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:

The PD-1–PD-L1 pathway maintains an immunosuppressive environment essential for neonatal heart regeneration

Abstract

The adult mouse heart responds to injury by scarring with consequent loss of contractile function, whereas the neonatal heart possesses the ability to regenerate. Activation of the immune system is among the first events upon tissue injury. It has been shown that immune response kinetics differ between regeneration and pathological remodeling, yet the underlying mechanisms of the distinct immune reactions during tissue healing remain unclear. Here we show that the immunomodulatory PD-1–PD-L1 pathway is highly active in regenerative neonatal hearts but rapidly silenced later in life. Deletion of the PD-1 receptor or inactivation of its ligand PD-L1 prevented regeneration of neonatal hearts after injury. Disruption of the pathway during neonatal cardiac injury led to increased inflammation and aberrant T cell activation, which ultimately impaired cardiac regeneration. Our findings reveal an immunomodulatory and cardioprotective role for the PD-1–PD-L1 pathway in heart regeneration and offer potential avenues for the control of adult tissue regeneration.

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: PD-1 and PD-L1 are highly enriched in regenerative hearts.
Fig. 2: PD-1 and PD-L1 are required for neonatal heart regeneration.
Fig. 3: Absence of PD-1 exacerbates the inflammatory response to MI in neonatal hearts.
Fig. 4: PD-1+ DN T cells infiltrate hearts upon P2-MI and are increased in the absence of PD-1.
Fig. 5: DN T cells become aberrantly activated in the absence of PD-1.

Similar content being viewed by others

Data availability

All data are available in the main text or the supplementary materials. All sequencing data generated from this study have been deposited in the NCBI Gene Expression Omnibus under accession no. GSE207290. Data used in this study include mm10/GRCm38, GSE207290, GSE123868, GSE153480 and GSE95755.

Code availability

All code used in this study is freely available on GitHub (https://github.com/CodeUTSW/Code_for_Paper/tree/main).

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Cui, M. et al. Dynamic transcriptional responses to injury of regenerative and non-regenerative cardiomyocytes revealed by single-nucleus RNA sequencing. Dev. Cell 53, 102–116 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Sadek, H. & Olson, E. N. Toward the goal of human heart regeneration. Cell Stem Cell 26, 7–16 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Liu, X. et al. Lymphoangiocrine signals promote cardiac growth and repair. Nature 588, 705–711 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cui, M. et al. Nrf1 promotes heart regeneration and repair by regulating proteostasis and redox balance. Nat. Commun. 12, 5270 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Aurora, A. B. et al. Macrophages are required for neonatal heart regeneration. J. Clin. Invest. 124, 1382–1392 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lavine, K. J. et al. Distinct macrophage lineages contribute to disparate patterns of cardiac recovery and remodeling in the neonatal and adult heart. Proc. Natl Acad. Sci. USA 113, E1414 (2016).

    Google Scholar 

  8. Wang, Z. et al. Resource cell-type-specific gene regulatory networks underlying murine neonatal heart regeneration at single-cell resolution. Cell. Rep. 33, 108472 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bajpai, G. et al. Tissue resident CCR2 and CCR2+ cardiac macrophages differentially orchestrate monocyte recruitment and fate specification following myocardial injury. Circ. Res. 124, 263–278 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Dick, S. A. et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat. Immunol. 20, 29–39 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. Tan, Y., Duan, X., Wang, B., Liu, X. & Zhan, Z. Murine neonatal cardiac B cells promote cardiomyocyte proliferation and heart regeneration. NPJ Regen. Med. 8, 7 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang, G. et al. ‘Default’ generation of neonatal regulatory T cells. J. Immunol. 185, 71–78 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Zacchigna, S. et al. Paracrine effect of regulatory T cells promotes cardiomyocyte proliferation during pregnancy and after myocardial infarction. Nat. Commun. 9, 2432 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Li, J. et al. Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner. Theranostics 9, 4324–4341 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Blanton, R. M., Carrillo-Salinas, F. J. & Alcaide, P. T-cell recruitment to the heart: friendly guests or unwelcome visitors? Am. J. Physiol. Heart Circ. Physiol. 317, H124–H140 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Dolejsi, T. et al. Adult T-cells impair neonatal cardiac regeneration. Eur. Heart J. https://doi.org/10.1093/eurheartj/ehac153 (2022).

