Skip to main content
Top
Published in: Current Treatment Options in Cardiovascular Medicine 10/2015

01-10-2015 | Regenerative Medicine and Stem-cell Therapy (S Wu and P Hsieh, Section Editors)

Harnessing the Induction of Cardiomyocyte Proliferation for Cardiac Regenerative Medicine

Authors: Arun Sharma, BS, Yuan Zhang, Sean M. Wu, MD PhD

Published in: Current Treatment Options in Cardiovascular Medicine | Issue 10/2015

Login to get access

Opinion statement

Adult human cardiomyocytes are terminally differentiated and have limited capacity for cell division. Hence, they are not naturally replaced following ischemic injury to the heart. As such, cardiac function is often permanently compromised after an event such as myocardial infarction. In recent years, investigators have focused intensively on ways to reactivate cardiomyocyte mitotic activity in both in vitro cell culture systems and in vivo animal models. In parallel, advances in stem cell biology have allowed for the mass production of patient-specific human cardiomyocytes from human-induced pluripotent stem cells. These cells can be produced via chemically defined differentiation of human pluripotent stem cells in a matter of weeks and could theoretically be utilized directly for therapeutic purposes to replace damaged myocardium. However, stem cell-derived cardiomyocytes, like their adult counterparts, are post-mitotic and incapable of large-scale expansion after reaching a certain stage of in vitro differentiation. Due to this shared characteristic, these stem cell-derived cardiomyocytes may provide a platform for studying genes, pathways, and small molecules that induce cell cycle reentry and proliferation of human cardiomyocytes. Ultimately, the discovery of novel mechanisms or pathways to induce human cardiomyocyte proliferation should improve our ability to regenerate adult cardiomyocytes and help restore cardiac function following injury.
Literature
1.
go back to reference Pagidipati NJ, Gaziano TA. Estimating deaths from cardiovascular disease: a review of global methodologies of mortality measurement. Circulation. 2013;127:749–56.PubMedCentralCrossRefPubMed Pagidipati NJ, Gaziano TA. Estimating deaths from cardiovascular disease: a review of global methodologies of mortality measurement. Circulation. 2013;127:749–56.PubMedCentralCrossRefPubMed
2.
go back to reference Oberpriller JO, Oberpriller JC. Response of the adult newt ventricle to injury. J Exp Zool. 1974;187:249–53.CrossRefPubMed Oberpriller JO, Oberpriller JC. Response of the adult newt ventricle to injury. J Exp Zool. 1974;187:249–53.CrossRefPubMed
3.
4.
go back to reference Li F, Wang X, Capasso JM, Gerdes AM. Rapid transition of cardiac myocytes from hyperplasia to hypertrophy during postnatal development. J Mol Cell Cardiol. 1996;28:1737–46.CrossRefPubMed Li F, Wang X, Capasso JM, Gerdes AM. Rapid transition of cardiac myocytes from hyperplasia to hypertrophy during postnatal development. J Mol Cell Cardiol. 1996;28:1737–46.CrossRefPubMed
5.•
go back to reference Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA, Olson EN, et al. Transient regenerative potential of the neonatal mouse heart. Science. 2011;331:1078–80. This study was the first to demonstrate that the mammalian heart harbors some regenerative potential via cardiomyocyte proliferation, but that this ability is lost quickly after birth.PubMedCentralCrossRefPubMed Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA, Olson EN, et al. Transient regenerative potential of the neonatal mouse heart. Science. 2011;331:1078–80. This study was the first to demonstrate that the mammalian heart harbors some regenerative potential via cardiomyocyte proliferation, but that this ability is lost quickly after birth.PubMedCentralCrossRefPubMed
6.
go back to reference Sturzu AC, Rajarajan K, Passer D, Plonowska K, Riley A, Tan TC, et al. Fetal mammalian heart generates a robust compensatory response to cell loss. Circulation. 2015;132:109–21.CrossRefPubMed Sturzu AC, Rajarajan K, Passer D, Plonowska K, Riley A, Tan TC, et al. Fetal mammalian heart generates a robust compensatory response to cell loss. Circulation. 2015;132:109–21.CrossRefPubMed
7.
