Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Identification of FDA-approved drugs that induce heart regeneration in mammals

Abstract

Targeting Meis1 and Hoxb13 transcriptional activity could be a viable therapeutic strategy for heart regeneration. In this study, we performd an in silico screening to identify FDA-approved drugs that can inhibit Meis1 and Hoxb13 transcriptional activity based on the resolved crystal structure of Meis1 and Hoxb13 bound to DNA. Paromomycin (Paro) and neomycin (Neo) induced proliferation of neonatal rat ventricular myocytes in vitro and displayed dose-dependent inhibition of Meis1 and Hoxb13 transcriptional activity by luciferase assay and disruption of DNA binding by electromobility shift assay. X-ray crystal structure revealed that both Paro and Neo bind to Meis1 near the Hoxb13-interacting domain. Administration of Paro–Neo combination in adult mice and in pigs after cardiac ischemia/reperfusion injury induced cardiomyocyte proliferation, improved left ventricular systolic function and decreased scar formation. Collectively, we identified FDA-approved drugs with therapeutic potential for induction of heart regeneration in mammals.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Identification of Paro and Neo as FDA-approved drugs targeting MEIS1–HOXB13.
Fig. 2: Paro and Neo interfere with Meis1, Hoxb13 and DNA interactions.
Fig. 3: Paro increases proliferation indices in adult mice.
Fig. 4: Administration of Paro–Neo combination increases proliferation indices in adult mice.
Fig. 5: Paro–Neo alters binding patterns of Meis1–Hoxb13 to their target genes in cardiomyocytes.
Fig. 6: Paro–Neo improves LVEF after I/R.
Fig. 7: Paro–Neo prevents cardiac remodeling in I/R pig model.

Similar content being viewed by others

Data availability

All data supporting the findings in this study are included in the main article and associated files. Source Data are provided with the manuscript. The crystal structure for the Meis1 homeobox domain bound to paromomycin or neomycin fragment was deposited to the Protein Data Bank with accession codes 8VTS and 8VTT, respectively. ChIP-seq data have been deposited in the Gene Expression Omnibus (GEO) under accession number GSE240437. RNA-seq data have been deposited in the GEO under accession number GSE240519.

References

  1. Bui, A. L., Horwich, T. B. & Fonarow, G. C. Epidemiology and risk profile of heart failure. Nat. Rev. Cardiol. 8, 30–41 (2011).

    Article  PubMed  Google Scholar 

  2. Senyo, S. E. et al. Mammalian heart renewal by pre-existing cardiomyocytes. Nature 493, 433–436 (2013).

    Article  ADS  CAS  PubMed  Google Scholar 

  3. Bergmann, O. et al. Evidence for cardiomyocyte renewal in humans. Science 324, 98–102 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Poss, K. D. Getting to the heart of regeneration in zebrafish. Semin. Cell Dev. Biol. 18, 36–45 (2007).

    Article  CAS  PubMed  Google Scholar 

  5. Walsh, S., Ponten, A., Fleischmann, B. K. & Jovinge, S. Cardiomyocyte cell cycle control and growth estimation in vivo—an analysis based on cardiomyocyte nuclei. Cardiovasc. Res. 86, 365–373 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Porrello, E. R. et al. Transient regenerative potential of the neonatal mouse heart. Science 331, 1078–1080 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Soonpaa, M. H., Kim, K. K., Pajak, L., Franklin, M. & Field, L. J. Cardiomyocyte DNA synthesis and binucleation during murine development. Am. J. Physiol. 271, H2183–H2189 (1996).

    CAS  PubMed  Google Scholar 

  8. Chaudhry, H. W. et al. Cyclin A2 mediates cardiomyocyte mitosis in the postmitotic myocardium. J. Biol. Chem. 279, 35858–35866 (2004).

