 631–648 [DOI] [PubMed] [Google Scholar]
- Nakajima H, Brindle PK, Handa M, Ihle JN (2001) Functional interaction of STAT5 and nuclear receptor co‐repressor SMRT: implications in negative regulation of STAT5‐dependent transcription. EMBO J 20: 6836–6844 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakamura M, Sadoshima J (2018) Mechanisms of physiological and pathological cardiac hypertrophy. Nat Rev Cardiol 15: 387–407 [DOI] [PubMed] [Google Scholar]
- Nosaka T, Kawashima T, Misawa K, Ikuta K, Mui ALF, Kitamura T (1999) STAT5 as a molecular regulator of proliferation, differentiation and apoptosis in hematopoietic cells. EMBO J 18: 4754–4765 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nüsslein‐Volhard C, Dahm R (2002) Zebrafish: a practical approach, 1st edn. Oxford: Oxford University Press; [Google Scholar]
- Odiete O, Hill MF, Sawyer DB (2012) Neuregulin in cardiovascular development and disease. Circ Res 111: 1376–1385 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Özcelik C, Erdmann B, Pilz B, Wettschureck N, Britsch S, Hübner N, Chien KR, Birchmeier C, Garratt AN (2002) Conditional mutation of the ErbB2 (HER2) receptor in cardiomyocytes leads to dilated cardiomyopathy. Proc Natl Acad Sci USA 99: 8880–8885 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Papatheodorou I, Moreno P, Manning J, Fuentes AMP, Gene Expression Omnibusrge N, Fexova S, Fonseca NA, Füllgrabe A, Green M, Huang N et al (2020) Expression atlas update: from tissues to single cells. Nucleic Acids Res 48: D77–D83 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paul A, Schinke M, Brown J, Riggi LE, Izumo S, Bartunek J, Allen P, M T (2004) Gene Expression Omnibus GSE1145 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE1145) [DATASET]
- Peng Y, Zhou B, Wang Y, Chen Y, Li H, Song Y, Zhang L, Rao L (2012) Association between polymorphisms in the signal transducer and activator of transcription and dilated cardiomyopathy in the Chinese Han population. Mol Cell Biochem 360: 197–203 [DOI] [PubMed] [Google Scholar]
- Pentassuglia L, Heim P, Lebboukh S, Morandi C, Xu L, Brink M (2016) Neuregulin‐1β promotes glucose uptake via PI3K/Akt in neonatal rat cardiomyocytes. Am J Physiol Endocrinol Metab 310: E782–E794 [DOI] [PubMed] [Google Scholar]
- Perez‐Riverol Y, Bai J, Bandla C, García‐Seisdedos D, Hewapathirana S, Kamatchinathan S, Kundu DJ, Prakash A, Frericks‐Zipper A, Eisenacher M et al (2022) The PRIDE database resources in 2022: a hub for mass spectrometry‐based proteomics evidences. Nucleic Acids Res 50: D543–D552 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Plowman GD, Green JM, Culouscou JM, Carlton GW, Rothwell VM, Buckley S (1993) Heregulin induces tyrosine phosphorylation of HER4/p180erbB4. Nature 366: 473–475 [DOI] [PubMed] [Google Scholar]
- Poss KD, Wilson LG, Keating MT (2002) Heart regeneration in zebrafish. Science 298: 2188–2190 [DOI] [PubMed] [Google Scholar]
- R Core Team (2017) R: a language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing; (www.R‐project.org/) [Google Scholar]
- Rasouli SJ, Stainier DYR (2017) Regulation of cardiomyocyte behavior in zebrafish trabeculation by neuregulin 2a signaling. Nat Commun 81: 1–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rigacci S, Guidotti V, Parri M, Berti A (2008) Modulation of STAT5 interaction with LMW‐PTP during early megakaryocyte differentiation. Biochemistry 47: 1482–1489 [DOI] [PubMed] [Google Scholar]
- Rohrbach S, Yan X, Weinberg EO, Hasan F, Bartunek J, Marchionni MA, Lorell BH (1999) Neuregulin in cardiac hypertrophy in rats with aortic stenosis. Circulation 100: 407–412 [DOI] [PubMed] [Google Scholar]
- Rohrbach S, Muller‐Werdan U, Werdan K, Koch S, Gellerich NF, Holtz J (2005a) Apoptosis‐modulating interaction of the neuregulin/erbB pathway with antracyclines in regulating Bcl‐xS and Bcl‐xL in cardiomyocytes. J Mol Cell Cardiol 38: 485–493 [DOI] [PubMed] [Google Scholar]
- Rohrbach S, Niemann B, Silber RE, Holtz J (2005b) Neuregulin receptors erbB2 and erbB4in failing human myocardium. Depressed expression and attenuated activation. Basic Res Cardiol 100: 240–249 [DOI] [PubMed] [Google Scholar]
- Romano N, Ceci M (2020) Are microRNAs responsible for cardiac hypertrophy in fish and mammals? What we can learn in the activation process in a zebrafish ex vivo model. Biochim Biophys Acta Mol Basis Dis 1866: 165896 [DOI] [PubMed] [Google Scholar]
- Rotwein P (2012) Mapping the growth hormone – Stat5b – IGF‐I transcriptional circuit. Trends Endocrinol Metab 23: 186–193 [DOI] [PMC free article] [PubMed] [Google Scholar]
- RStudio team (2019) RStudio: integrated development for R. Boston, MA: RStudio, PBC. (https://www.posit.co) [Google Scholar]