go back to reference Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114:763–76.CrossRefPubMed Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114:763–76.CrossRefPubMed
8.
go back to reference Kimura W, Xiao F, Canseco DC, Muralidhar S, Thet S, Zhang HM, et al. Hypoxia fate mapping identifies cycling cardiomyocytes in the adult heart. Nature. 2015;523:226–30.CrossRefPubMed Kimura W, Xiao F, Canseco DC, Muralidhar S, Thet S, Zhang HM, et al. Hypoxia fate mapping identifies cycling cardiomyocytes in the adult heart. Nature. 2015;523:226–30.CrossRefPubMed
9.
go back to reference Ryan TJ, Anderson JL, Antman EM, Braniff BA, Brooks NH, Califf RM, et al. Acc/aha guidelines for the management of patients with acute myocardial infarction: executive summary. A report of the American college of cardiology/american heart association task force on practice guidelines (committee on management of acute myocardial infarction). Circulation. 1996;94:2341–50.CrossRefPubMed Ryan TJ, Anderson JL, Antman EM, Braniff BA, Brooks NH, Califf RM, et al. Acc/aha guidelines for the management of patients with acute myocardial infarction: executive summary. A report of the American college of cardiology/american heart association task force on practice guidelines (committee on management of acute myocardial infarction). Circulation. 1996;94:2341–50.CrossRefPubMed
10.
go back to reference Abouna GM. Organ shortage crisis: problems and possible solutions. Transplant Proc. 2008;40:34–8.CrossRefPubMed Abouna GM. Organ shortage crisis: problems and possible solutions. Transplant Proc. 2008;40:34–8.CrossRefPubMed
11.
go back to reference Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A, et al. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest. 2001;108:407–14.PubMedCentralCrossRefPubMed Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A, et al. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest. 2001;108:407–14.PubMedCentralCrossRefPubMed
12.
go back to reference Kattman SJ, Witty AD, Gagliardi M, Dubois NC, Niapour M, Hotta A, et al. Stage-specific optimization of activin/nodal and bmp signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell. 2011;8:228–40.CrossRefPubMed Kattman SJ, Witty AD, Gagliardi M, Dubois NC, Niapour M, Hotta A, et al. Stage-specific optimization of activin/nodal and bmp signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell. 2011;8:228–40.CrossRefPubMed
13.
go back to reference Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical wnt signaling. Proc Natl Acad Sci U S A. 2012;109:E1848–57.PubMedCentralCrossRefPubMed Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical wnt signaling. Proc Natl Acad Sci U S A. 2012;109:E1848–57.PubMedCentralCrossRefPubMed
14.•
go back to reference Burridge PW, Matsa E, Shukla P, Lin ZC, Churko JM, Ebert AD, et al. Chemically defined generation of human cardiomyocytes. Nat Methods. 2014;11:855–60. This study established the first chemically-defined protocol for the production of human cardiomyocytes from pluripotent stem cells, allowing for the utilization of patient-specific cardiomyocytes for downstream disease modeling and drug discovery assays without employing undefined components such as serum.PubMedCentralCrossRefPubMed Burridge PW, Matsa E, Shukla P, Lin ZC, Churko JM, Ebert AD, et al. Chemically defined generation of human cardiomyocytes. Nat Methods. 2014;11:855–60. This study established the first chemically-defined protocol for the production of human cardiomyocytes from pluripotent stem cells, allowing for the utilization of patient-specific cardiomyocytes for downstream disease modeling and drug discovery assays without employing undefined components such as serum.PubMedCentralCrossRefPubMed
15.
go back to reference Niebruegge S, Nehring A, Bar H, Schroeder M, Zweigerdt R, Lehmann J. Cardiomyocyte production in mass suspension culture: embryonic stem cells as a source for great amounts of functional cardiomyocytes. Tissue Eng Part A. 2008;14:1591–601.CrossRefPubMed Niebruegge S, Nehring A, Bar H, Schroeder M, Zweigerdt R, Lehmann J. Cardiomyocyte production in mass suspension culture: embryonic stem cells as a source for great amounts of functional cardiomyocytes. Tissue Eng Part A. 2008;14:1591–601.CrossRefPubMed
16.