  17. Li, J. et al. Specific ablation of CD4+ T-cells promotes heart regeneration in juvenile mice. Theranostics 10, 8018–8035 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wang, Z. et al. Mechanistic basis of neonatal heart regeneration revealed by transcriptome and histone modification profiling. Proc. Natl Acad. Sci. USA 116, 18455–18465 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Dong, H., Zhu, G., Tamada, K. & Chen, L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 5, 1365–1369 (1999).

    Article  CAS  PubMed  Google Scholar 

  20. Freeman, G. J. et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 192, 1027–1034 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Latchman, Y. et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat. Immunol. 2, 261–268 (2001).

    Article  CAS  PubMed  Google Scholar 

  22. Laroumanie, F. et al. CD4+ T cells promote the transition from hypertrophy to heart failure during chronic pressure overload. Circulation 129, 2111–2124 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Nevers, T. et al. Left ventricular T-cell recruitment contributes to the pathogenesis of heart failure. Circ. Heart Fail. 8, 776–787 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kallikourdis, M. et al. T cell costimulation blockade blunts pressure overload-induced heart failure. Nat. Commun. 8, 14680 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Aghajanian, H. et al. Targeting cardiac fibrosis with engineered T cells. Nature 573, 430–433 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Rurik, J. G. et al. CAR T cells produced in vivo to treat cardiac injury. Science 375, 91–96 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hayashi, T. et al. The programmed death-1 signaling axis modulates inflammation and LV structure/function in a stress-induced cardiomyopathy model. JACC Basic Transl. Sci. 7, 1120–1139 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Grabie, N. et al. Endothelial programmed death-1 ligand 1 (PD-L1) regulates CD8+ T-cell-mediated injury in the heart. Circulation 116, 2062–2071 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Spitzer, M. H. & Nolan, G. P. Mass cytometry: single cells, many features. Cell 165, 780–791 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Quaife-Ryan, G. A. et al. Multicellular transcriptional analysis of mammalian heart regeneration. Circulation 136, 1123–1139 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Keir, M. E., Freeman, G. J. & Sharpe, A. H. PD-1 regulates self-reactive CD8+ T cell responses to antigen in lymph nodes and tissues. J. Immunol. 179, 5064–5070 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Gao, S., Ho, D., Vatner, D. E. & Vatner, S. F. Echocardiography in mice. Curr. Protoc. Mouse Biol. 1, 71–83 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Suthahar, N., Meijers, W. C., Silljé, H. H. W. & de Boer, R. A. From inflammation to fibrosis—molecular and cellular mechanisms of myocardial tissue remodelling and perspectives on differential treatment opportunities. Curr. Heart Fail. Rep. 14, 235–250 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kanisicak, O. et al. Genetic lineage tracing defines myofibroblast origin and function in the injured heart. Nat. Commun. 7, 12260 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Morath, A. & Schamel, W. W. αβ and γδ T cell receptors: similar but different. J. Leukoc. Biol. 107, 1045–1055 (2020).

    Article  CAS  PubMed  Google Scholar 

  36. Geltink, R. I. K., Kyle, R. L. & Pearce, E. L. Unraveling the complex interplay between T cell metabolism and function. Annu. Rev. Immunol. 36, 461–488 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Won, T. et al. Cardiac myosin-specific autoimmune T cells contribute to immune-checkpoint-inhibitor-associated myocarditis. Cell Rep. 41, 111611 (2022).