- Samsa LA, Ito CE, Brown DR, Qian L, Liu J (2016) IgG‐containing isoforms of Neuregulin‐1 are dispensable for cardiac trabeculation in zebrafish. PLoS One 11: e0166734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sawyer DB, Zuppinger C, Miller TA, Eppenberger HM, Suter TM (2002) Modulation of anthracycline‐induced myofibrillar disarray in rat ventricular myocytes by neuregulin‐1β and anti‐erbB2. Circulation 105: 1551–1554 [DOI] [PubMed] [Google Scholar]
- Schindelin J, Arganda‐Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, Preibisch S, Rueden C, Saalfeld S, Schmid B et al (2012) Fiji: an open‐source platform for biological‐image analysis. Nat Methods 9: 676–682 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sefat‐E‐Khuda YM, Xing Y, Shimasaki T, Takeya M, Kuwahara K, Sakaguchi N (2004) The Sac3 homologue shd1 is involved in mitotic progression in mammalian cells. J Biol Chem 279: 46182–46190 [DOI] [PubMed] [Google Scholar]
- Smeets P, Vogels‐van den Bosch H, Willemsen P, Stassen A, Ayoubi T, van der Vusse G, van Bilsen M (2008) Gene Expression Omnibus GSE12337 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE12337). [DATASET]
- Solntsev SK, Shortreed MR, Frey BL, Smith LM (2018) Enhanced global post‐translational modification discovery with MetaMorpheus. J Proteome Res 17: 1844–1851 [DOI] [PubMed] [Google Scholar]
- Sundvall M, Korhonen A, Vaparanta K, Anckar J, Halkilahti K, Salah Z, Aqeilan RI, Palvimo JJ, Sistonen L, Elenius K (2012) Protein inhibitor of activated STAT3 (PIAS3) protein promotes SUMOylation and nuclear sequestration of the intracellular domain of ErbB4 protein. J Biol Chem 287: 23216–23226 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vaparanta K, Jokilammi A, Tamirat M, Merilahti JAM, Salokas K, Varjosalo M, Ivaska J, Johnson MS, Elenius K (2022) An extracellular receptor tyrosine kinase motif orchestrating intracellular STAT activation. Nat Commun 13: 6953 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Villalta JI, Galli S, Iacaruso MF, Antico Arciuch VG, Poderoso JJ, Jares-Erijman EA, Pietrasanta LI (2011) New algorithm to determine true colocalization in combination with image restoration and time-lapse confocal microscopy to map kinases in mitochondria. PLoS One 6: e19031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z, Chan HW, Gambarotta G, Smith NJ, Purdue BW, Pennisi DJ, Porrello ER, O'Brien SL, Reichelt ME, Thomas WG et al (2021) Stimulation of the four isoforms of receptor tyrosine kinase ErbB4, but not ErbB1, confers cardiomyocyte hypertrophy. J Cell Physiol 236: 8160–8170 [DOI] [PubMed] [Google Scholar]
- Welch S, Plank D, Witt S, Glascock B, Schaefer E, Chimenti S, Andreoli AM, Limana F, Leri A, Kajstura J et al (2002) Cardiac‐specific IGF‐1 expression attenuates dilated cardiomyopathy in tropomodulin‐overexpressing transgenic mice. Circ Res 90: 641–648 [DOI] [PubMed] [Google Scholar]
- White RM, Sessa A, Burke C, Bowman T, LeBlanc J, Ceol C, Bourque C, Dovey M, Goessling W, Burns CE et al (2008) Transparent adult zebrafish as a tool for in vivo transplantation analysis. Cell Stem Cell 2: 183–189 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu Q, Wang T, Chen S, Zhou Q, Li H, Hu N, Feng Y, Dong N, Yao S, Xia Z (2017) Cardiac protective effects of remote ischaemic preconditioning in children undergoing tetralogy of fallot repair surgery: a randomized controlled trial. Eur Heart J 105: 151–154 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia W, Mullin RJ, Keith BR, Liu LH, Ma H, Rusnak DW, Owens G, Alligood KJ, Spector NL (2002) Anti‐tumor activity of GW572016: a dual tyrosine kinase inhibitor blocks EGF activation of EGFR/erbB2 and downstream Erk1/2 and AKT pathways. Oncogene 2141: 6255–6263 [DOI] [PubMed] [Google Scholar]
- Yu CL, Jin YJ, Burakoff SJ (2000) Cytosolic tyrosine dephosphorylation of STAT5: Potential Role Of SHP‐2 in STAT5 regulation. J Biol Chem 275: 599–604 [DOI] [PubMed] [Google Scholar]
- Yu JH, Zhu BM, Wickre M, Riedlinger G, Chen W, Hosui A, Robinson GW, Hennighausen L (2010) The transcription factors STAT5A and STAT5B negatively regulate cell proliferation through the activation of Cdkn2b and Cdkn1a expression. Hepatology 52: 1808–1818 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zensun Sci. & Tech. Co., Ltd (2018) Survival study of the recombinant human neuregulin‐1β in subjects with chronic heart failure. ClinicalTrials.gov NCT03388539. https://clinicaltrials.gov/ct2/show/NCT03388593
- Zhao YY, Sawyer DR, Baliga RR, Opel DJ, Han X, Marchionni MA, Kelly RA (1998) Neuregulins promote survival and growth of cardiac myocytes. Persistence of ErbB2 and ErbB4 expression in neonatal and adult ventricular myocytes. J Biol Chem 273: 10261–10269 [DOI] [PubMed] [Google Scholar]
- Zhong W, Mao S, Tobis S, Angelis E, Jordan MC, Roos KP, Fishbein MC, De Alborán IM, MacLellan WR (2006) Hypertrophic growth in cardiac myocytes is mediated by Myc through a cyclin D2‐dependent pathway. EMBO J 25: 3869–3879 [DOI] [PMC free article] [PubMed] [Google Scholar]