go back to reference Lundy SD, Zhu WZ, Regnier M, Laflamme MA. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells Dev. 2013;22:1991–2002.PubMedCentralCrossRefPubMed Lundy SD, Zhu WZ, Regnier M, Laflamme MA. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells Dev. 2013;22:1991–2002.PubMedCentralCrossRefPubMed
17.
go back to reference Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O, Winterstern A, et al. Modelling the long qt syndrome with induced pluripotent stem cells. Nature. 2011;471:225–9.CrossRefPubMed Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O, Winterstern A, et al. Modelling the long qt syndrome with induced pluripotent stem cells. Nature. 2011;471:225–9.CrossRefPubMed
18.
go back to reference Sharma A, Wu JC, Wu SM. Induced pluripotent stem cell-derived cardiomyocytes for cardiovascular disease modeling and drug screening. Stem Cell Res Ther. 2013;4:150.PubMedCentralCrossRefPubMed Sharma A, Wu JC, Wu SM. Induced pluripotent stem cell-derived cardiomyocytes for cardiovascular disease modeling and drug screening. Stem Cell Res Ther. 2013;4:150.PubMedCentralCrossRefPubMed
19.
go back to reference Sharma A, Marceau C, Hamaguchi R, Burridge PW, Rajarajan K, Churko JM, et al. Human induced pluripotent stem cell-derived cardiomyocytes as an in vitro model for coxsackievirus b3-induced myocarditis and antiviral drug screening platform. Circ Res. 2014;115:556–66.PubMedCentralCrossRefPubMed Sharma A, Marceau C, Hamaguchi R, Burridge PW, Rajarajan K, Churko JM, et al. Human induced pluripotent stem cell-derived cardiomyocytes as an in vitro model for coxsackievirus b3-induced myocarditis and antiviral drug screening platform. Circ Res. 2014;115:556–66.PubMedCentralCrossRefPubMed
20.
go back to reference Mordwinkin NM, Burridge PW, Wu JC. A review of human pluripotent stem cell-derived cardiomyocytes for high-throughput drug discovery, cardiotoxicity screening, and publication standards. J Cardiovasc Transl Res. 2013;6:22–30.PubMedCentralCrossRefPubMed Mordwinkin NM, Burridge PW, Wu JC. A review of human pluripotent stem cell-derived cardiomyocytes for high-throughput drug discovery, cardiotoxicity screening, and publication standards. J Cardiovasc Transl Res. 2013;6:22–30.PubMedCentralCrossRefPubMed
21.
go back to reference Chong JJ, Yang X, Don CW, Minami E, Liu YW, Weyers JJ, et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature. 2014;510:273–7.PubMedCentralCrossRefPubMed Chong JJ, Yang X, Don CW, Minami E, Liu YW, Weyers JJ, et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature. 2014;510:273–7.PubMedCentralCrossRefPubMed
22.
go back to reference Zhang D, Shadrin IY, Lam J, Xian HQ, Snodgrass HR, Bursac N. Tissue-engineered cardiac patch for advanced functional maturation of human esc-derived cardiomyocytes. Biomaterials. 2013;34:5813–20.PubMedCentralCrossRefPubMed Zhang D, Shadrin IY, Lam J, Xian HQ, Snodgrass HR, Bursac N. Tissue-engineered cardiac patch for advanced functional maturation of human esc-derived cardiomyocytes. Biomaterials. 2013;34:5813–20.PubMedCentralCrossRefPubMed
23.
go back to reference Stevens KR, Pabon L, Muskheli V, Murry CE. Scaffold-free human cardiac tissue patch created from embryonic stem cells. Tissue Eng Part A. 2009;15:1211–22.PubMedCentralCrossRefPubMed Stevens KR, Pabon L, Muskheli V, Murry CE. Scaffold-free human cardiac tissue patch created from embryonic stem cells. Tissue Eng Part A. 2009;15:1211–22.PubMedCentralCrossRefPubMed
24.