    Article  CAS  PubMed  Google Scholar 

  38. Axelrod, M. L. et al. T cells specific for α-myosin drive immunotherapy-related myocarditis. Nature 611, 818–826 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhang, W. et al. Necrotic myocardial cells release damage-associated molecular patterns that provoke fibroblast activation in vitro and trigger myocardial inflammation and fibrosis in vivo. J. Am. Heart Assoc. 4, e001993 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Sattler, S. & Rosenthal, N. The neonate versus adult mammalian immune system in cardiac repair and regeneration. Biochim. Biophys. Acta Mol. Cell Res. 1863, 1813–1821 (2016).

    Article  CAS  Google Scholar 

  41. Trajano, L. F. & Smart, N. Immunomodulation for optimal cardiac regeneration: insights from comparative analyses. NPJ Regen. Med. https://doi.org/10.1038/s41536-021-00118-2 (2021).

  42. Forte, E., Furtado, M. B. & Rosenthal, N. The interstitium in cardiac repair: role of the immune–stromal cell interplay. Nat. Rev. Cardiol. 15, 601–616 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Chen, Z., Huang, J., Kwak-Kim, J. & Wang, W. Immune checkpoint inhibitors and reproductive failures. J. Reprod. Immunol. 156, 103799 (2023).

    Article  CAS  PubMed  Google Scholar 

  44. Montero, A. M. & Huang, A. H. The regenerative capacity of neonatal tissues. Development 149, dev199819 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Nishimura, H. et al. Developmentally regulated expression of the PD-1 protein on the surface of double-negative (CD4CD8) thymocytes. Int. Immunol. 8, 773–780 (1996).

    Article  CAS  PubMed  Google Scholar 

  46. Rodríguez-Rodríguez, N. et al. Pro-inflammatory self-reactive T cells are found within murine TCR-αβ+CD4CD8PD-1+ cells. Eur. J. Immunol. 46, 1383–1391 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Sadasivam, M. et al. Activation and proliferation of PD-1+ kidney double-negative T Cells is dependent on nonclassical MHC proteins and IL-2. J. Am. Soc. Nephrol. 30, 277–292 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Velikkakam, T., Gollob, K. J. & Dutra, W. O. Double-negative T cells: setting the stage for disease control or progression. Immunology 165, 371–385 (2022).

    Article  CAS  PubMed  Google Scholar 

  49. Qin, W. et al. The diverse function of PD-1/PD-L pathway beyond cancer. Front. Immunol. 10, 1–16 (2019).

    Article  Google Scholar 

  50. Lv, H. J. et al. Impaired thymic tolerance to α-myosin directs autoimmunity to the heart in mice and humans. J. Clin. Invest. 121, 1561–1573 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gabrielsen, I. S. M. et al. Transcriptomes of antigen presenting cells in human thymus. PLoS ONE 14, e0218858 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Nishimura, H. et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 291, 319–322 (2001).

    Article  CAS  PubMed  Google Scholar 

  53. Okazaki, T. et al. Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat. Med. 9, 1477–1483 (2003).

    Article  CAS  PubMed  Google Scholar 

  54. Mahmoud, A. I., Porrello, E. R., Kimura, W., Olson, E. N. & Sadek, H. A. Surgical models for cardiac regeneration in neonatal mice. Nat. Protoc. 9, 305–311 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Porrello, E. R. et al. Regulation of neonatal and adult mammalian heart regeneration by the miR-15 family. Proc. Natl Acad. Sci. USA 110, 187–192 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Aronoff, L., Epelman, S. & Clemente-Casares, X. Isolation and identification of extravascular immune cells of the heart. J. Vis. Exp. 1, e58114 (2018).

    Google Scholar 

  57. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. 37, 38–44 (2019).

    Article  CAS  Google Scholar 

  58. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Büttner, M., Ostner, J., Müller, C. L., Theis, F. J. & Schubert, B. scCODA is a Bayesian model for compositional single-cell data analysis. Nat. Commun. 12, 6876 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Kleshchevnikov, V. et al. Cell2location maps fine-grained cell types in spatial transcriptomics. Nat. Biotechnol. 40, 661–671 (2022).