    Article  CAS  PubMed  Google Scholar 

  9. Soonpaa, M. H. et al. Cyclin D1 overexpression promotes cardiomyocyte DNA synthesis and multinucleation in transgenic mice. J. Clin. Invest. 99, 2644–2654 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sim, C. B. et al. Dynamic changes in the cardiac methylome during postnatal development. FASEB J. 29, 1329–1343 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Trivedi, C. M., Lu, M. M., Wang, Q. & Epstein, J. A. Transgenic overexpression of Hdac3 in the heart produces increased postnatal cardiac myocyte proliferation but does not induce hypertrophy. J. Biol. Chem. 283, 26484–26489 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Puente, B. N. et al. The oxygen-rich postnatal environment induces cardiomyocyte cell-cycle arrest through DNA damage response. Cell 157, 565–579 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Porrello, E. R. et al. miR-15 family regulates postnatal mitotic arrest of cardiomyocytes. Circ. Res. 109, 670–679 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Eulalio, A. et al. Functional screening identifies miRNAs inducing cardiac regeneration. Nature 492, 376–381 (2012).

    Article  ADS  CAS  PubMed  Google Scholar 

  15. Heallen, T. et al. Hippo pathway inhibits Wnt signaling to restrain cardiomyocyte proliferation and heart size. Science 332, 458–461 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chen, J. et al. mir-17–92 cluster is required for and sufficient to induce cardiomyocyte proliferation in postnatal and adult hearts. Circ. Res. 112, 1557–1566 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kiserud, T. Physiology of the fetal circulation. Semin. Fetal Neonatal Med. 10, 493–503 (2005).

    Article  PubMed  Google Scholar 

  18. Nguyen, N. U. N. et al. A calcineurin–Hoxb13 axis regulates growth mode of mammalian cardiomyocytes. Nature 582, 271–276 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Henley, M. J. & Koehler, A. N. Advances in targeting ‘undruggable’ transcription factors with small molecules. Nat. Rev. Drug Discov. 20, 669–688 (2021).

    Article  CAS  PubMed  Google Scholar 

  20. Arkin, M. R., Tang, Y. & Wells, J. A. Small-molecule inhibitors of protein–protein interactions: progressing toward the reality. Chem. Biol. 21, 1102–1114 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Grembecka, J. et al. Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat. Chem. Biol. 8, 277–284 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ahmed, M. S. et al. Identification of tetracycline combinations as EphB1 tyrosine kinase inhibitors for treatment of neuropathic pain. Proc. Natl Acad. Sci. USA 118, e2016265118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yin, Y. et al. Impact of cytosine methylation on DNA binding specificities of human transcription factors. Science 356, eaaj2239 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Nair, A. B. & Jacob, S. A simple practice guide for dose conversion between animals and human. J. Basic Clin. Pharm. 7, 27–31 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Musa, A. M. et al. Paro for the treatment of visceral leishmaniasis in Sudan: a randomized, open-label, dose-finding study. PLoS Negl. Trop. Dis. 4, e855 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Goeman, F. et al. Molecular imaging of nuclear factor-Y transcriptional activity maps proliferation sites in live animals. Mol. Biol. Cell 23, 1467–1474 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kocabas, F. et al. Meis1 regulates the metabolic phenotype and oxidant defense of hematopoietic stem cells. Blood 120, 4963–4972 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Liu, Y. et al. Pharmacokinetics of neomycin sulfate after intravenous and oral administrations in swine. J. Vet. Pharmacol. Ther. 44, 850–853 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Bruni, G. N. & Kralj, J. M. Membrane voltage dysregulation driven by metabolic dysfunction underlies bactericidal activity of aminoglycosides. eLife 9, e58706 (2020).

  30. Jospe-Kaufman, M., Siomin, L. & Fridman, M. The relationship between the structure and toxicity of aminoglycoside antibiotics. Bioorg. Med. Chem. Lett. 30, 127218 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zeitz, M. J. & Smyth, J. W. Translating translation to mechanisms of cardiac hypertrophy. J. Cardiovasc. Dev. Dis. 7, 9 (2020).

  32. Ye, L. et al. Cardiac repair in a porcine model of acute myocardial infarction with human induced pluripotent stem cell-derived cardiovascular cells. Cell Stem Cell 15, 750–761 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Halgren, T. A. Merck molecular force field. I. Basis, form, scope, parameterization, and performance of MMFF94. J. Comput. Chem. 17, 490–519 (1996).

    Article  CAS  Google Scholar 

  34. Naïm, M. et al. Solvated interaction energy (SIE) for scoring protein–ligand binding affinities. 1. Exploring the parameter space. J. Chem. Inf. Model. 47, 122–133 (2007).

