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:

Macrophages promote the transition from myocardial ischemia reperfusion injury to cardiac fibrosis in mice through GMCSF/CCL2/CCR2 and phenotype switching

Abstract

Following acute myocardial ischemia reperfusion (MIR), macrophages infiltrate damaged cardiac tissue and alter their polarization phenotype to respond to acute inflammation and chronic fibrotic remodeling. In this study we investigated the role of macrophages in post-ischemic myocardial fibrosis and explored therapeutic targets for myocardial fibrosis. Male mice were subjected to ligation of the left coronary artery for 30 min. We first detected the levels of chemokines in heart tissue that recruited immune cells infiltrating into the heart, and found that granulocyte-macrophage colony-stimulating factor (GMCSF) released by mouse cardiac microvascular endothelial cells (MCMECs) peaked at 6 h after reperfusion, and c-c motif chemokine ligand 2 (CCL2) released by GMCSF-induced macrophages peaked at 24 h after reperfusion. In co-culture of BMDMs with MCMECs, we demonstrated that GMCSF derived from MCMECs stimulated the release of CCL2 by BMDMs and effectively promoted the migration of BMDMs. We also confirmed that GMCSF promoted M1 polarization of macrophages in vitro, while GMCSF neutralizing antibodies (NTABs) blocked CCL2/CCR2 signaling. In MIR mouse heart, we showed that GMCSF activated CCL2/CCR2 signaling to promote NLRP3/caspase-1/IL-1β-mediated and amplified inflammatory damage. Knockdown of CC chemokine receptor 2 gene (CCR2−/−), or administration of specific CCR2 inhibitor RS102895 (5 mg/kg per 12 h, i.p., one day before MIR and continuously until the end of the experiment) effectively reduced the area of myocardial infarction, and down-regulated inflammatory mediators and NLRP3/Caspase-1/IL-1β signaling. Mass cytometry confirmed that M2 macrophages played an important role during fibrosis, while macrophage-depleted mice exhibited significantly reduced transforming growth factor-β (Tgf-β) levels in heart tissue after MIR. In co-culture of macrophages with fibroblasts, treatment with recombinant mouse CCL2 stimulated macrophages to release a large amount of Tgf-β, and promoted the release of Col1α1 by fibroblasts. This effect was diminished in BMDMs from CCR2−/− mice. After knocking out or inhibiting CCR2-gene, the levels of Tgf-β were significantly reduced, as was the level of myocardial fibrosis, and cardiac function was protected. This study confirms that the acute injury to chronic fibrosis transition after MIR in mice is mediated by GMCSF/CCL2/CCR2 signaling in macrophages through NLRP3 inflammatory cascade and the phenotype switching.

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: Macrophages infiltration during the transition from acute MIR injury to fibrosis.
Fig. 2: Cytokine levels in mouse heart tissue at different time points after MIR.
Fig. 3: MCMECs releases GMCSF under starvation treatment, inducing macrophage migration and releasing CCL2.
Fig. 4: GMCSF/CCL2/CCR2 promotes the transition from MIR injury to cardiac fibrosis through the NLRP3 signaling pathway.
Fig. 5: CCL2 reverses M1 macrophage-induced by GMCSF to the M2 macrophage, which releases Tgf-β to promote the transformation of fibroblasts into myofibroblasts.
Fig. 6: The effect of inhibiting CCR2 on fibrosis and cardiac function after MIR injury.
Fig. 7: Mass cytometry detection of the cardiac immune microenvironment status of mouse on the seventh day after MIR.
Fig. 8: Mass cytometry detection of the cardiac immune microenvironment status of mouse on the 7th day after MIR.
Fig. 9: After myocardial ischemic injury, cardiac microvascular endothelial cells release a large amount of GMCSF to attract monocytes to migrate to the heart, which differentiate into macrophages and transform into pro-inflammatory M1 phenotype under GMCSF induction, releasing a large amount of inflammatory factors and CCL2.

Similar content being viewed by others

References

  1. Prabhu SD, Frangogiannis NG. The biological basis for cardiac repair after myocardial infarction: from inflammation to fibrosis. Circ Res. 2016;119:91–112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Savarese G, Lund LH. Global public health burden of heart failure. Card Fail Rev. 2017;3:7–11.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Chiu J-J, Chien S. Effects of disturbed flow on vascular endothelium: pathophysiological basis and clinical perspectives. Physiol Rev. 2011;91:327–87.