    Article  CAS  PubMed  Google Scholar 

  61. Lim, J. F., Berger, H. & Su, I.-H. Isolation and activation of murine lymphocytes. J. Vis. Exp. 2016, e54596 (2016).

    Google Scholar 

Download references

Acknowledgements

We thank S. Bezprozvannaya at the University of Texas Southwestern Medical Center (UTSW) for technical help; J. Cabrera (UTSW) for assistance on graphic design; A. Mobley and the Flow Cytometry Core at UTSW for assistance with CyTOF; the Moody Foundation Flow Cytometry Facility for assistance with cell sorting; J. Xu and Y. Jung Kim from the Children’s Research Institute for performing the Illumina sequencing; L. Straub from the Touchstone Diabetes Center at UTSW for experimental advice and scientific discussions; K. Chen for support on the analysis of CyTOF data; L. Pathira Kankanamge and S. Miriyala from Boston Children’s Hospital for assistance with scCODA single-cell and spatial transcriptome analysis, respectively; B. Li from the Lyda Hill Department of Bioinformatics; Z. Zhong from the Department of Immunology for scientific discussions; and K. O’Donnell from Department of Molecular Biology at UTSW for scientific discussions and reagents. S.V.A. is supported by a Postdoctoral Fellowship from the American Heart Association (no. 916883) and M.C. is supported by grants from the National Institutes of Health (K99HL153683 and R00HL1536833). This work was supported by grants from the National Institutes of Health (HL130253 and HL157281, to E.N.O. and R.B.-D.), the Robert A. Welch Foundation (1-0025, to E.N.O.) and the Leducq Foundation Transatlantic Network of Excellence (20CVD04, to E.N.O.).

Author information

Authors and Affiliations

Authors

Contributions

S.V.A., M.C. and E.N.O. were responsible for conceptualization. S.V.A., M.C., W.T. and E.S.-O. were responsible for the methodology. S.V.A. and M.C. were responsible for investigation. S.V.A. and M.C. were responsible for visualization. E.N.O., R.B.-D., S.V.A. and M.C. were responsible for funding acquisition. E.N.O., N.L. and R.B.-D. were responsible for supervision. S.V.A., M.C., N.L., E.N.O. and R.B.-D. were responsible for writing.

Corresponding author

Correspondence to Eric N. Olson.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks Sikander Hayat, Douglas Mann, Javid Moslehi, Nadia Rosenthal and Han Zhu 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 PD-1 and PD-L1 are expressed in the neonatal heart.

a, Expression of PD-L1 and PD-L2 in P1 mouse hearts at steady state, as measured by qPCR. Fold change expression normalized to PD-L1 is shown. n = 3 mice per group. p = 0.0026 (PD-L1 vs. PD-L2). b, t-SNE visualization of cardiac cell types identified by CyTOF represented as a composite of all samples combined. c, Median intensity of marker genes for each cardiac cell type (scaled across individual columns) in combined samples, as measured by CyTOF. d, t-SNE plots depicting PD-L1, CD45, and CD31 expression in cardiac cell types from hearts at d3 after MI or sham performed at P2 (P2-MI or P2-sham) or at P8 (P8-MI or P8-sham), as measured by CyTOF. e, Immunofluorescence of PD-L1 (red), MOMA-2 (green) and DAPI (blue) on heart sections at d1 after MI performed at P1 (P1-MI). Sections were collected at 200μm below the ligation. Scale bar, 50μm. f, Immunofluorescence of PD-L1 (red), PECAM-1 (green) and DAPI (blue) on heart sections at d1 after P8-sham. Scale bar, 50μm. g, Violin plots of PD-L1 expression in individual cells in the conditions depicted, measured by CyTOF. These values represent the raw data used to create the heat map in Fig. 1d. The boxplots contain the 25th (Q1) to 75th (Q3) percentile of the dataset (IQR, interquartile range), and the center line denotes the median value. Whiskers mark the minimum (Q1-1.5*IQR) and maximum (Q3 + 1.5*IQR) values. Statistics were calculated using a two-sided Wilcoxon test and are shown only for selected comparisons. h, PD-L1 (left) and PD-1 (right) expression in cardiomyocytes, fibroblasts, immune cells, and endothelial cells from hearts collected at d3 after MI performed at P1 (P1-MI) or at P56 (P56-MI), measured by RNA Sequencing. Data have been previously published as GSE95755. Expression is shown as count per million reads (CPM). n = 4 biologically independent samples. **p < 0.01 by two-tailed Student’s t-test, compared to the expression in corresponding cell type from P1 hearts. p = 0.0027 (PD-L1 Immune cell: P1 vs. P56), p = 0.0085 (PD-L1 Endothelial cell: P1 vs. P56), p = 0.0038 (PD-1 Immune cell: P1 vs. P56). i, t-SNE plots depicting CD3 (upper panel) and PD-1 (lower panel) expression, as measured by CyTOF. t-SNE plots are composites of all samples combined. Red circle denotes CD3+ cells. j, Absolute counts of PD-1+ T cells and PD-1+ macrophages/monocytes/neutrophils in hearts at d3 after P2-MI (n = 7 mice per group) or P8-MI (n = 9 mice per group), as measured by flow cytometry. p = 9.4 ×10−8 (P2-MI PD-1+ T cells vs. P2-MI PD-1+ macrophages/monocytes/neutrophils), p = 1.74 ×10−7 (P2-MI PD-1+ T cells vs. P8-MI PD-1+ T cells). Results are shown as mean ± s.d (h) or mean ± s.e.m (a, j); nsp > 0.05, **p < 0.01, ****p < 0.0001 by two-tailed Student’s t-test (a) and by two-way ANOVA with Fisher’s LSD test on multiple comparisons (j).