    Article  PubMed  Google Scholar 

  35. Labute, P. Protonate3D: assignment of ionization states and hydrogen coordinates to macromolecular structures. Proteins 75, 187–205 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Hara, H. et al. Discovery of a small molecule to increase cardiomyocytes and protect the heart after ischemic injury. JACC Basic Transl. Sci. 3, 639–653 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Arsic, N. et al. Induction of functional neovascularization by combined VEGF and angiopoietin-1 gene transfer using AAV vectors. Mol. Ther. 7, 450–459 (2003).

    Article  CAS  PubMed  Google Scholar 

  39. Lam, N. T. et al. Targeting calcineurin induces cardiomyocyte proliferation in adult mice. Nat. Cardiovasc. Res. 1, 679–688 (2022).

    Article  Google Scholar 

  40. Mahmoud, A. I. et al. Meis1 regulates postnatal cardiomyocyte cell cycle arrest. Nature 497, 249–253 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2013).

    Article  PubMed  Google Scholar 

  43. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Liao, Y., Wang, J., Jaehnig, E. J., Shi, Z. & Zhang, B. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res. 47, W199–w205 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  46. Minor, W., Cymborowski, M., Otwinowski, Z. & Chruszcz, M. HKL-3000: the integration of data reduction and structure solution—from diffraction images to an initial model in minutes. Acta Crystallogr. D Biol. Crystallogr. 62, 859–866 (2006).

    Article  ADS  PubMed  Google Scholar 

  47. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  49. Afonine, P. V. et al. Joint X-ray and neutron refinement with phenix.refine. Acta Crystallogr. D Biol. Crystallogr. 66, 1153–1163 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors gratefully acknowledge the Molecular Pathology Core at The University of Texas Southwestern Medical Center (UTSW) and J. Shelton for help with histology; J. Xu and Y. J. Kim from the Next Generation Sequencing Core Facility at the Children’s Research Institute at UTSW for performing the Illumina sequencing; and E. Olson, M. Parker, Z. Wang, B. McConnell and the Whole Brain Microscopy Facility at UTSW for their support and assistance in this work. The authors thank the Structural Biology Laboratory and H. Aronovich at UTSW for support with X-ray crystallographic studies. Results shown in this report are derived from work performed at the Argonne National Laboratory and the Structural Biology Center at the Advanced Photon Source, under Department of Energy Office of Biological and Environmental Research contract DE-AC02-06CH11357. H.A.S. was supported by grants from the National Institutes of Health (NIH) (R01 HL137415-02, R01 HL147276-01, R01 HL149137-01, 1P01HL160476-01A1, R35 HL166563-01 and P01HL160488), the Cancer Prevention and Research Institute of Texas (RP160520), the Hamon Center for Regenerative Science and Medicine and the Leducq Foundation (Redox Regulation of Cardiomyocyte Renewal). M.S.A was funded by NIH R01GM119336. N.U.N.N. was supported by grants from the American Heart Association (856552 and 19POST34450039). N.V.G was supported by grants from the NIH (GM127390) and the Welch Foundation (I-1505). C.C.Z. was supported by Leukemia & Lymphoma Society grant (6629-21) and NIH (R01CA248736). Work in the M.G. laboratory was supported by European Research Council Advanced Grant 787971 ‘CuRE’; British Heart Foundation Programme Grant RG/19/11/34633; and grants 825670 ‘CardioReGenix’ and 874764 ‘REANIMA’ from the European Commissionʼs Horizon 2020 program.