    Article  PubMed  Google Scholar 

  4. Zuidema MY, Zhang C. Ischemia/reperfusion injury: the role of immune cells. World J Cardiol. 2010;2:325–32.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Venugopal H, Hanna A, Humeres C, Frangogiannis NG. Properties and functions of fibroblasts and myofibroblasts in myocardial infarction. Cells. 2022;11:1386.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Xu L, Sharkey D, Cantley LG. Tubular GMCSF promotes late MCP-1/CCR2-mediated fibrosis and inflammation after ischemia/reperfusion injury. J Am Soc Nephrol. 2019;30:1825–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tan-Garcia A, Lai F, Yeong JPS, Irac SE, Ng PY, Msallam R, et al. Liver fibrosis and CD206+ macrophage accumulation are suppressed by anti-GMCSF therapy. JHEP Rep. 2019;2:100062.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Anzai A, Choi JL, He S, Fenn AM, Nairz M, Rattik S, et al. The infarcted myocardium solicits GMCSF for the detrimental oversupply of inflammatory leukocytes. J Exp Med. 2017;214:3293–310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Peet C, Ivetic A, Bromage DI, Shah AM. Cardiac monocytes and macrophages after myocardial infarction. Cardiovasc Res. 2020;116:1101–12.

    Article  CAS  PubMed  Google Scholar 

  10. Duncan SE, Gao S, Sarhene M, Coffie JW, Linhua D, Bao X, et al. Macrophage activities in myocardial infarction and heart failure. Cardiol Res Pract. 2020;2020:4375127.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Chen B, Frangogiannis NG. Chemokines in myocardial infarction. J Cardiovasc Transl Res. 2021;14:35–52.

    Article  CAS  PubMed  Google Scholar 

  12. Georgakis MK, Bernhagen J, Heitman LH, Weber C, Dichgans M. Targeting the CCL2–CCR2 axis for atheroprotection. Eur Heart J. 2022;43:1799–808.

    Article  CAS  PubMed  Google Scholar 

  13. Xia Y, Frangogiannis NG. MCP-1/CCL2 as a therapeutic target in myocardial infarction and ischemic cardiomyopathy. Inflamm Allergy Drug Targets. 2007;6:101–7.

    Article  CAS  PubMed  Google Scholar 

  14. Flamini S, Sergeev P, Viana de Barros Z, Mello T, Biagioli M, Paglialunga M, et al. Glucocorticoid-induced leucine zipper regulates liver fibrosis by suppressing CCL2-mediated leukocyte recruitment. Cell Death Dis. 2021;12:421.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Braga TT, Correa-Costa M, Silva RC, Cruz MC, Hiyane MI, da Silva JS, et al. CCR2 contributes to the recruitment of monocytes and leads to kidney inflammation and fibrosis development. Inflammopharmacology. 2018;26:403–11.

    Article  CAS  PubMed  Google Scholar 

  16. Chung S, Overstreet JM, Li Y, Wang Y, Niu A, Wang S, et al. Tgf-β promotes fibrosis after severe acute kidney injury by enhancing renal macrophage infiltration. JCI insight. 2018;3:e123563.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Humeres C, Shinde AV, Hanna A, Alex L, Hernández SC, Li R, et al. Smad7 effects on Tgf-β and ErbB2 restrain myofibroblast activation and protect from postinfarction heart failure. J Clin Invest. 2022;132:e146926.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zaidi Y, Aguilar EG, Troncoso M, Ilatovskaya DV, DeLeon-Pennell KY. Immune regulation of cardiac fibrosis post myocardial infarction. Cell Signal. 2021;77:109837.

    Article  CAS  PubMed  Google Scholar 

  19. Vierhout M, Ayoub A, Naiel S, Yazdanshenas P, Revill SD, Reihani A, et al. Monocyte and macrophage derived myofibroblasts: Is it fate? A review of the current evidence. Wound Repair Regen. 2021;29:548–62.

    Article  PubMed  Google Scholar 

  20. Meng X-M, Wang S, Huang X-R, Yang C, Xiao J, Zhang Y, et al. Inflammatory macrophages can transdifferentiate into myofibroblasts during renal fibrosis. Cell Death Dis. 2016;7:e2495.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Haider N, Boscá L, Zandbergen HR, Kovacic JC, Narula N, González-Ramos S, et al. Transition of macrophages to fibroblast-like cells in healing myocardial infarction. J Am Coll Cardiol. 2019;74:3124–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ruberti OM, Rodrigues B. Estrogen deprivation and myocardial infarction: Role of aerobic exercise training, inflammation and metabolomics. Curr Cardiol Rev. 2020;16:292–305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Liu K, Jin H, Tang M, Zhang S, Tian X, Zhang M, et al. Lineage tracing clarifies the cellular origin of tissue-resident macrophages in the developing heart. J Cell Biol. 2022;221:e202108093.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Nguyen PD, Hsiao ST, Sivakumaran P, Lim SY, Dilley RJ. Enrichment of neonatal rat cardiomyocytes in primary culture facilitates long-term maintenance of contractility in vitro. Am J Physiol Cell Physiol. 2012;303:C1220–8.