Source data

Extended Data Fig. 2 Gating strategies used for flow cytometry analyses throughout the study.

a, Gating strategy used to detect depicted innate immune cells in the heart. Tissue-resident macrophages, TRMs. b, Gating strategy used to detect depicted immune cells in the spleen. c, Gating strategy used to detect depicted T cell populations in the heart.

Extended Data Fig. 3 Ablation of the PD-1/PD-L1 pathway impairs neonatal heart regeneration.

a-c, Ejection fraction (a), left ventricular end-diastolic volume (b) and left ventricular end-systolic volume (c) of IgG- or anti-PD-L1-treated mice at 1, 2, and 3 weeks after P3-MI. IgG sham: n = 6, 5, and 5 mice at 1, 2, 3 weeks, respectively. anti-PD-L1 sham: n = 6 mice. IgG MI: n = 5, 6, and 6 mice at 1, 2, 3 weeks, respectively. anti-PD-L1 MI: n = 7 mice (a, b c). p = 0.00052 (1 week), p = 0.000008 (2 weeks), p = 0.000009 (3 weeks) (a). p = 0.004638 (2 weeks), p = 0.000823 (3 weeks) (b). p = 0.002964 (2 weeks), p = 0.000145 (3 weeks) (c). d-f, Ejection fraction (d), left ventricular end-diastolic volume (e) and left ventricular end-systolic volume (f) of Pd-1+/ and Pd-1/ mice at 1, 2, and 3 weeks after P2-MI. Pd-1+/ sham: n = 5, 5, and 4 mice at 1, 2, 3 weeks, respectively. Pd-1/ sham: n = 5 mice. Pd-1+/ MI: n = 4 mice. Pd-1/ MI: n = 6, 5, and 5 mice at 1, 2, 3 weeks, respectively (d, e, f). p = 0.000703 (1 week), p = 0.001559 (2 weeks), p = 0.021383 (3 weeks) (d). p = 0.01633 (2 weeks), p = 0.019505 (3 weeks) (e). p = 0.004214 (2 weeks), p = 0.042845 (3 weeks) (f). The same cohort of mice was measured every week. Results are shown as mean ± s.e.m; *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 by two-tailed Student’s t-test.