go back to reference Sun L, Yu J, Qi S, Hao Y, Liu Y, Li Z. Bone morphogenetic protein-10 induces cardiomyocyte proliferation and improves cardiac function after myocardial infarction. J Cell Biochem. 2014;115:1868–76.PubMed Sun L, Yu J, Qi S, Hao Y, Liu Y, Li Z. Bone morphogenetic protein-10 induces cardiomyocyte proliferation and improves cardiac function after myocardial infarction. J Cell Biochem. 2014;115:1868–76.PubMed
25.
go back to reference Chaudhry HW, Dashoush NH, Tang H, Zhang L, Wang X, Wu EX, et al. Cyclin a2 mediates cardiomyocyte mitosis in the postmitotic myocardium. J Biol Chem. 2004;279:35858–66.CrossRefPubMed Chaudhry HW, Dashoush NH, Tang H, Zhang L, Wang X, Wu EX, et al. Cyclin a2 mediates cardiomyocyte mitosis in the postmitotic myocardium. J Biol Chem. 2004;279:35858–66.CrossRefPubMed
26.
go back to reference Woo YJ, Panlilio CM, Cheng RK, Liao GP, Atluri P, Hsu VM, et al. Therapeutic delivery of cyclin a2 induces myocardial regeneration and enhances cardiac function in ischemic heart failure. Circulation. 2006;114:I206–13.CrossRefPubMed Woo YJ, Panlilio CM, Cheng RK, Liao GP, Atluri P, Hsu VM, et al. Therapeutic delivery of cyclin a2 induces myocardial regeneration and enhances cardiac function in ischemic heart failure. Circulation. 2006;114:I206–13.CrossRefPubMed
27.
go back to reference Bicknell KA, Coxon CH, Brooks G. Forced expression of the cyclin b1-cdc2 complex induces proliferation in adult rat cardiomyocytes. Biochem J. 2004;382:411–6.PubMedCentralCrossRefPubMed Bicknell KA, Coxon CH, Brooks G. Forced expression of the cyclin b1-cdc2 complex induces proliferation in adult rat cardiomyocytes. Biochem J. 2004;382:411–6.PubMedCentralCrossRefPubMed
28.
go back to reference Tamamori-Adachi M, Ito H, Sumrejkanchanakij P, Adachi S, Hiroe M, Shimizu M, et al. Critical role of cyclin d1 nuclear import in cardiomyocyte proliferation. Circ Res. 2003;92:e12–9.CrossRefPubMed Tamamori-Adachi M, Ito H, Sumrejkanchanakij P, Adachi S, Hiroe M, Shimizu M, et al. Critical role of cyclin d1 nuclear import in cardiomyocyte proliferation. Circ Res. 2003;92:e12–9.CrossRefPubMed
29.
go back to reference Evans-Anderson HJ, Alfieri CM, Yutzey KE. Regulation of cardiomyocyte proliferation and myocardial growth during development by foxo transcription factors. Circ Res. 2008;102:686–94.CrossRefPubMed Evans-Anderson HJ, Alfieri CM, Yutzey KE. Regulation of cardiomyocyte proliferation and myocardial growth during development by foxo transcription factors. Circ Res. 2008;102:686–94.CrossRefPubMed
30.
go back to reference Jung J, Kim TG, Lyons GE, Kim HR, Lee Y. Jumonji regulates cardiomyocyte proliferation via interaction with retinoblastoma protein. J Biol Chem. 2005;280:30916–23.CrossRefPubMed Jung J, Kim TG, Lyons GE, Kim HR, Lee Y. Jumonji regulates cardiomyocyte proliferation via interaction with retinoblastoma protein. J Biol Chem. 2005;280:30916–23.CrossRefPubMed
31.
