We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

How can the adult zebrafish and neonatal mice teach us about stimulating cardiac regeneration in the human heart?

    Michela Sorbini‡

    *Author for correspondence:

    E-mail Address: m.sorbini@smd17.qmul.ac.uk

    Barts and the London School of Medicien and Dentistry, Queen Mary University of London, E1 2AD, London, UK

    Imperial College School of Medicine, SW7 2AZ, London, UK

    ‡All authors contributed equally to this work

    Search for more papers by this author

    ,
    Sammy Arab‡

    Imperial College School of Medicine, SW7 2AZ, London, UK

    ‡All authors contributed equally to this work

    Search for more papers by this author

    ,
    Tara Soni‡

    Imperial College School of Medicine, SW7 2AZ, London, UK

    ‡All authors contributed equally to this work

    Search for more papers by this author

    ,
    Angelos Frisiras‡

    Imperial College School of Medicine, SW7 2AZ, London, UK

    ‡All authors contributed equally to this work

    Search for more papers by this author

    &
    Samay Mehta‡

    Imperial College School of Medicine, SW7 2AZ, London, UK

    ‡All authors contributed equally to this work

    Search for more papers by this author

    Published Online:https://doi.org/10.2217/rme-2022-0161

    The proliferative capacity of mammalian cardiomyocytes diminishes shortly after birth. In contrast, adult zebrafish and neonatal mice can regenerate cardiac tissues, highlighting new potential therapeutic avenues. Different factors have been found to promote cardiomyocyte proliferation in zebrafish and neonatal mice; these include maintenance of mononuclear and diploid cardiomyocytes and upregulation of the proto-oncogene c-Myc. The growth factor NRG-1 controls cell proliferation and interacts with the Hippo–Yap pathway to modulate regeneration. Key components of the extracellular matrix such as Agrin are also crucial for cardiac regeneration. Novel therapies explored in this review, include intramyocardial injection of Agrin or zebrafish-ECM and NRG-1 administration. These therapies may induce regeneration in patients and should be further explored.

    Plain language summary

    The heart pumps blood across the body carrying nutrients and oxygen where they are needed. If the heart is damaged (e.g., after a heart attack), it may lose its ability to pump blood, and this can lead to heart failure, where the heart cannot meet the body's needs, leaving the affected person tired and breathless. This occurs because the human heart unfortunately has a limited ability to heal and regain function. Current therapies for heart injuries focus on minimizing the problems resulting from the injury but cannot recover damaged heart tissue. Scientists have found that in contrast to adult human hearts, the hearts of baby mice and zebrafish can repair themselves after injuries and recover normal function. This review highlights some important mechanisms that occur in the hearts of baby mice and zebrafish, which may help contribute to their regenerative abilities. These mechanisms involve small messenger chemicals that stimulate heart cells to replicate and reform normal heart tissues. Further research into these pathways may help develop new therapies for damaged human hearts and help them regain function.

    Tweetable abstract

    Unlike humans, adult zebrafish and neonatal mice can regenerate cardiac tissues. Key regenerative processes that occur in zebrafish and neonatal mice may help the development of future therapeutic avenues for many cardiovascular diseases.