Source data

Extended Data Fig. 4 Ablation of PD-1 promotes cardiomyocyte apoptosis post-MI.

a,b, Immunofluorescence of TUNEL (magenta), cardiac troponin T (cTnT, green) and DAPI (blue) on heart sections from Pd-1+/ (a) and Pd-1/ (b) mice at d7 after MI performed at P2. Sections were collected at 200μm below the ligation. Scale bar, 100μm. Four Pd-1+/ biological replicates and three Pd-1-/- biological replicates are shown. Replicate, rep.

Extended Data Fig. 5 Ablation of PD-1 impairs cardiomyocyte proliferation post-MI.

a,b, Immunofluorescence of phosphorylated histone H3 (pH3, red), cardiac troponin T (cTnT, green) and DAPI (blue) on heart sections from Pd-1+/- (a) and Pd-1-/- (b) mice at d7 after MI performed at P2. Sections were collected at 200μm below the ligation. Scale bar, 100μm. Four Pd-1+/- biological replicates and three PD-1-/- biological replicates are shown. Replicate, rep.

Extended Data Fig. 6 PD-1 is required for neonatal heart regeneration.

a, Illustration showing time points of antibody injection, MI or sham and sample collection. b, c, Fractional shortening (b) and ejection fraction (c) of IgG-treated or anti-PD-1 antibody (anti-PD-1)-treated mice at 1, 2 and 3 weeks post-MI or sham. The same cohort of mice was measured every week. IgG/ anti-PD-1 sham: n = 5 mice. IgG MI: n = 8, 7, and 7 mice at 1, 2, 3 weeks, respectively. anti-PD-1 MI: n = 8 mice (b c). p = 0.0022 (1 week), p = 0.0175 (2 weeks), p = 0.0175 (3 weeks) (b). p = 0.0014 (1 week), p = 0.0227 (2 weeks), p = 0.0227 (3 weeks) (c). d, Immunofluorescence of TUNEL (magenta), cardiac troponin T (cTnT, green) and DAPI (blue) on heart sections from IgG-treated or anti-PD-1-treated mice at d7 post-MI with infarct and remote zones depicted. Sections were collected at 200μm below the ligation. Scale bar, 100μm. e, Quantification of TUNEL+ cells in (d). p = 0.002577 (Infarct: IgG vs. anti-PD-1). f, Immunofluorescence of pH3 (red), cardiac troponin T (cTnT, green) and DAPI (blue) in hearts from IgG-treated or anti-PD-1-treated mice at d7 post-MI. Sections were collected at 200μm below the ligation, and pictures were taken at the border zone of the injury. Scale bar, 100μm. g, Quantification of pH3+ cardiomyocytes from (f). p = 0.0395 (IgG vs. anti-PD-1). n = 6 mice per group (3 images counted per mouse) (e, g). Results are shown as mean ± s.e.m; *p < 0.05, **p < 0.01 by two-tailed Student’s t-test (e, g) or two-tailed Student’s t-test with correction for multiple comparisons using the Holm-Šidák method (b, c).

Source data

Extended Data Fig. 7 Identification of immune cell types by scRNA-seq.

a, Violin plots showing distribution of numbers of genes (nFeature), numbers of RNA molecules (nCount) and percentage of mitochondrial DNA (percent.mt) that were detected in Pd-1+/- or Pd-1-/- hearts at d7 after MI or sham performed at P2. Sham, Sh. b, UMAP plot of scRNA-seq data showing identified immune cell types in combined samples. c, Heat map showing the expression of top 10 marker genes of each cell type from (b) in combined samples, as measured by scRNA-seq. d, UMAP plot showing macrophage subclusters separated by sample. e, Heat map showing expression of the pro-inflammatory cytokines Il6, Il18, Il23a and Nos2, separated by cell type, genotype, and condition. Sham, Sh.