go back to reference O'Meara CC, Wamstad JA, Gladstone RA, Fomovsky GM, Butty VL, Shrikumar A, et al. Transcriptional reversion of cardiac myocyte fate during mammalian cardiac regeneration. Circ Res. 2015;116:804–15.CrossRefPubMed O'Meara CC, Wamstad JA, Gladstone RA, Fomovsky GM, Butty VL, Shrikumar A, et al. Transcriptional reversion of cardiac myocyte fate during mammalian cardiac regeneration. Circ Res. 2015;116:804–15.CrossRefPubMed
32.
go back to reference Novoyatleva T, Sajjad A, Pogoryelov D, Patra C, Schermuly RT, Engel FB. Fgf1-mediated cardiomyocyte cell cycle reentry depends on the interaction of fgfr-1 and fn14. FASEB J. 2014;28:2492–503.CrossRefPubMed Novoyatleva T, Sajjad A, Pogoryelov D, Patra C, Schermuly RT, Engel FB. Fgf1-mediated cardiomyocyte cell cycle reentry depends on the interaction of fgfr-1 and fn14. FASEB J. 2014;28:2492–503.CrossRefPubMed
33.
go back to reference Rochais F, Sturny R, Chao CM, Mesbah K, Bennett M, Mohun TJ, et al. Fgf10 promotes regional foetal cardiomyocyte proliferation and adult cardiomyocyte cell-cycle re-entry. Cardiovasc Res. 2014;104:432–42.CrossRefPubMed Rochais F, Sturny R, Chao CM, Mesbah K, Bennett M, Mohun TJ, et al. Fgf10 promotes regional foetal cardiomyocyte proliferation and adult cardiomyocyte cell-cycle re-entry. Cardiovasc Res. 2014;104:432–42.CrossRefPubMed
34.
go back to reference Shimoji K, Yuasa S, Onizuka T, Hattori F, Tanaka T, Hara M, et al. G-csf promotes the proliferation of developing cardiomyocytes in vivo and in derivation from escs and ipscs. Cell Stem Cell. 2010;6:227–37.CrossRefPubMed Shimoji K, Yuasa S, Onizuka T, Hattori F, Tanaka T, Hara M, et al. G-csf promotes the proliferation of developing cardiomyocytes in vivo and in derivation from escs and ipscs. Cell Stem Cell. 2010;6:227–37.CrossRefPubMed
35.
go back to reference McDevitt TC, Laflamme MA, Murry CE. Proliferation of cardiomyocytes derived from human embryonic stem cells is mediated via the igf/pi 3-kinase/akt signaling pathway. J Mol Cell Cardiol. 2005;39:865–73.PubMedCentralCrossRefPubMed McDevitt TC, Laflamme MA, Murry CE. Proliferation of cardiomyocytes derived from human embryonic stem cells is mediated via the igf/pi 3-kinase/akt signaling pathway. J Mol Cell Cardiol. 2005;39:865–73.PubMedCentralCrossRefPubMed
36.
go back to reference Engels MC, Rajarajan K, Feistritzer R, Sharma A, Nielsen UB, Schalij MJ, et al. Insulin-like growth factor promotes cardiac lineage induction in vitro by selective expansion of early mesoderm. Stem Cells. 2014;32:1493–502.PubMedCentralCrossRefPubMed Engels MC, Rajarajan K, Feistritzer R, Sharma A, Nielsen UB, Schalij MJ, et al. Insulin-like growth factor promotes cardiac lineage induction in vitro by selective expansion of early mesoderm. Stem Cells. 2014;32:1493–502.PubMedCentralCrossRefPubMed
37.
go back to reference Bersell K, Arab S, Haring B, Kuhn B. Neuregulin1/erbb4 signaling induces cardiomyocyte proliferation and repair of heart injury. Cell. 2009;138:257–70.CrossRefPubMed Bersell K, Arab S, Haring B, Kuhn B. Neuregulin1/erbb4 signaling induces cardiomyocyte proliferation and repair of heart injury. Cell. 2009;138:257–70.CrossRefPubMed
38.