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Laflamme MA, Murry CE. Heart regeneration. Nature 473(7347), 326–335 (2011).
    • 2. Torabi A, Cleland JG, Rigby AS, Sherwi N. Development and course of heart failure after a myocardial infarction in younger and older people. J. Geriatr. Cardiol. 11(1), 1–12 (2014).
    • 3. Drexler H, Fuchs M. Current limitations in treatment of heart failure: new avenues and treatment options. J. Cardiovasc. Electrophysiol. 13(Suppl. 1), S53–6 (2002).
    • 4. Tonsho M, Michel S, Ahmed Z et al. Heart transplantation: challenges facing the field. Cold Spring Harb. Perspect. Med. 4(5), a015636 (2014).
    • 5. Poss KD, Wilson LG, Keating MT. Heart regeneration in zebrafish. Science 298(5601), 2188–90 (2002).
    • 6. Porrello ER, Mahmoud AI, Simpson E et al. Transient regenerative potential of the neonatal mouse heart. Science 331(6020), 1078–1080 (2011).
    • 7. González-Rosa JM, Martín V, Peralta M et al. Extensive scar formation and regression during heart regeneration after cryoinjury in zebrafish. Development 138(9), 1663–1674 (2011).
    • 8. González-Rosa JM, Mercader N. Cryoinjury as a myocardial infarction model for the study of cardiac regeneration in the zebrafish. Nat. Protoc. 7(4), 782–788 (2012).
    • 9. Wang J, Panáková D, Kikuchi K et al. The regenerative capacity of zebrafish reverses cardiac failure caused by genetic cardiomyocyte depletion. Development 138(16), 3421–3430 (2011).
    • 10. Mukherjee D, Wagh G, Mokalled MH et al. Ccn2a is an injury-induced matricellular factor that promotes cardiac regeneration in zebrafish. Development 148(2), dev193219 (2021).
    • 11. Raya A, Koth CM, Büscher D et al. Activation of Notch signaling pathway precedes heart regeneration in zebrafish. Proc. Natl Acad. Sci. USA 100, 11889–95 (2003).
    • 12. Chablais F, Veit J, Rainer G, Jaźwińska A. The zebrafish heart regenerates after cryoinjury-induced myocardial infarction. BMC Dev. Biol. 11(1), 1–13 (2011).
    • 13. Schnabel K, Wu C-C, Kurth T, Weidinger G. Regeneration of cryoinjury induced necrotic heart lesions in zebrafish is associated with epicardial activation and cardiomyocyte proliferation. PLOS ONE 6 (4), e18503 (2011).
    • 14. Curado S, Anderson RM, Jungblut B et al. Conditional targeted cell ablation in zebrafish: a new tool for regeneration studies. Dev. Dyn. 236(4), 1025–1035 (2007).
    • 15. Sun F, Shoffner AR, Poss KD. A genetic cardiomyocyte ablation model for the study of heart regeneration in zebrafish. Cardiac Regeneration 2158, 71–80 (2021).
    • 16. Missinato MA, Zuppo DA, Watkins SC et al. Zebrafish heart regenerates after chemoptogenetic cardiomyocyte depletion. Dev. Dyn. 250(7), 986–1000 (2021).
    • 17. Kolk MVV, Meyberg D, Deuse T et al. LAD-ligation: a murine model of myocardial infarction. J. Vis. Exp. 32(14), 1438 (2009).
    • 18. Haubner BJ, Adamowicz-Brice M, Khadayate S et al. Complete cardiac regeneration in a mouse model of myocardial infarction. Aging 4(12), 966 (2012).
    • 19. Haubner BJ, Schuetz T, Penninger JM. A reproducible protocol for neonatal ischemic injury and cardiac regeneration in neonatal mice. Basic Res. Cardiol. 111(6), 64 (2016).
    • 20. Liu C, Wang L, Wang X, Hou X. A complete heart regeneration model with inflammation as a key component. Exp. Anim. 70(4), 479–487 (2021).
    • 21. Li F, Wang X, Capasso JM, Gerdes AM. Rapid transition of cardiac myocytes from hyperplasia to hypertrophy during postnatal development. J. Mol. Cell. Cardiol. 28(8), 1737–1746 (1996).
    • 22. Yutzey KE. Cardiomyocyte Proliferation: Teaching an Old Dogma New Tricks. Circ. Res. 120(4), 627–629 (2017).
    • 23. Bergmann O, Bhardwaj RD, Bernard S et al. Evidence for cardiomyocyte renewal in humans. Science 324(5923), 98–102 (2009).