Extended Data Fig. 8 Ablation of PD-1 causes local increased immune infiltration in the heart post-MI.

a,b, a, Quantification of spatial transcriptomic data shown in Fig. 3h. Baseline cell abundance was computed by averaging cell abundance across all spatial positions. EndoEC, endocardial cell; Mono, monocyte; Macro, macrophage; DC-like, dendric cell-like cells; FB, fibroblast. b, c, Immunofluorescence of MOMA-2 (green), cardiac troponin T (cTnT, red) and DAPI (blue) on heart sections from Pd-1+/- (b) and Pd-1-/- (c) mice at d7 after MI performed at P2. Absence of cTnT staining indicates damaged myocardium. Sections were collected at 200μm below the ligation. Scale bar, 100μm. Three Pd-1+/- and three Pd-1-/- biological replicates are shown. Replicate, rep. d, Quantification of MOMA-2+ cells from (b) and (c). n = 3 mice per group. p = 0.033 (Pd-1+/- vs. Pd-1-/-). Results are shown as mean ± s.e.m; *p < 0.05, by two-tailed Student’s t-test.

Source data

Extended Data Fig. 9 TCRγδ+ PD-1+ DN T cells accumulate in hearts injured at P2.

a, Relative composition of T cell populations depicted in Fig. 4a. Double-positive, DP. Double-negative, DN. Plotted values are shown for CD8+, CD4+ and DN T cells. b, t-SNE plots depicting CD4, CD8 and PD-1 expression in T cells from hearts at d3 after MI or sham performed at P2 (P2-MI or P2-sham) or P8 (P8-MI or P8-sham), as measured by CyTOF. c, Expression of TCRγδ, TCRβ and CD90 in DN, CD4+, CD8+ T cells from hearts at d3 after P2-MI, as measured by flow cytometry. d, Percentage of DN T cells among all T cells in spleens at steady state at 2 and 8 days of age, as measured by flow cytometry. n = 5 mice per group. p = 0.0010 (P2 vs. P8). Results are shown as mean ± s.e.m; **p < 0.01 by two-tailed Student’s t-test.

Source data

Extended Data Fig. 10 Neonatal DN T cells become aberrantly activated upon disruption of the PD-1/PD-L1 axis during MI.

a,b, Absolute counts of T cell populations (a) and neutrophils (b) in hearts of IgG- or αPD-L1-treated mice, at steady state and at d5 after MI performed at P3 (P3-MI), as measured by flow cytometry. n = 6 mice per group. p = 0.0495 (DN: IgG vs. αPD-L1) (a), p = 0.0004 (IgG vs. αPD-L1) (b). c, Heat map showing expression of marker genes in double-negative (DN), double-positive (DP), CD8+ and CD4+ T cell populations, as measured by scRNA-seq. d, Expression of DN T cell marker genes [log2(fold change) >0.6, p-adj <0.01] in DN, DP, CD8+ and CD4+ T cell populations (left) and their enriched gene ontology (GO) terms (right), as measured by scRNA-seq. A complete gene list is provided in Supplementary Data 3. Red shaded area denotes p-value (-Log10 (p-value)) by two-sided Wilcoxon Rank Sum test. e, Volcano plot depicting differentially expressed genes between Pd-1-/- DN T cells and Pd-1+/- DN T cells upon MI. p-value < 0.01, two-sided Wilcoxon Rank Sum test. A complete gene list is provided in Supplementary Data 4. f, Gene ontology (GO) terms enriched in genes upregulated in Pd-1-/- MI DN T cells vs Pd-1+/- MI DN T cells. Red shaded area denotes p-value (-Log10 (p-value)) by two-sided Wilcoxon Rank Sum test. g, NK1.1 expression in DN T cells in the heart of Pd-1+/- and Pd-1-/- mice at d5 after MI performed at P2 (P2-MI), as measured by flow cytometry. Results are shown as mean ± s.e.m; *p < 0.05, ***p < 0.001 by two-way ANOVA with Šidák correction on multiple comparisons (a) or two-tailed Student’s t-test (b).

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–4 and captions for Supplementary Data files.

Reporting Summary

Supplementary Data

Supplementary Data 1–4.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vargas Aguilar, S., Cui, M., Tan, W. et al. The PD-1–PD-L1 pathway maintains an immunosuppressive environment essential for neonatal heart regeneration. Nat Cardiovasc Res 3, 389–402 (2024). https://doi.org/10.1038/s44161-024-00447-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-024-00447-7

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