go back to reference Hou CJ, Qi YM, Zhang DZ, Wang QG, Cui CS, Kuang L, et al. The proliferative and migratory effects of physical injury and stromal cell-derived factor-1 alpha on rat cardiomyocytes and fibroblasts. Eur Rev Med Pharmacol Sci. 2015;19:1252–7.PubMed Hou CJ, Qi YM, Zhang DZ, Wang QG, Cui CS, Kuang L, et al. The proliferative and migratory effects of physical injury and stromal cell-derived factor-1 alpha on rat cardiomyocytes and fibroblasts. Eur Rev Med Pharmacol Sci. 2015;19:1252–7.PubMed
39.
go back to reference Uosaki H, Magadum A, Seo K, Fukushima H, Takeuchi A, Nakagawa Y, et al. Identification of chemicals inducing cardiomyocyte proliferation in developmental stage-specific manner with pluripotent stem cells. Circ Cardiovasc Genet. 2013;6:624–33.PubMedCentralCrossRefPubMed Uosaki H, Magadum A, Seo K, Fukushima H, Takeuchi A, Nakagawa Y, et al. Identification of chemicals inducing cardiomyocyte proliferation in developmental stage-specific manner with pluripotent stem cells. Circ Cardiovasc Genet. 2013;6:624–33.PubMedCentralCrossRefPubMed
40.
go back to reference Engel FB, Schebesta M, Duong MT, Lu G, Ren S, Madwed JB, et al. P38 map kinase inhibition enables proliferation of adult mammalian cardiomyocytes. Genes Dev. 2005;19:1175–87.PubMedCentralCrossRefPubMed Engel FB, Schebesta M, Duong MT, Lu G, Ren S, Madwed JB, et al. P38 map kinase inhibition enables proliferation of adult mammalian cardiomyocytes. Genes Dev. 2005;19:1175–87.PubMedCentralCrossRefPubMed
41.
go back to reference Porrello ER, Johnson BA, Aurora AB, Simpson E, Nam YJ, Matkovich SJ, et al. Mir-15 family regulates postnatal mitotic arrest of cardiomyocytes. Circ Res. 2011;109:670–9.PubMedCentralCrossRefPubMed Porrello ER, Johnson BA, Aurora AB, Simpson E, Nam YJ, Matkovich SJ, et al. Mir-15 family regulates postnatal mitotic arrest of cardiomyocytes. Circ Res. 2011;109:670–9.PubMedCentralCrossRefPubMed
42.
go back to reference Chen J, Huang ZP, Seok HY, Ding J, Kataoka M, Zhang Z, et al. Mir-17-92 cluster is required for and sufficient to induce cardiomyocyte proliferation in postnatal and adult hearts. Circ Res. 2013;112:1557–66.PubMedCentralCrossRefPubMed Chen J, Huang ZP, Seok HY, Ding J, Kataoka M, Zhang Z, et al. Mir-17-92 cluster is required for and sufficient to induce cardiomyocyte proliferation in postnatal and adult hearts. Circ Res. 2013;112:1557–66.PubMedCentralCrossRefPubMed
43.
go back to reference Yang Y, Cheng H, Qiu Y, Dupee DK, Noonan M, Lin YD, Fisch S, Unno K, Sereti KI, Liao R. Microrna-34a plays a key role in cardiac repair and regeneration following myocardial infarction. Circ Res. 2015. Yang Y, Cheng H, Qiu Y, Dupee DK, Noonan M, Lin YD, Fisch S, Unno K, Sereti KI, Liao R. Microrna-34a plays a key role in cardiac repair and regeneration following myocardial infarction. Circ Res. 2015.