    Article  CAS  PubMed  Google Scholar 

  25. Kim Y, Nurakhayev S, Nurkesh A, Zharkinbekov Z, Saparov A. Macrophage polarization in cardiac tissue repair following myocardial infarction. Int J Mol Sci. 2021;22:2715.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Dick SA, Macklin JA, Nejat S, Momen A, Clemente-Casares X, Althagafi MG, et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat Immunol. 2019;20:29–39.

    Article  CAS  PubMed  Google Scholar 

  27. Revelo XS, Parthiban P, Chen C, Barrow F, Fredrickson G, Wang H, et al. Cardiac resident macrophages prevent fibrosis and stimulate angiogenesis. Circ Res. 2021;129:1086–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bonaventura A, Montecucco F, Dallegri F. Cellular recruitment in myocardial ischaemia/reperfusion injury. Eur J Clin Invest. 2016;46:590–601.

    Article  CAS  PubMed  Google Scholar 

  29. Shapouri‐Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, et al. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 2018;233:6425–40.

    Article  PubMed  Google Scholar 

  30. Cochain C, Rodero MP, Vilar J, Recalde A, Richart AL, Loinard C, et al. Regulation of monocyte subset systemic levels by distinct chemokine receptors controls post-ischaemic neovascularization. Cardiovasc Res. 2010;88:186–95.

    Article  CAS  PubMed  Google Scholar 

  31. Germano DB, Oliveira SB, Bachi ALL, Juliano Y, Novo NF, do Amaral JB, et al. Monocyte chemokine receptors as therapeutic targets in cardiovascular diseases. Immunol Lett. 2023;256-257:1–8.

    Article  CAS  PubMed  Google Scholar 

  32. Frangogiannis NG. The inflammatory response in myocardial injury, repair, and remodelling. Nat Rev Cardiol. 2014;11:255–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sun K, Li Y-y, Jin J. A double-edged sword of immuno-microenvironment in cardiac homeostasis and injury repair. Signal Transduct Target Ther. 2021;6:79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Burke RM, Villar KNB, Small EM. Fibroblast contributions to ischemic cardiac remodeling. Cell Signal. 2021;77:109824.

    Article  CAS  PubMed  Google Scholar 

  35. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11:723–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Jaguin M, Houlbert N, Fardel O, Lecureur V. Polarization profiles of human M-CSF-generated macrophages and comparison of M1-markers in classically activated macrophages from GM-CSF and M-CSF origin. Cell Immunol. 2013;281:51–61.

    Article  CAS  PubMed  Google Scholar 

  37. Chen B, Yang Y, Yang C, Duan J, Chen L, Lu K, et al. M2 macrophage accumulation contributes to pulmonary fibrosis, vascular dilatation, and hypoxemia in rat hepatopulmonary syndrome. J Cell Physiol. 2021;236:7682–97.

    Article  CAS  PubMed  Google Scholar 

  38. Xi S, Zheng X, Li X, Jiang Y, Wu Y, Gong J, et al. Activated hepatic stellate cells induce infiltration and formation of CD163+ macrophages via CCL2/CCR2 pathway. Front Med. 2021;8:627927.

    Article  Google Scholar 

Download references

Acknowledgements

The work was supported by the National Natural Science Foundation of China (82170263, 82200282), the National Key Research and Development Program of China (2021YFA0804904), the Natural Science Foundation of Hefei City (2022043).

Author information

Authors and Affiliations

Authors

Contributions

SCS and LKM designed the experiments. SCS and CG performed most of the experiments. JX, CC, XJX, BYH and MZ performed some experiments. SCS and MZ analyzed the results. SCS, CG and LKM wrote the paper with help from other authors.

Corresponding authors

Correspondence to Chen Gong or Li-kun Ma.

Ethics declarations

Competing interests

The authors declare no competing interests.

Supplementary information

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

Shen, Sc., Xu, J., Cheng, C. et al. Macrophages promote the transition from myocardial ischemia reperfusion injury to cardiac fibrosis in mice through GMCSF/CCL2/CCR2 and phenotype switching. Acta Pharmacol Sin 45, 959–974 (2024). https://doi.org/10.1038/s41401-023-01222-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41401-023-01222-3

Keywords

Search

Quick links