    • 24. Patterson M, Barske L, Van Handel B et al. Frequency of mononuclear diploid cardiomyocytes underlies natural variation in heart regeneration. Nat. Genet. 49(9), 1346–1353 (2017).
    • 25. Alkass K, Panula J, Westman M et al. No evidence for cardiomyocyte number expansion in preadolescent mice. Cell 163(4), 1026–1036 (2015).
    • 26. González-Rosa JM, Sharpe M, Field D et al. Myocardial polyploidization creates a barrier to heart regeneration in zebrafish. Dev. Cell 44(4), 433–446.e7 (2018).
    • 27. Adler CP, Friedburg H, Herget GW et al. Variability of cardiomyocyte DNA content, ploidy level and nuclear number in mammalian hearts. Virchows Arch. 429(2-3), 159–164 (1996).
    • 28. Herget GW, Neuburger M, Plagwitz R, Adler CP. DNA content, ploidy level and number of nuclei in the human heart after myocardial infarction. Cardiovasc. Res. 36(1), 45–51 (1997).
    • 29. Bergmann O, Zdunek S, Felker A et al. Dynamics of cell generation and turnover in the human heart. Cell 161(7), 1566–1575 (2015).
    • 30. Meckert PC, Rivello HG, Vigliano C et al. Endomitosis and polyploidization of myocardial cells in the periphery of human acute myocardial infarction. Cardiovasc. Res. 67(1), 116–123 (2005).
    • 31. Mollova M, Bersell K, Walsh S et al. Cardiomyocyte proliferation contributes to heart growth in young humans. Proc. Natl Acad. Sci. USA 110(4), 1446–1451 (2013).
    • 32. Miyaoka Y, Ebato K, Kato H et al. Hypertrophy and unconventional cell division of hepatocytes underlie liver regeneration. Curr. Biol. 22(13), 1166–1175 (2012).
    • 33. Chen J, Huang Z-P, Seok HY et al. mir-17-92 cluster is required for and sufficient to induce cardiomyocyte proliferation in postnatal and adult hearts. Circ. Res. 112(12), 1557–1566 (2013).
    • 34. Porrello ER, Mahmoud AI, Simpson E et al. Regulation of neonatal and adult mammalian heart regeneration by the miR-15 family. Proc. Natl Acad. Sci. USA 110(1), 187–192 (2013). • This study investigates the important role of the miR-15 family in mice cardiac regeneration and its potential therapeutic use following cardiac injury.
    • 35. Porrello ER, Johnson BA, Aurora AB et al. MiR-15 family regulates postnatal mitotic arrest of cardiomyocytes. Circ. Res. 109(6), 670–679 (2011).
    • 36. Yin VP, Lepilina A, Smith A, Poss KD. Regulation of zebrafish heart regeneration by miR-133. Dev. Biol. 365(2), 319–327 (2012).
    • 37. Diez-Cuñado M, Wei K, Bushway PJ et al. MiRNAs that induce human cardiomyocyte proliferation converge on the Hippo pathway. Cell Rep. 23(7), 2168–2174 (2018).
    • 38. Jackson T, Allard MF, Sreenan CM et al. The c-myc proto-oncogene regulates cardiac development in transgenic mice. Mol. Cell. Biol. 10(7), 3709–3716 (1990).
    • 39. Ahuja P, Zhao P, Angelis E et al. Myc controls transcriptional regulation of cardiac metabolism and mitochondrial biogenesis in response to pathological stress in mice. J. Clin. Invest. 120(5), 1494–1505 (2010).
    • 40. Wang Y, Li Y, Feng J et al. Mydgf promotes cardiomyocyte proliferation and neonatal heart regeneration. Theranostics 10(20), 9100–9112 (2020).
    • 41. Kisby T, Lázaro I, Fisch S et al. Adenoviral mediated delivery of OSKM factors induces partial reprogramming of mouse cardiac cells in vivo. Adv. Ther. 4(2), 2000141 (2021).
    • 42. Bywater MJ, Burkhart DL, Straube J et al. Reactivation of Myc transcription in the mouse heart unlocks its proliferative capacity. Nat. Commun. 11(1), 1827 (2020). • This study explores c-Myc-induced cardiac regeneration and proposes an innovative way of enhancing it by manipulating P-TEFβ levels in mice.
    • 43. Sano M, Abdellatif M, Oh H et al. Activation and function of cyclin T–Cdk9 (positive transcription elongation factor-b) in cardiac muscle-cell hypertrophy. Nat. Med. 8(11), 1310–1317 (2002).
    • 44. Miklas JW, Levy S, Hofsteen P et al. Amino acid primed mTOR activity is essential for heart regeneration. iScience 25(1), 103574 (2022).