44.•
go back to reference Aguirre A, Montserrat N, Zacchigna S, Nivet E, Hishida T, Krause MN, et al. In vivo activation of a conserved microrna program induces mammalian heart regeneration. Cell Stem Cell. 2014;15:589–604. In this study, the authors identified a set of microRNAs that are critical for regulating heart regeneration in zebrafish, and by inhibiting these miRNAs, were able to extend this regenerative mechanism to a murine model in order to induce cardiomyocyte dedifferentiation and proliferation post-infarction.CrossRefPubMed Aguirre A, Montserrat N, Zacchigna S, Nivet E, Hishida T, Krause MN, et al. In vivo activation of a conserved microrna program induces mammalian heart regeneration. Cell Stem Cell. 2014;15:589–604. In this study, the authors identified a set of microRNAs that are critical for regulating heart regeneration in zebrafish, and by inhibiting these miRNAs, were able to extend this regenerative mechanism to a murine model in order to induce cardiomyocyte dedifferentiation and proliferation post-infarction.CrossRefPubMed
45.
go back to reference Liu N, Bezprozvannaya S, Williams AH, Qi X, Richardson JA, Bassel-Duby R, et al. Microrna-133a regulates cardiomyocyte proliferation and suppresses smooth muscle gene expression in the heart. Genes Dev. 2008;22:3242–54.PubMedCentralCrossRefPubMed Liu N, Bezprozvannaya S, Williams AH, Qi X, Richardson JA, Bassel-Duby R, et al. Microrna-133a regulates cardiomyocyte proliferation and suppresses smooth muscle gene expression in the heart. Genes Dev. 2008;22:3242–54.PubMedCentralCrossRefPubMed
46.
go back to reference Eulalio A, Mano M, Dal Ferro M, Zentilin L, Sinagra G, Zacchigna S, et al. Functional screening identifies mirnas inducing cardiac regeneration. Nature. 2012;492:376–81.CrossRefPubMed Eulalio A, Mano M, Dal Ferro M, Zentilin L, Sinagra G, Zacchigna S, et al. Functional screening identifies mirnas inducing cardiac regeneration. Nature. 2012;492:376–81.CrossRefPubMed
47.
go back to reference Tian Y, Liu Y, Wang T, Zhou N, Kong J, Chen L, et al. A microrna-hippo pathway that promotes cardiomyocyte proliferation and cardiac regeneration in mice. Sci Transl Med. 2015;7:279ra238.CrossRef Tian Y, Liu Y, Wang T, Zhou N, Kong J, Chen L, et al. A microrna-hippo pathway that promotes cardiomyocyte proliferation and cardiac regeneration in mice. Sci Transl Med. 2015;7:279ra238.CrossRef
48.
go back to reference Li X, Wang J, Jia Z, Cui Q, Zhang C, Wang W, et al. Mir-499 regulates cell proliferation and apoptosis during late-stage cardiac differentiation via sox6 and cyclin d1. PLoS One. 2013;8, e74504.PubMedCentralCrossRefPubMed Li X, Wang J, Jia Z, Cui Q, Zhang C, Wang W, et al. Mir-499 regulates cell proliferation and apoptosis during late-stage cardiac differentiation via sox6 and cyclin d1. PLoS One. 2013;8, e74504.PubMedCentralCrossRefPubMed
49.
go back to reference Naqvi N, Li M, Calvert JW, Tejada T, Lambert JP, Wu J, et al. A proliferative burst during preadolescence establishes the final cardiomyocyte number. Cell. 2014;157:795–807.PubMedCentralCrossRefPubMed Naqvi N, Li M, Calvert JW, Tejada T, Lambert JP, Wu J, et al. A proliferative burst during preadolescence establishes the final cardiomyocyte number. Cell. 2014;157:795–807.PubMedCentralCrossRefPubMed
50.
go back to reference Kuhn B, del Monte F, Hajjar RJ, Chang YS, Lebeche D, Arab S, et al. Periostin induces proliferation of differentiated cardiomyocytes and promotes cardiac repair. Nat Med. 2007;13:962–9.CrossRefPubMed Kuhn B, del Monte F, Hajjar RJ, Chang YS, Lebeche D, Arab S, et al. Periostin induces proliferation of differentiated cardiomyocytes and promotes cardiac repair. Nat Med. 2007;13:962–9.CrossRefPubMed
51.
