In BrdU+ cardiomyocytes and total cardiomyocytes, suggesting that NGF increases cardiomyocyte proliferation in vivo. This observation was completely consistent with our demonstration that the cardiotoxin induced reduction in cardiomyocyte numbers could be reversed by NGF. In this context it is of note that in chick dorsal root ganglion sensory neuronal cultures, NGF potently increased neuregulin-1 (NRG1) [25], and that NRG1 through its receptor ErbB4 induced mammalian adult cardiomyocytes to proliferate [5]. Together it is therefore possible that NGF may trigger proliferation of cardiomyocytes via upregulation of NRG1 in the heart. TA-02 Recent work suggested that GATA4 is activated in cardiomyocytes during the regenerative process of the cardiac resection model [10], however in this study the addition of NGF following AA treatment did not change gata4 mRNA Dimethylenastron transcript levels in the heart. Additionally, a recent study in adult mouse heart demonstrated that cardiomyocyte specific deletion of glycogen synthase kinase (GSK)-3b following surgically induced myocardial infarction increased cardiomyocyte proliferation [26]. Of interest, in rat sympathetic neurons, PC12 cells, and mouse embryonic dorsal root ganglion neurons, NGF promotes axon growth by activation of phosphatidylinositol 3-kinase (PI3K) which inactivate GSK-3b through the TrkA receptor [27,28,29,30,31]. In contrast, in mouse embryonic hippocampal neurons, NGF promotes axon elongation by inactivation of GSK-3b mediated by p75NTR receptor [32]. Importantly, NGF ultimately inactivates GSK-3b regardless of which NGF receptor signalling pathway it activates. Taken together, it is conceivable that NGF may inactivate GSK-3b in cardiomyocytes to induce proliferation. In adult zebrafish, cardiac regeneration is primarily mediated by cardiomyocyte proliferation [10,11], and NGF is modestly upregulated in the heart post amputation [33]. Our study supports that NGF augments cardiomyocyte proliferation and may play an important role in cardiac regeneration. While our findings areNGF Rescues Heart Failureconsistent broadly with the concept of a cardiomyocyte proliferation mediated regenerative response, it also highlights the importance of differences in experimental paradigms. In the present study, 24195657 in our globally injured heart model we show that there is an insufficient regenerative response, unlike that in the locally, surgically damaged zebrafish heart. In summary, the present study demonstrates that NGF may attenuate the progression of HF via the induction of a regenerative program based upon cardiomyocyte proliferation rather than by an anti-apoptotic mechanism. These studies are complimentary to prior observations which demonstrate that NGF expression is reduced in the failing heart, and importantly taken together suggest that a deficiency of NGF within the failing heart could impair the capacity of any residual regenerative capacity. Importantly, further work is required to establish the translational relevance of these findings to that in human disease in older individuals. Regenerating the myocardium following injury to prevent the onset of heart failure is one of the ultimate goals of cardiac repair [6]. We hypothesise that NGF may provide a more favourable environment which sufficiently regenerates the heart to prevent the development of heart failure. Further studies are required to evaluate the precise molecular mechanism by whichthis process is mediated, and importantly to establish.In BrdU+ cardiomyocytes and total cardiomyocytes, suggesting that NGF increases cardiomyocyte proliferation in vivo. This observation was completely consistent with our demonstration that the cardiotoxin induced reduction in cardiomyocyte numbers could be reversed by NGF. In this context it is of note that in chick dorsal root ganglion sensory neuronal cultures, NGF potently increased neuregulin-1 (NRG1) [25], and that NRG1 through its receptor ErbB4 induced mammalian adult cardiomyocytes to proliferate [5]. Together it is therefore possible that NGF may trigger proliferation of cardiomyocytes via upregulation of NRG1 in the heart. Recent work suggested that GATA4 is activated in cardiomyocytes during the regenerative process of the cardiac resection model [10], however in this study the addition of NGF following AA treatment did not change gata4 mRNA transcript levels in the heart. Additionally, a recent study in adult mouse heart demonstrated that cardiomyocyte specific deletion of glycogen synthase kinase (GSK)-3b following surgically induced myocardial infarction increased cardiomyocyte proliferation [26]. Of interest, in rat sympathetic neurons, PC12 cells, and mouse embryonic dorsal root ganglion neurons, NGF promotes axon growth by activation of phosphatidylinositol 3-kinase (PI3K) which inactivate GSK-3b through the TrkA receptor [27,28,29,30,31]. In contrast, in mouse embryonic hippocampal neurons, NGF promotes axon elongation by inactivation of GSK-3b mediated by p75NTR receptor [32]. Importantly, NGF ultimately inactivates GSK-3b regardless of which NGF receptor signalling pathway it activates. Taken together, it is conceivable that NGF may inactivate GSK-3b in cardiomyocytes to induce proliferation. In adult zebrafish, cardiac regeneration is primarily mediated by cardiomyocyte proliferation [10,11], and NGF is modestly upregulated in the heart post amputation [33]. Our study supports that NGF augments cardiomyocyte proliferation and may play an important role in cardiac regeneration. While our findings areNGF Rescues Heart Failureconsistent broadly with the concept of a cardiomyocyte proliferation mediated regenerative response, it also highlights the importance of differences in experimental paradigms. In the present study, 24195657 in our globally injured heart model we show that there is an insufficient regenerative response, unlike that in the locally, surgically damaged zebrafish heart. In summary, the present study demonstrates that NGF may attenuate the progression of HF via the induction of a regenerative program based upon cardiomyocyte proliferation rather than by an anti-apoptotic mechanism. These studies are complimentary to prior observations which demonstrate that NGF expression is reduced in the failing heart, and importantly taken together suggest that a deficiency of NGF within the failing heart could impair the capacity of any residual regenerative capacity. Importantly, further work is required to establish the translational relevance of these findings to that in human disease in older individuals. Regenerating the myocardium following injury to prevent the onset of heart failure is one of the ultimate goals of cardiac repair [6]. We hypothesise that NGF may provide a more favourable environment which sufficiently regenerates the heart to prevent the development of heart failure. Further studies are required to evaluate the precise molecular mechanism by whichthis process is mediated, and importantly to establish.