    • 45. Dicks S, Jürgensen L, Leuschner F et al. Cardiac regeneration and tumor growth-what do they have in common? Front. Genet. 11, 586658 (2020).
    • 46. Chen Y, Lüttmann FF, Schoger E et al. Reversible reprogramming of cardiomyocytes to a fetal state drives heart regeneration in mice. Science.373(6562), 1537–1540 (2021).
    • 47. Aharonov A, Shakked A, Umansky KB et al. ERBB2 drives YAP activation and EMT-like processes during cardiac regeneration. Nat. Cell Biol. 22(11), 1346–1356 (2020).
    • 48. Plouffe SW, Lin KC, Moore JL 3rd et al. The Hippo pathway effector proteins YAP and TAZ have both distinct and overlapping functions in the cell. J. Biol. Chem. 293(28), 11230–11240 (2018).
    • 49. Xiao Y, Dong J. The Hippo signaling pathway in cancer: a cell cycle perspective. Cancers 13(24), 6214(2021).
    • 50. Xin M, Kim Y, Sutherland LB et al. Hippo pathway effector Yap promotes cardiac regeneration. Proc. Natl Acad. Sci. USA 110(34), 13839–13844 (2013).
    • 51. Heallen T, Morikawa Y, Leach J et al. Hippo signaling impedes adult heart regeneration. Development 140(23), 4683–4690 (2013).
    • 52. Von Gise A, Lin Z, Schlegelmilch K et al. YAP1, the nuclear target of Hippo signaling, stimulates heart growth through cardiomyocyte proliferation but not hypertrophy. Proc. Natl Acad. Sci. USA 109(7), 2394–2399 (2012).
    • 53. Nishimoto M, Uranishi K, Asaka MN et al. Transformation of normal cells by aberrant activation of YAP via cMyc with TEAD. Sci. Rep. 9(1), 10933 (2019).
    • 54. Flinn MA, Jeffery BE, O’Meara CC, Link BA. Yap is required for scar formation but not myocyte proliferation during heart regeneration in zebrafish. Cardiovasc. Res. 115(3), 570–577 (2019).
    • 55. Bo B, Li S, Zhou K, Wei J. The regulatory role of oxygen metabolism in exercise-induced cardiomyocyte regeneration. Front. Cell Dev. Biol. 9, 664527 (2021).
    • 56. Fukuda R, Marín-Juez R, El-Sammak H et al. Stimulation of glycolysis promotes cardiomyocyte proliferation after injury in adult zebrafish. EMBO Rep. 21(8), e49752 (2020).
    • 57. Lopaschuk GD, Karwi QG, Tian R et al. Cardiac energy metabolism in heart failure. Circ. Res. 128(10), 1487–1513 (2021).
    • 58. Elhelaly WM, Lam NT, Hamza M et al. Redox regulation of heart regeneration: an evolutionary tradeoff. Front. Cell. Dev. Biol 4, 137 (2016).
    • 59. Puente BN, Kimura W, Muralidhar SA et al. The oxygen-rich postnatal environment induces cardiomyocyte cell-cycle arrest through DNA damage response. Cell 157(3), 565–579 (2014).
    • 60. Bai C-G, Liu X-H, Liu W-Q, Ma D-L. Regional expression of the hypoxia-inducible factor (HIF) system and association with cardiomyocyte cell cycle re-entry after myocardial infarction in rats. Heart Vessels 23(3), 193–200 (2008).
    • 61. Moslehi J, Minamishima YA, Shi J et al. Loss of hypoxia-inducible factor prolyl hydroxylase activity in cardiomyocytes phenocopies ischemic cardiomyopathy. Circulation 122(10), 1004–1016 (2010).
    • 62. Gauron C, Rampon C, Bouzaffour M et al. Sustained production of ROS triggers compensatory proliferation and is required for regeneration to proceed. Sci. Rep. 3, 2084 (2013).
    • 63. Han P, Zhou X-H, Chang N et al. Hydrogen peroxide primes heart regeneration with a derepression mechanism. Cell Res. 24(9), 1091–1107 (2014). • This study provides evidence for the role of reactive oxygen species in cardiac repair in zebrafish and identifies downstream molecules such as PTPs, which may be potential therapeutic targets for cardiac regeneration.
    • 64. Smith AM, Maguire-Nguyen KK, Rando TA et al. The protein tyrosine phosphatase 1B inhibitor MSI-1436 stimulates regeneration of heart and multiple other tissues. NPJ Regen. Med. 2, 4 (2017).
    • 65. Kimura W, Xiao F, Canseco DC et al. Hypoxia fate mapping identifies cycling cardiomyocytes in the adult heart. Nature 523(7559), 226–230 (2015).