Author information

Authors and Affiliations

Authors

Contributions

M.S.A. and N.U.N.N. conducted and interpreted experiments related to protein expression and purification and mouse proliferation experiments without myocardial injury. M.S.A. and C.-C.H. performed all mouse injections. N.U.N.N. and C.-C.H. performed mouse surgeries and echocardiography experiments and interpreted the results. M.S.A. and W.E. performed and interpreted non-injury mouse echocardiography. N.U.N.N., C.-C.H. and N.L. conducted mouse proliferation experiments with myocardial injury and interpreted results. N.U.N.N. and N.T.L. performed and interpreted proliferation from dispersed hearts. S.T., M.S.A., C.-C.H. and N.U.N.N. managed mouse colonies. Y.N., G.W., X.L. and J.Z. performed and interpreted the pig-related experiment. M.S.A. and A.F. designed and performed the docking simulations and scoring experiments. N.U.N.N., P.W., D.T. and M.S.A. conducted and interpreted crystallization experiments. J.P. and N.V.G. helped with crystal structure interpretation and figure generation. N.U.N.N., M.S.A., C.-C.H. and N.T.L. conducted immunofluorescent staining. M.S.A., A.F. and I.M.-M. performed and interpreted the NRVM screening and BrdU experiments. N.U.N.N. and M.S.A. conducted MADM-related experiments and interpreted results. N.U.N.N., C.-C.H., M.C. and M.D.S. conducted ChIP-seq. N.U.N.N., C.-C.H. and I.M.-M. conducted RNA-seq. Y.L., N.U.N.N., C.X., E.O., D.Z. and I.M.-M. interpreted ChIP-seq and RNA-seq results. I.S., M.T., L.Z. and M.G. performed and interpreted CycleTrack experiments. X.L. and C.-C.Z. performed and interpreted HSC experiments. N.U.N.N. and C.-C.H. performed and interpreted experiments related to protein purification, cell culture, transfection, western blotting, luciferase assays, EMSA, qPCR and co-IP. M.S.A. and N.U.N.N. designed and conducted experiments, interpreted results and contributed to manuscript preparation. H.A.S. designed the experiments, conceived the project and contributed to manuscript preparation.

Corresponding author

Correspondence to Hesham A. Sadek.

Ethics declarations

Competing interests

M.G. is founder, consultant, member of the board and equity holder in Forcefield Therapeutics, Heqet Therapeutics and Purespring Therapeutics. All other authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks the anonymous reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Identification of Paro and Neo via a structure-based drug repurposing.

a, Chemical structures of Neo and Paro. b, Docked poses for Neo (magenta) and Paro (green) against S1-S3 sites for MEIS1-HOXB13 crystal structure. Luciferase transcriptional activity assay for p15 with c, Meis1 and d, Hoxb13 against hesperidin and rutin, compared to DMSO (Ctrl). Statistical analyses: two way ANOVA with Tukey’s post-hoc test (c,d); *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Source data

Extended Data Fig. 2 Purification and crystallization of Meis1 and Hoxb13.

a-b, Size-exclusion chromatography (Superdex 200) of DBD Meis1 and Hoxb13 purified proteins, respectively. The Y axis shows the absorbance at 280 nm and the X axis shows the elution volume in ml. c-d, Purified and peak fractions of (c) Meis1 and (d) Hoxb13 were analysed by SDS-PAGE and visualised with Coomassie Blue staining. e-f, Quantification for (e) Meis1 and (f) Hoxb13 EMSA showing that Paro, Neo, and Paro–Neo combination disrupt their DNA binding capacities. g, Paro–Neo dissociation constant for Meis1-DNA binding in EMSA assays across 0.001-50 µM drug concentrations. Data are presented as mean ± s.e.m. Statistical analyses: one way ANOVA with Tukey’s post-hoc test (e, f); *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Source data

Extended Data Fig. 3 Electron density for Paro and Neo bound Meis1.

a, Crystals of Meis1 grown with 10 mM Neo in hanging drop. b, Close-up of Ribostamycin (grey) bound to Meis1 interacting domains showing the contacts of H 297, P 298, and Y 299 as well as the role of E 303 to form hydrogen-bond interactions. c, Electron density for Paro-bound Meis1: Shown in grey mesh is the ½2mFo-DFc½ electron density, contoured at 0.5 s. Shown in stick representation are H 297 (green) in chain A, H 294 (yellow) in chain D and the Ribostamycin fragment of Paro (cyan). d, Electron density for Neo-bound Meis1: shown in Grey mesh is the ½2mFo-DFc½ electron density, contoured at 0.7 s. Shown in stick representation are H 297 (green) in chain G, H 294 (yellow) in the symmetry-related chain D and the Ribostamycin fragment of neomycin (cyan). e, Ribbon cartoon representation for Hoxb13 (Cyan) and Meis1 (Orange) along with Ribostamycin (Purple) and close-up for the contributing amino acid residues from Hoxb13 and Meis1 along with Ribostamycin. This was modelled using alignment of PDB_ID; 5EGO and Paro bound to Meis1 at RMSD 0.373 A°. f, Ribbon cartoon representation for Hoxb13 (Cyan) and Meis1 (Orange) along with Ribostamycin (Purple) and close-up for the contributing amino acid residues from Hoxb13 and Meis1 along with Ribostamycin. This was modelled using alignment of PDB_ID: 5EGO and Neo bound to Meis1 at RMSD 0.373 A°.