go back to reference Reuter S, Soonpaa MH, Firulli AB, Chang AN, Field LJ. Recombinant neuregulin 1 does not activate cardiomyocyte DNA synthesis in normal or infarcted adult mice. PLoS One. 2014;9, e115871.PubMedCentralCrossRefPubMed Reuter S, Soonpaa MH, Firulli AB, Chang AN, Field LJ. Recombinant neuregulin 1 does not activate cardiomyocyte DNA synthesis in normal or infarcted adult mice. PLoS One. 2014;9, e115871.PubMedCentralCrossRefPubMed
52.
go back to reference Becker JR, Chatterjee S, Robinson TY, Bennett JS, Panakova D, Galindo CL, et al. Differential activation of natriuretic peptide receptors modulates cardiomyocyte proliferation during development. Development. 2014;141:335–45.PubMedCentralCrossRefPubMed Becker JR, Chatterjee S, Robinson TY, Bennett JS, Panakova D, Galindo CL, et al. Differential activation of natriuretic peptide receptors modulates cardiomyocyte proliferation during development. Development. 2014;141:335–45.PubMedCentralCrossRefPubMed
53.
go back to reference Beigi F, Schmeckpeper J, Pow-Anpongkul P, Payne JA, Zhang L, Zhang Z, et al. C3orf58, a novel paracrine protein, stimulates cardiomyocyte cell-cycle progression through the pi3k-akt-cdk7 pathway. Circ Res. 2013;113:372–80.CrossRefPubMed Beigi F, Schmeckpeper J, Pow-Anpongkul P, Payne JA, Zhang L, Zhang Z, et al. C3orf58, a novel paracrine protein, stimulates cardiomyocyte cell-cycle progression through the pi3k-akt-cdk7 pathway. Circ Res. 2013;113:372–80.CrossRefPubMed
54.
go back to reference Tseng AS, Engel FB, Keating MT. The gsk-3 inhibitor bio promotes proliferation in mammalian cardiomyocytes. Chem Biol. 2006;13:957–63.CrossRefPubMed Tseng AS, Engel FB, Keating MT. The gsk-3 inhibitor bio promotes proliferation in mammalian cardiomyocytes. Chem Biol. 2006;13:957–63.CrossRefPubMed
55.
go back to reference Pasumarthi KB, Nakajima H, Nakajima HO, Soonpaa MH, Field LJ. Targeted expression of cyclin d2 results in cardiomyocyte DNA synthesis and infarct regression in transgenic mice. Circ Res. 2005;96:110–8.CrossRefPubMed Pasumarthi KB, Nakajima H, Nakajima HO, Soonpaa MH, Field LJ. Targeted expression of cyclin d2 results in cardiomyocyte DNA synthesis and infarct regression in transgenic mice. Circ Res. 2005;96:110–8.CrossRefPubMed
56.
go back to reference Shapiro SD, Ranjan AK, Kawase Y, Cheng RK, Kara RJ, Bhattacharya R, et al. Cyclin a2 induces cardiac regeneration after myocardial infarction through cytokinesis of adult cardiomyocytes. Sci Transl Med. 2014;6:224ra227. Shapiro SD, Ranjan AK, Kawase Y, Cheng RK, Kara RJ, Bhattacharya R, et al. Cyclin a2 induces cardiac regeneration after myocardial infarction through cytokinesis of adult cardiomyocytes. Sci Transl Med. 2014;6:224ra227.
57.
go back to reference Ieda M, Fu JD, Delgado-Olguin P, Vedantham V, Hayashi Y, Bruneau BG, et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell. 2010;142:375–86.PubMedCentralCrossRefPubMed Ieda M, Fu JD, Delgado-Olguin P, Vedantham V, Hayashi Y, Bruneau BG, et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell. 2010;142:375–86.PubMedCentralCrossRefPubMed
Metadata
Title
Harnessing the Induction of Cardiomyocyte Proliferation for Cardiac Regenerative Medicine
Authors
Arun Sharma, BS
Yuan Zhang
Sean M. Wu, MD PhD
Publication date
01-10-2015
Publisher
Springer US
Published in
Current Treatment Options in Cardiovascular Medicine / Issue 10/2015
Print ISSN: 1092-8464
Electronic ISSN: 1534-3189
DOI
https://doi.org/10.1007/s11936-015-0404-z