    • 66. Faeh D, Moser A, Panczak R et al. Independent at heart: persistent association of altitude with ischaemic heart disease mortality after consideration of climate, topography and built environment. J. Epidemiol. Community Health 70(8), 798–806 (2016).
    • 67. Ye L, Qiu L, Feng B et al. Role of blood oxygen saturation during post-natal human cardiomyocyte cell cycle activities. JACC Basic Transl. Sci. 5(5), 447–460 (2020).
    • 68. Francis R, Chong J, Ramlall M et al. Effect of remote ischaemic conditioning on infarct size and remodelling in ST-segment elevation myocardial infarction patients: the CONDI-2/ERIC-PPCI CMR substudy. Basic Res. Cardiol. 116(1), 59 (2021).
    • 69. Bae J, Paltzer WG, Mahmoud AI. The role of metabolism in heart failure and regeneration. Front. Cardiovasc. Med.8, 702920 (2021)
    • 70. Tan J, Yang M, Wang H et al. Moderate heart rate reduction promotes cardiac regeneration through stimulation of the metabolic pattern switch. Cell Rep. 38(10), 110468 (2022).
    • 71. Ali RL, Hakim JB, Boyle PM et al. Arrhythmogenic propensity of the fibrotic substrate after atrial fibrillation ablation: a longitudinal study using magnetic resonance imaging-based atrial models. Cardiovasc. Res. 115(12), 1757 (2019).
    • 72. Chen Z-T, Gao Q-Y, Wu M-X et al. Glycolysis inhibition alleviates cardiac fibrosis after myocardial infarction by suppressing cardiac fibroblast activation. Front. Cardiovasc. Med. 8, 701745 (2021).
    • 73. White IA, Gordon J, Balkan W, Hare JM. Sympathetic reinnervation is required for mammalian cardiac regeneration. Circ. Res. 117(12), 990–994 (2015).
    • 74. Rodriguez-Pallares J, Parga JA, Muñoz A et al. Mechanism of 6-hydroxydopamine neurotoxicity: the role of NADPH oxidase and microglial activation in 6-hydroxydopamine-induced degeneration of dopaminergic neurons. J. Neurochem. 103(1), 145–156 (2007).
    • 75. Del Campo CV, Liaw NY, Gunadasa-Rohling M et al. Regenerative potential of epicardium-derived extracellular vesicles mediated by conserved miRNA transfer. Cardiovasc. Res. 118(2), 597–611 (2022).
    • 76. Mahmoud AI, O’Meara CC, Gemberling M et al. Nerves regulate cardiomyocyte proliferation and heart regeneration. Dev. Cell 34(4), 387–399 (2015).
    • 77. Dae MW, Herre JM, O’Connell JW et al. Scintigraphic assessment of sympathetic innervation after transmural versus nontransmural myocardial infarction. J. Am. Coll. Cardiol. 17(6), 1416–1423 (1991).
    • 78. Lepilina A, Coon AN, Kikuchi K et al. A dynamic epicardial injury response supports progenitor cell activity during zebrafish heart regeneration. Cell 127(3), 607–619 (2006).
    • 79. Huang Y, Harrison MR, Osorio A et al. Igf signaling is required for cardiomyocyte proliferation during zebrafish heart development and regeneration. PLOS ONE.8(6), e67266 (2013).
    • 80. Kikuchi K, Holdway JE, Werdich AA et al. Primary contribution to zebrafish heart regeneration by gata4(+) cardiomyocytes. Nature 464(7288), 601–605 (2010).
    • 81. Zhao L, Borikova AL, Ben-Yair R et al. Notch signaling regulates cardiomyocyte proliferation during zebrafish heart regeneration. Proc. Natl Acad. Sci. USA 111(4), 1403–1408 (2014).
    • 82. D’Uva G, Aharonov A, Lauriola M et al. ERBB2 triggers mammalian heart regeneration by promoting cardiomyocyte dedifferentiation and proliferation. Nat. Cell Biol. 17(5), 627–638 (2015).
    • 83. Honkoop H, De Bakker DE, Aharonov A et al. Single-cell analysis uncovers that metabolic reprogramming by ErbB2 signaling is essential for cardiomyocyte proliferation in the regenerating heart. Elife 8, e50163 (2019).
    • 84. Gemberling M, Karra R, Dickson AL, Poss KD. Nrg1 is an injury-induced cardiomyocyte mitogen for the endogenous heart regeneration program in zebrafish. Elife 4, e05871 (2015).
    • 85. Polizzotti BD, Ganapathy B, Walsh S et al. Neuregulin stimulation of cardiomyocyte regeneration in mice and human myocardium reveals a therapeutic window. Sci. Transl. Med. 7(281), 281ra45 (2015).
    • 86. Ma H, Yin C, Zhang Y et al. ErbB2 is required for cardiomyocyte proliferation in murine neonatal hearts. Gene 592(2), 325–330 (2016).
    • 87. Takahashi T, Ishida K, Itoh K et al. IGF-I gene transfer by electroporation promotes regeneration in a muscle injury model. Gene Ther. 10(8), 612–620 (2003).
    • 88. Ali SR, Elhelaly W, Nguyen NUN et al. Angiocrine IGFBP3 spatially coordinates IGF signaling during neonatal cardiac regeneration. bioRxiv 460522(2021).
    • 89. Sallin P, Jaźwińska A. Acute stress is detrimental to heart regeneration in zebrafish. Open Biol. 6(3), 160012(2016).
    • 90. Bassat E, Mutlak YE, Genzelinakh A et al. The extracellular matrix protein agrin promotes heart regeneration in mice. Nature 547(7662), 179–184 (2017).
    • 91. Garcia-Puig A, Mosquera JL, Jiménez-Delgado S et al. Proteomics analysis of extracellular matrix remodeling during zebrafish heart regeneration. Mol. Cell. Proteomics 18(9), 1745–1755 (2019). • This study explores the composition of extracellular matrix (ECM) in zebrafish and demonstrates that there is a sharp increase in specific ECM proteins following amputation. It highlights the differences in these ECM protein levels before and after resection and outlines their individual roles in cardiac regeneration.
    • 92. Notari M, Ventura-Rubio A, Bedford-Guaus SJ et al. The local microenvironment limits the regenerative potential of the mouse neonatal heart. Sci. Adv. 4(5), eaao5553 (2018). • The study shows that the composition of ECM in zebrafish undergoes an abundance of changes following amputation and highlights the influence of ECM handling on cardiac regeneration. The authors also highlight crucial changes in ECM stiffness during regeneration.
    • 93. Yahalom-Ronen Y, Rajchman D, Sarig R et al. Reduced matrix rigidity promotes neonatal cardiomyocyte dedifferentiation, proliferation and clonal expansion. Elife.4, e07455 (2015).
    • 94. Chen WCW, Wang Z, Missinato MA et al. Decellularized zebrafish cardiac extracellular matrix induces mammalian heart regeneration. Sci. Adv. 2(11), e1600844 (2016).
    • 95. Yutzey KE. Regenerative biology: Neuregulin 1 makes heart muscle. Nature 520(7548), 445–446 (2015).
    • 96. Pfefferli C, Jaźwińska A. The careg element reveals a common regulation of regeneration in the zebrafish myocardium and fin. Nat. Commun. 8, 15151 (2017).
    • 97. Chablais F, Jazwinska A. The regenerative capacity of the zebrafish heart is dependent on TGFβ signaling. Development 139(11), 1921–1930 (2012).
    • 98. George EL, Baldwin HS, Hynes RO. Fibronectins are essential for heart and blood vessel morphogenesis but are dispensable for initial specification of precursor cells. Blood 90(8), 3073–3081 (1997).
    • 99. Trinh LA, Stainier DYR. Fibronectin regulates epithelial organization during myocardial migration in zebrafish. Dev. Cell 6(3), 371–382 (2004).
    • 100. Jaźwińska A, Sallin P. Regeneration versus scarring in vertebrate appendages and heart. J. Pathol. 238(2), 233–246 (2016).
    • 101. Kühn B, del Monte F, Hajjar RJ et al. Periostin induces proliferation of differentiated cardiomyocytes and promotes cardiac repair. Nat. Med. 13(8), 962–969 (2007).
    • 102. Baehr A, Umansky KB, Bassat E et al. Agrin promotes coordinated therapeutic processes leading to improved cardiac repair in pigs. Circulation 142(9), 868–881 (2020).
    • 103. Chakraborty S, Njah K, Pobbati AV et al. Agrin as a mechanotransduction signal regulating YAP through the Hippo pathway. Cell Rep. 18(10), 2464–2479 (2017).
    • 104. Han C, Nie Y, Lian H et al. Acute inflammation stimulates a regenerative response in the neonatal mouse heart. Cell Res. 25(10), 1137–1151 (2015).
    • 105. Aurora AB, Porrello ER, Tan W et al. Macrophages are required for neonatal heart regeneration. J. Clin. Invest. 124(3), 1382–1392 (2014). • This study highlights the importance of the role of the immune system in regulating cardiac regeneration and specifically the differential role of macrophages in regeneration of myocardium of neonatal mice. It shows that macrophage depletion removes the ability of neonatal mice to regenerate their myocardium.
    • 106. Glaros T, Larsen M, Li L. Macrophages and fibroblasts during inflammation, tissue damage and organ injury. Front. Biosci. 14(10), 3988–3993 (2009).
    • 107. Schulz C, Gomez Perdiguero E, Chorro L et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336(6077), 86–90 (2012).
    • 108. Hashimoto D, Chow A, Noizat C et al. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 38(4), 792–804 (2013).
    • 109. Yona S, Kim K-W, Wolf Y et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38(1), 79–91 (2013).
    • 110. Ginhoux F, Greter M, Leboeuf M et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330(6005), 841–845 (2010).
    • 111. Hoeffel G, Wang Y, Greter M et al. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J. Exp. Med. 209(6), 1167–1181 (2012).
    • 112. Lavine KJ, Epelman S, Uchida K et al. Distinct macrophage lineages contribute to disparate patterns of cardiac recovery and remodeling in the neonatal and adult heart. Proc. Natl. Acad. Sci. U. S. A. 111(45), 16029–16034 (2014).
    • 113. Epelman S, Lavine KJ, Beaudin AE et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity 40(1), 91–104 (2014).
    • 114. Bajpai G, Bredemeyer A, Li W et al. Tissue resident CCR2- and CCR2+ cardiac macrophages differentially orchestrate monocyte recruitment and fate specification following myocardial injury. Circ. Res. 124(2), 263–278 (2019).
    • 115. Roberts R, DeMello V, Sobel BE. Deleterious effects of methylprednisolone in patients with myocardial infarction. Circulation 53(Suppl. 3), I204–6 (1976).
    • 116. Bevan L, Lim ZW, Venkatesh B et al. Specific macrophage populations promote both cardiac scar deposition and subsequent resolution in adult zebrafish. Cardiovasc. Res. 116(7), 1357–1371 (2020).
    • 117. Kirchmaier S, Naruse K, Wittbrodt J, Loosli F. The genomic and genetic toolbox of the teleost medaka (Oryzias latipes). Genetics 199(4), 905–918 (2015).
    • 118. Ito K, Morioka M, Kimura S et al. Differential reparative phenotypes between zebrafish and medaka after cardiac injury. Dev. Dyn. 243(9), 1106–1115 (2014).
    • 119. Lai S-L, Marín-Juez R, Moura PL et al. Reciprocal analyses in zebrafish and medaka reveal that harnessing the immune response promotes cardiac regeneration. Elife.6, 6 (2017).
    • 120. Potts HG, Stockdale WT, Mommersteeg MTM. Unlocking the secrets of the regenerating fish heart: comparing regenerative models to shed light on successful regeneration. J. Cardiovasc. Dev. Dis. 8(1), 4 (2021).
    • 121. Wodsedalek DJ, Paddock SJ, Wan TC et al. IL-13 promotes in vivo neonatal cardiomyocyte cell cycle activity and heart regeneration. Am. J. Physiol. Heart Circ. Physiol. 316(1), H24–H34 (2019).
    • 122. Hui SP, Sheng DZ, Sugimoto K et al. Zebrafish regulatory T cells mediate organ-specific regenerative programs. Dev. Cell 43(6), 659–672.e5 (2017).
    • 123. Miller DM, Thomas SD, Islam A et al. c-Myc and cancer metabolism. Clin. Cancer Res. 18(20), 5546–5553 (2012).
    • 124. Tanner JJ, Parsons ZD, Cummings AH et al. Redox regulation of protein tyrosine phosphatases: structural and chemical aspects. Antioxid. Redox Signal. 15(1), 77–97 (2011).
    • 125. Adler CP, Friedburg H. Myocardial DNA content, ploidy level and cell number in geriatric hearts: post-mortem examinations of human myocardium in old age. J. Mol. Cell. Cardiol. 18(1), 39–53 (1986).