Extended Data Fig. 4 Paro treatment promotes adult cardiomyocyte division.

a-c, (a) BW, (b) HW, and (c) HW/BW in control and Paro-treated 10-weeks CD-1 mice. Ctrl, n = 5; Paro-treated groups, n = 5. d, Representative images of immunostaining for pH3 (green), cTnT (red) and nucleus (blue), showing additional examples of mitotic cardiomyocytes in Paro-treated mice. e, Representative images of immunostaining for AurkB (green), cTnT (red) and nucleus (blue), showing additional examples of cytokinesis cardiomyocytes in Paro-treated mice. f, Representative images and quantification for pH3 and cTnT staining in Paro-treated isolated cardiomyocytes. Scale bars, 10 μm (d-f). Data are presented as mean ± s.e.m.; Student’s unpaired two-sided t-test. Data in a-c were done for n = 5 for each group. Data in f were done for n = 4 for each group. *P < 0.05, **P < 0.01, ***P < 0.001.

Source data

Extended Data Fig. 5 Paro–Neo treatment prolongs neonatal cardiomyocyte proliferation.

a, Schematic for Paro–Neo administration to neonates CD-1 pups at 300 mg kg−1, i.p from p1 to p14. Hearts were collected for histological analysis. b, (Left, upper and right) Representative images of immunostaining for pH3 (green), cTnT (red) and nucleus (blue) showing mitotic cardiomyocytes (arrowheads) in Paro–Neo-treated mice; and (left, lower) the quantification. Ctrl, n = 6; Paro–Neo-treated groups, n = 3. c, (Left, upper and right) Representative images of immunostaining for aurora B kinase (green), cTnT (red) and nucleus (blue) showing cytokinesis cardiomyocytes (arrowheads) in Paro–Neo-treated mice; and (left, lower) the quantification. Ctrl, n = 6; Paro–Neo-treated groups, n = 3. d, Representative images and quantification for CSA cardiomyocyte size from WGA (green) and nucleus (blue) staining show no difference in cell size between Paro–Neo and vehicle-treated hearts. Ctrl, n = 3; Paro–Neo-treated groups, n = 4. Data are presented as mean ± s.e.m.; Student’s unpaired two-sided t-test (b,c). Data in b-d were done for at least n = 3 for each group. *P < 0.05, **P < 0.01, ***P < 0.001. Scale bars, 10 μm (b,c) d-f and 100 μm (d).

Source data

Extended Data Fig. 6 Paro–Neo treatment promotes adult cardiomyocyte division.

a, Schematic for Paro–Neo administration to adult CD-1 mice at 300 mg kg−1, i.p. for 2 weeks. Hearts were collected for histological analysis. b-d, (b) BW, (c) HW, and (d) HW/BW in control and Paro–Neo-treated mice. e, Representative images of immunostaining for pH3 (green), cTnT (red) and nucleus (blue), showing additional examples of mitotic cardiomyocytes in Paro–Neo-treated mice. f, Representative images of immunostaining for AurkB (green), cTnT (red) and nucleus (blue), showing additional examples of cytokinesis cardiomyocytes (arrowheads) in Paro–Neo-treated mice. g, (Upper) Representative images and (lower) quantification for pH3 and cTnT staining in Paro–Neo-treated isolated cardiomyocytes. Data are presented as mean ± s.e.m.; Student’s unpaired two-sided t-test (b-d, g). Data in b-d were done for n = 5 for control group and n = 8 for Paro–Neo-treated group. Data in g were done for n = 4 for each group. *P < 0.05, **P < 0.01, ***P < 0.001. Scale bars, 10 μm (e-g).

Source data

Extended Data Fig. 7 RNA-seq with ChIP-seq guided targets.

a, Representative sequencing tracks showing Meis1 ChIP-seq peaks in vehicle-treated (blue) and Paro–Neo-treated (red) hearts at Myh6 locus demonstrating the absence of the peaks in Paro–Neo-treated samples. b, Unbiased RNA-seq (with a fold change of 1.5). Venn diagrams show the common genes of two cohorts (DiKO vs Paro–Neo-treated hearts, normalised to Ctrl). c-d, GO terms of (c) dKO and (d) Paro–Neo-treated hearts.

Extended Data Fig. 8 In vivo pharmacological effect of aminoglycosides on the expression of cell cycle inhibitors, Meis1, Hoxb13, and HSCs.

a, Neonatal CD-1 mice received twice daily injections, i.p. of PBS, Paro (100 mg kg−1), Neo (100 mg kg−1), and Paro–Neo (150 mg kg−1) starting from p14 till p28. b-d, (b) Representative WB and densitometry quantification of (c) p15/p16 and (d) p21 protein expression, n = 4 for each group. Gapdh serves as loading controls. e-f, mRNA expression of (e) Meis1 and (f) Hoxb13 following drug treatment, n = 3 for each group. g-i, (g) Representative WB and densitometry quantification of (h) Meis1 and (i) Hoxb13 protein expression, n = 3 for each group. Gapdh serves as loading controls. j-l, Paro–Neo administration increases HSC number. 10-week-old mice received daily injections of Ctrl or Paro–Neo for 14 days. k, Representative flow cytometry for Ctrl and Paro–Neo-treated samples. l, Histograms of the means of LT-HSCs. Ctrl, n = 8; Paro/Neo-treated groups, n = 7. Data are presented as mean ± s.e.m. Statistical analyses: one-way ANOVA with Tukey’s post-hoc test (c-f, h, i), Student’s unpaired two-sided t-test (l); *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Source data

Extended Data Fig. 9 Aminoglycosides prevent cardiac remodelling in MI mouse model.

a, Schematic of MI model in drug(s)-treated mice b, Serial echocardiography assessment of LVEF showing maintained LVEF post-MI in Paro- and Paro–Neo-treated mice, compared with controls. However, higher LVEF post-MI in Paro–Neo-treated mice was observed. c, Representative echocardiography images for Ctrl, Paro-, and Paro–Neo-treated hearts at 1 week (pre-injection) and 8 weeks post-MI. d-k, Serial echocardiographic measurement of Ctrl, Paro-, and Paro–Neo-treated mice. l, Masson’s trichrome staining of hearts post-MI shows a marked decrease in LV dilatation and remodelling of Paro–Neo-treated hearts, compared with Ctrl hearts. m, Paro-treated hearts show a non-significant decline in the infarcted size, while Paro–Neo-treated hearts show a significant decrease in the infarcted size midline length, compared with controls. Ctrl, n = 6; Paro, n = 5, and Paro–Neo-treated groups, n = 8. n-p, (n) BW, (o) HW, and (p) HW/BW in Ctrl, Paro-, and Paro–Neo-treated mice. Ctrl, n = 6; Paro, n = 5, and Paro–Neo-treated groups, n = 5. q, Wet to dry lung ratio shows a significant decrease in the Paro and Paro–Neo-treated mice, compared with controls. Ctrl, n = 6; Paro, n = 5, and Paro–Neo-treated groups, n = 5. Data are presented as mean ± s.e.m. Statistical analyses: two-way ANOVA with Tukey’s post-hoc test (b); one-way ANOVA with Tukey’s post-hoc test (m-q); *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Source data

Extended Data Fig. 10 Schematic of Paro and Neo inhibiting Meis1 and Hoxb13 transcriptional activity.

In-silico screening identifies the FDA-approved drugs Paromomycin and Neomycin as inhibitors of Meis1 and Hoxb13 DNA binding, thereby promoting heart regeneration.

Supplementary information

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ahmed, M.S., Nguyen, N.U.N., Nakada, Y. et al. Identification of FDA-approved drugs that induce heart regeneration in mammals. Nat Cardiovasc Res 3, 372–388 (2024). https://doi.org/10.1038/s44161-024-00450-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-024-00450-y

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing