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:

TRPM2 deficiency in mice protects against atherosclerosis by inhibiting TRPM2–CD36 inflammatory axis in macrophages

Subjects

An Author Correction to this article was published on 23 June 2023

An Author Correction to this article was published on 08 April 2022

A Publisher Correction to this article was published on 05 April 2022

This article has been updated

Abstract

Atherosclerosis is the major cause of ischemic heart disease and stroke, the leading causes of mortality worldwide. The central pathological features of atherosclerosis include macrophage infiltration and foam cell formation. However, the detailed mechanisms regulating these two processes are unclear. Here we show that oxidative stress-activated Ca2+-permeable transient receptor potential melastatin 2 (TRPM2) plays a critical role in atherogenesis. Both global and macrophage-specific Trpm2 deletions protect Apoe/ mice against atherosclerosis. Trpm2 deficiency reduces oxidized low-density lipoprotein (oxLDL) uptake by macrophages, thereby minimizing macrophage infiltration, foam cell formation and inflammatory responses. Activation of the oxLDL receptor CD36 induces TRPM2 activity and vice versa. In cultured macrophages, TRPM2 is activated by the CD36 ligands oxLDL and thrombospondin-1 (TSP1); deleting Trpm2 or inhibiting TRPM2 activity suppresses the activation of the CD36 signaling cascade induced by oxLDL and TSP1. Our findings establish the TRPM2–CD36 axis as a molecular mechanism underlying atherogenesis and suggest TRPM2 as a potential therapeutic target for atherosclerosis.

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: Global Trpm2 deletion protects mice against atherosclerosis.
Fig. 2: Trpm2 deletion attenuates inflammation in the aortas.
Fig. 3: Trpm2 deletion in macrophages protects mice against atherosclerosis.
Fig. 4: Deletion of Trpm2 inhibits foam cell formation.
Fig. 5: Trpm2 deletion inhibits the activation of CD36 signaling by TSP1.
Fig. 6: TRPM2 mediates the activation of CD36 signaling.
Fig. 7: TRPM2 mediates macrophage activation induced by oxLDL or TSP1.
Fig. 8: Inhibiting TRPM2 activation suppresses foam cell formation.

Similar content being viewed by others

Data availability

All data generated or analyzed are included in the main article and associated files. Source data are provided with this paper.

Change history

References

  1. Libby, P. et al. Atherosclerosis. Nat. Rev. Dis. Primers 5, 56 (2019).

    PubMed  Google Scholar 

  2. Moore, K. J., Sheedy, F. J. & Fisher, E. A. Macrophages in atherosclerosis: a dynamic balance. Nat. Rev. Immunol. 13, 709–721 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Moore, K. J. & Tabas, I. Macrophages in the pathogenesis of atherosclerosis. Cell 145, 341–355 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Silverstein, R. L., Li, W., Park, Y. M. & Rahaman, S. O. Mechanisms of cell signaling by the scavenger receptor CD36: implications in atherosclerosis and thrombosis. Trans. Am. Clin. Climatol. Assoc. 121, 206–220 (2010).

    PubMed  PubMed Central  Google Scholar 

  5. Moore, K. J. & Freeman, M. W. Scavenger receptors in atherosclerosis: beyond lipid uptake. Arterioscler. Thromb. Vasc. Biol. 26, 1702–1711 (2006).

    CAS  PubMed  Google Scholar 

  6. Baldrighi, M., Mallat, Z. & Li, X. NLRP3 inflammasome pathways in atherosclerosis. Atherosclerosis 267, 127–138 (2017).

    CAS  PubMed  Google Scholar 

  7. Cho, S. CD36 as a therapeutic target for endothelial dysfunction in stroke. Curr. Pharm. Des. 18, 3721–3730 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Hara, Y. et al. LTRPC2 Ca2+-permeable channel activated by changes in redox status confers susceptibility to cell death. Mol. Cell 9, 163–173 (2002).

    CAS  PubMed  Google Scholar 

  9. Perraud, A. L. et al. ADP-ribose gating of the calcium-permeable LTRPC2 channel revealed by Nudix motif homology. Nature 411, 595–599 (2001).

    CAS  PubMed  Google Scholar 

  10. Sano, Y. et al. Immunocyte Ca2+ influx system mediated by LTRPC2. Science 293, 1327–1330 (2001).

    CAS  PubMed  Google Scholar 

  11. Takahashi, N., Kozai, D., Kobayashi, R., Ebert, M. & Mori, Y. Roles of TRPM2 in oxidative stress. Cell Calcium 50, 279–287 (2011).

    CAS  PubMed  Google Scholar 

  12. Desai, B. N. & Leitinger, N. Purinergic and calcium signaling in macrophage function and plasticity. Front. Immunol. 5, 580 (2014).

    PubMed  PubMed Central  Google Scholar 

  13. Syed Mortadza, S. A., Wang, L., Li, D. & Jiang, L.-H. TRPM2 channel-mediated ROS-sensitive Ca2+ signaling mechanisms in immune cells. Front. Immunol. 6, 407 (2015).

    PubMed  PubMed Central  Google Scholar 

  14. Di, A. et al. The redox-sensitive cation channel TRPM2 modulates phagocyte ROS production and inflammation. Nat. Immunol. 13, 29–34 (2011).

    PubMed  PubMed Central  Google Scholar 

  15. Wolf, D. & Ley, K. Immunity and inflammation in atherosclerosis. Circ. Res. 124, 315–327 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Arida, A., Protogerou, A. D., Kitas, G. D. & Sfikakis, P. P. Systemic inflammatory response and atherosclerosis: the paradigm of chronic inflammatory rheumatic diseases. Int. J. Mol. Sci. 19, 1890 (2018).

    PubMed  PubMed Central  Google Scholar 

  17. Sheedy, F. J. et al. CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat. Immunol. 14, 812–820 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Tsiantoulas, D. et al. APRIL limits atherosclerosis by binding to heparan sulfate proteoglycans. Nature 597, 92–96 (2021).

    CAS  PubMed  Google Scholar 

  19. Fonfria, E. et al. TRPM2 is elevated in the tMCAO stroke model, transcriptionally regulated, and functionally expressed in C13 microglia. J. Recept. Signal Transduct. Res. 26, 179–198 (2006).

    CAS  PubMed  Google Scholar 

  20. Belrose, J. C. & Jackson, M. F. TRPM2: a candidate therapeutic target for treating neurological diseases. Acta Pharmacol. Sin. 39, 722–732 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Deshmane, S. L., Kremlev, S., Amini, S. & Sawaya, B. E. Monocyte chemoattractant protein-1 (MCP-1): an overview. J. Interferon Cytokine Res. 29, 313–326 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Nowak, W. N., Deng, J., Ruan, X. Z. & Xu, Q. Reactive oxygen species generation and atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 37, e41–e52 (2017).

    CAS  PubMed  Google Scholar 

  23. Buttery, L. D. et al. Inducible nitric oxide synthase is present within human atherosclerotic lesions and promotes the formation and activity of peroxynitrite. Lab. Invest. 75, 77–85 (1996).

    CAS  PubMed  Google Scholar 

  24. Zhong, Z. et al. TRPM2 links oxidative stress to NLRP3 inflammasome activation. Nat. Commun. 4, 1611 (2013).

    PubMed  Google Scholar 

  25. Duewell, P. et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464, 1357–1361 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Park, Y. M., Febbraio, M. & Silverstein, R. L. CD36 modulates migration of mouse and human macrophages in response to oxidized LDL and may contribute to macrophage trapping in the arterial intima. J. Clin. Invest. 119, 136–145 (2009).

    CAS  PubMed  Google Scholar 

  27. Barrett, T. J. Macrophages in atherosclerosis regression. Arterioscler. Thromb. Vasc. Biol. 40, 20–33 (2020).

    CAS  PubMed  Google Scholar 

  28. Miller, B. A. et al. TRPM2 channels protect against cardiac ischemia-reperfusion injury: role of mitochondria. J. Biol. Chem. 289, 7615–7629 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Ferron, M. & Vacher, J. Targeted expression of Cre recombinase in macrophages and osteoclasts in transgenic mice. Genesis 41, 138–145 (2005).

    CAS  PubMed  Google Scholar 

  30. Beckers, C. M. L. et al. Cre/lox studies identify resident macrophages as the major source of circulating coagulation factor XIII-A. Arterioscler. Thromb. Vasc. Biol. 37, 1494–1502 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Yoshimura, T., Robinson, E. A., Tanaka, S., Appella, E. & Leonard, E. J. Purification and amino acid analysis of two human monocyte chemoattractants produced by phytohemagglutinin-stimulated human blood mononuclear leukocytes. J. Immunol. 142, 1956–1962 (1989).

    CAS  PubMed  Google Scholar 

  32. Harris, J., VanPatten, S., Deen, N. S., Al-Abed, Y. & Morand, E. F. Rediscovering MIF: new tricks for an old cytokine. Trends Immunol. 40, 447–462 (2019).

    CAS  PubMed  Google Scholar 

  33. Zhao, M. et al. Activation of the p38 MAP kinase pathway is required for foam cell formation from macrophages exposed to oxidized LDL. APMIS 110, 458–468 (2002).

    CAS  PubMed  Google Scholar 

  34. Ricci, R. et al. Requirement of JNK2 for scavenger receptor A-mediated foam cell formation in atherogenesis. Science 306, 1558–1561 (2004).

    CAS  PubMed  Google Scholar 

  35. Rahaman, S. O. et al. A CD36-dependent signaling cascade is necessary for macrophage foam cell formation. Cell Metab. 4, 211–221 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Wang, Y., Wang, G. Z., Rabinovitch, P. S. & Tabas, I. Macrophage mitochondrial oxidative stress promotes atherosclerosis and nuclear factor-κB-mediated inflammation in macrophages. Circ. Res. 114, 421–433 (2014).

    CAS  PubMed  Google Scholar 

  37. Yan, B., Bengtson, C. P., Buchthal, B., Hagenston, A. M. & Bading, H. Coupling of NMDA receptors and TRPM4 guides discovery of unconventional neuroprotectants. Science 370, eaay3302 (2020).

    CAS  PubMed  Google Scholar 

  38. Dugan, L. L. et al. Mitochondrial production of reactive oxygen species in cortical neurons following exposure to N-methyl-d-aspartate. J. Neurosci. 15, 6377–6388 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Hoffmann, A., Kann, O., Ohlemeyer, C., Hanisch, U.-K. & Kettenmann, H. Elevation of basal intracellular calcium as a central element in the activation of brain macrophages (microglia): suppression of receptor-evoked calcium signaling and control of release function. J. Neurosci. 23, 4410–4419 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Yoshimura, T. The chemokine MCP-1 (CCL2) in the host interaction with cancer: a foe or ally? Cell. Mol. Immunol. 15, 335–345 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Yamamoto, S. et al. TRPM2-mediated Ca2+ influx induces chemokine production in monocytes that aggravates inflammatory neutrophil infiltration. Nat. Med. 14, 738–747 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Chu, L.-Y., Ramakrishnan, D. P. & Silverstein, R. L. Thrombospondin-1 modulates VEGF signaling via CD36 by recruiting SHP-1 to VEGFR2 complex in microvascular endothelial cells. Blood 122, 1822–1832 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Lopez-Dee, Z., Pidcock, K. & Gutierrez, L. S. Thrombospondin-1: multiple paths to inflammation. Mediators Inflamm. 2011, 296069 (2011).

    PubMed  PubMed Central  Google Scholar 

  44. Ganguly, R. et al. TSP-1 (thrombospondin-1) deficiency protects ApoE−/− mice against leptin-induced atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 41, e112–e127 (2021).

    CAS  PubMed  Google Scholar 

  45. Ganguly, R. et al. Oral chromium picolinate impedes hyperglycemia-induced atherosclerosis and inhibits proatherogenic protein TSP-1 expression in STZ-induced type 1 diabetic ApoE-/- mice. Sci. Rep. 7, 45279 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Du, J., Xie, J. & Yue, L. Intracellular calcium activates TRPM2 and its alternative spliced isoforms. Proc. Natl Acad. Sci. USA 106, 7239–7244 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Sumoza-Toledo, A. & Penner, R. TRPM2: a multifunctional ion channel for calcium signalling. J. Physiol. 589, 1515–1525 (2011).

    CAS  PubMed  Google Scholar 

  48. Leung, A. W. Y., Chan, R. S. M., Sea, M. M. M. & Woo, J. An overview of factors associated with adherence to lifestyle modification programs for weight management in adults. Int. J. Environ. Res. Public Health 14, 922 (2017).

    PubMed  PubMed Central  Google Scholar 

  49. Jeurissen, M. L. J. et al. Prevention of oxLDL uptake leads to decreased atherosclerosis in hematopoietic NPC1-deficient Ldlr−/− mice. Atherosclerosis 255, 59–65 (2016).

    CAS  PubMed  Google Scholar 

  50. Rahaman, S. O., Zhou, G. & Silverstein, R. L. Vav protein guanine nucleotide exchange factor regulates CD36 protein-mediated macrophage foam cell formation via calcium and dynamin-dependent processes. J. Biol. Chem. 286, 36011–36019 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Murakami, T. et al. Critical role for calcium mobilization in activation of the NLRP3 inflammasome. Proc. Natl Acad. Sci. USA 109, 11282–11287 (2012).

  52. Fernandez, D. M. & Giannarelli, C. Immune cell profiling in atherosclerosis: role in research and precision medicine. Nat. Rev. Cardiol. 19, 43–58 (2022).

    PubMed  Google Scholar 

  53. Lin, J.-D. et al. Single-cell analysis of fate-mapped macrophages reveals heterogeneity, including stem-like properties, during atherosclerosis progression and regression. JCI Insight 4, e124574 (2019).

    PubMed  PubMed Central  Google Scholar 

  54. Miller, B. A. et al. The second member of transient receptor potential-melastatin channel family protects hearts from ischemia-reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 304, H1010–H1022 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Paigen, B., Morrow, A., Holmes, P. A., Mitchell, D. & Williams, R. A. Quantitative assessment of atherosclerotic lesions in mice. Atherosclerosis 68, 231–240 (1987).

    CAS  PubMed  Google Scholar 

  56. Amini-Nik, S. et al. β-Catenin-regulated myeloid cell adhesion and migration determine wound healing. J. Clin. Invest. 124, 2599–2610 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Du, J. et al. TRPM7-mediated Ca2+ signals confer fibrogenesis in human atrial fibrillation. Circ. Res. 106, 992–1003 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Sun, Y. et al. A human platelet receptor protein microarray identifies the high affinity immunoglobulin E receptor subunit α (FcεR1α) as an activating platelet endothelium aggregation receptor 1 (PEAR1) ligand. Mol. Cell. Proteomics 14, 1265–1274 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank A. M. Scharenberg (University of Washington) for providing the TRPM2 plasmid. CD36-bio-His was a gift from G. Wright (Addgene plasmid no. 52025; http://n2t.net/addgene:52025; Research Resource Identifier: Addgene_52025)58. We thank Q. Lin (University of Connecticut) for preparing the artistic graphic works for our study. This work was partially supported by the National Institutes of Health (no. R01-HL143750) and American Heart Association (no. 19TPA34890022) to L.Y. and Canadian Institutes of Health Research (no. MOP 86655) to J.V.

Author information

Authors and Affiliations

Authors

Contributions

L.Y. conceived the research. P.Z. designed and performed the in vitro experiments. Z.Y. and J.F. performed most of the in vivo experiments. A.S.Y. conducted some of the in vitro experiments. J. V., B. M. and Y.M. generated the transgenic mice. E.R.J. and P.Z. conducted the flow cytometry experiments. P.Z. and L.Y. wrote the manuscript with contributions from all authors.

Corresponding author

Correspondence to Lixia Yue.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks Lin-Hua Jiang and the other, 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.

Integrated supplementary information

Extended Data Fig. 1 Knockout of Trpm2 in Apoe-/- mice.

(a) Representative PCR genotyping results showing a 514 bp and 740 bp products for WT and M2KO mice. (b) Representative WB analysis of TRPM2 expression in macrophages isolated from Apoe single knockout (WT (n = 3)) and Apoe / Trpm2 double knockout (M2KO (n = 3)) mice (c-e) Representative recording (c, I-V curve; d, time-current trace) and quantification of TRPM2 current in macrophages isolated from Apoe single knockout (WT) and Apoe / Trpm2 double knockout (M2KO) mice. ACA is a TRPM2 blocker. (***: p < 0.001; unpaired t test; mean ± SEM) (f) Graphic illustration showing the atherosclerotic area chosen for taking images of F4/80&CD80 staining in Fig. 1i and Fig. 3h. (g) Representative WB analysis of TRPM2 expression in macrophages isolated from Trpm2fl/flCd11b-cre- (n = 3) and Trpm2fl/flCd11b-cre+ mice (n = 3) with Apoe knockout. (h, i) Representative recording and quantification of TRPM2 current in macrophages isolated from Trpm2fl/flCd11b-cre- and Trpm2fl/flCd11b-cre+ mice with Apoe knockout.

Source data

Extended Data Fig. 2 Expression of TRPM2 is increased during atherogenesis.

(a, b) Representative WB analysis of TRPM2 expression in aorta from wild-type mice (WT) with or without the treatment of high-fat diet (HFD) for 4 months (n = 6/group). (c, d) Representative WB analysis of TRPM2 expression in macrophages isolated from WT mice with or without the treatment of oxLDL (50 µg/ml) for 24 h (n = 6/group). (***: p < 0.001; unpaired t test, two-tailed; mean ± SEM)

Source data

Extended Data Fig. 3 In vitro macrophage migration and emigration assay.

(a) Graphic illustration of in vitro examination of macrophage infiltration across endothelial cells induced by MCP1. Aorta-derived endothelial cells were plated on the transwell inserts (pore size: 12 µm) for 3-5 days. Bone marrow derived macrophages were added into the upper chamber after endothelial cells completely covered the upper surface of transwells. After 4 h, F4/80 and CD80 staining of macrophages in the lower chamber was performed as in Fig. 2l. (b) Graphic illustration of in vitro examination of macrophage emigration across endothelial cells induced by MCP1. Aorta-derived endothelial cells were plated on the transwell inserts (pore size: 12 µm) for 3-5 days. Bone marrow derived macrophages preloaded with oxLDL for 24 h were added into the upper chamber after endothelial cells completely covered the upper surface of transwells. After 24 h, F4/80 and CD80 staining of macrophages in lower chamber was performed as in Fig. 2n. (c) In vitro macrophage migration assay induced by MIF instead of MCP1 as shown in Extended Data Fig. 3a. l, F4/80 and CD80 staining of macrophages in the lower chamber (Red: F4/80; Blue: DAPI; Green: CD80). (d), Quantification of the number of infiltrated macrophages within a x 10 field. 6 dishes from each group were chosen for quantification. (***: p < 0.001; two-way ANOVA, Bonferroni’s test; mean ± SEM).

Source data

Extended Data Fig. 4 TRPM2 is required for CD36 activation in macrophages induced by oxLDL.

(a) Quantification of Fig. 4a by counting percentage of Oil red O staining macrophages (n = 8/group). (b, c) 30-min oxLDL treatment (50 µg/ml) induce the activation of CD36 signaling without upregulating CD36 expression. Representative WB analysis of CD36, pFyn, pJNK and pp38 expression in macrophages after oxLDL treatment for 30 min (n = 6/group). (d, e) NADPH oxidase inhibitor apocynin does not inhibit CD36 activation induced by 24-h oxLDL treatment (50 µg/ml) in macrophages isolated from wild-type (WT) mice. Representative WB analysis of CD36, pFyn, pJNK and pp38 expression in macrophages after oxLDL treatment for 24 h (n = 6/group). (f) Quantification of Fig. 4k by counting percentage of Oil red O staining macrophages (n = 6/group). (g) Quantification of Fig. 4o by counting percentage of Oil red O staining macrophages (n = 8/group). (h) A set of original Fura-2 real time recording traces without normalization during oxLDL treatment as in Fig. 4o. (i, j) Trpm2 deletion does not influence the production of MCP1/MIF in endothelial cells isolated from aorta in response to 24-h oxLDL treatment (50 µg/ml). Representative WB analysis of MCP1 and MIF expression in endothelial cells after oxLDL treatment for 24 h (n = 6/group). (ns: no statistical significance; *: p < 0.05; **: p < 0.01; ***: p < 0.001; ANOVA, two-way, Bonferroni’s test; mean ± SEM).

Source data

Extended Data Fig. 5 TRPM2 is required for CD36 activation in macrophages induced by TSP1.

(a, b) Representative WB analysis of TRPM2 expression in macrophages isolated from WT mice with or without the treatment of TSP1 (10 µg/ml) for 24 h (n = 6/group). (c, d) 30-min TSP1 (10 µg/ml) treatment induce the activation of CD36 signaling without upregulating CD36 expression. Representative WB analysis of CD36, pFyn, pJNK and pp38 expression in macrophages after TSP1 treatment for 30 min (n = 6/group). (e, f) NADPH oxidase inhibitor apocynin does not inhibit CD36 activation induced by 24-h TSP1 (10 µg/ml) treatment in macrophages isolated from wild-type (WT) mice. Representative WB analysis of CD36, pFyn, pJNK and pp38 expression in macrophages after TSP1 treatment for 24 h. (g) A set of original Fura-2 real time recording traces without normalization during TSP1 treatment as in Fig. 5g. (h, i) Trpm2 deletion does not influence the production of MCP1/MIF in endothelial cells isolated from aorta in response to 24-h TSP1 (10 µg/ml) treatment. Representative WB analysis of MCP1 and MIF expression in endothelial cells after TSP1 treatment for 24 h (n = 6/group). (ns: no statistical significance; *: p < 0.05; **: p < 0.01; ***: p < 0.001; ANOVA, two-way, Bonferroni’s test; mean ± SEM).

Source data

Extended Data Fig. 6 Different inhibitors suppressed the activation of TRPM2 by oxLDL or TSP1 treatment.

(a, b) Representative recording of TRPM2 current in HEK293T cells transfected with CD36 and TRPM2 during oxLDL treatment (50 µg/ml) as in a, and during TSP1 treatment (10 µg/ml) as in b. Transfected cells were treated with different inhibitors as indicated before current recording.

Source data

Extended Data Fig. 7 Inhibition of CD36 or TRPM2 did not produce additional inhibitory effect on M2KO macrophages.

(a) Representative WB analysis of the expression of CD36, pFyn, Fyn, pJNK, JNK, pp38 and p38 in isolated macrophages from M2KO mice after oxLDL treatment (50 µg/ml). Macrophages were treated with different inhibitors as indicated before protein extraction. (b) Quantification of WB bands (n = 3/group). (c) Representative WB analysis of the expression of CD36, pFyn, Fyn, pJNK, JNK, pp38 and p38 in isolated macrophages from M2KO mice after TSP1 treatment (10 µg/ml). Macrophages were treated with different inhibitors as indicated before protein extraction. (d) Quantification of WB bands (n = 3/group). (e) Representative WB analysis of the expression of iNOS in isolated macrophages from M2KO mice after oxLDL treatment (50 µg/ml). Macrophages were treated with different inhibitors as indicated before protein extraction. (f) Representative WB analysis of the expression of iNOS in isolated macrophages from M2KO mice after TSP1 treatment (10 µg/ml). Macrophages were treated with different inhibitors as indicated before protein extraction. (g) Quantification of WB bands (n = 3/group). (h) Quantification of WB bands (n = 3/group). (i) Representative WB analysis of the expression of MCP1 and MIF in isolated macrophages from M2KO mice after oxLDL treatment (50 μg/ml). Macrophages were treated with different inhibitors as indicated before protein extraction. (j) Representative WB analysis of the expression of MCP1 and MIF in isolated macrophages from M2KO mice after TSP1 treatment (10 μg/ml). Macrophages were treated with different inhibitors as indicated before protein extraction. (k) Quantification of WB bands (n = 3/group). (l) Quantification of WB bands (n = 3/group). (ns: no statistical significance; ANOVA, two-way, Bonferroni’s test; mean ± SEM).

Source data

Extended Data Fig. 8 Graphic abstract, the activation of CD36 and TRPM2 form a positive feedback loop in atherogenesis.

(a) Global Trpm2 deletion and macrophage-specific Trpm2 deletion protect against atherosclerosis in Apoe-/- mice fed with a high-fat diet (HFD). (b) Trpm2 deficiency in macrophages inhibits atherogenesis by inhibiting macrophage infiltration and minimizing foam cell formation. (c) TRPM2 activation is required for CD36-induced oxLDL uptake and subsequent inflammatory responses in macrophages. (d) The ligands of CD36, oxLDL and TSP1, activate TRPM2, thereby perpetuating TRPM2-CD36 inflammatory cycle in atherogenesis cascade. (e) Our data establish TRPM2-CD36 axis in macrophages as an important atherogenesis mechanism and TRPM2 as a promising therapeutic target for atherosclerosis.

Supplementary information

Supplementary Information

Flow cytometry gating strategy for Figs. 1 and 3 and all uncropped original scanned gel images in the Extended Data figures.

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Current traces and statistical source data.

Source Data Fig. 2

Uncropped gels.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Uncropped gels.

Source Data Fig. 4

Fura2 imaging traces and statistical source data.

Source Data Fig. 4

Uncropped gels.

Source Data Fig. 5

Fura2 imaging traces and statistical source data.

Source Data Fig. 5

Uncropped gels.

Source Data Fig. 6

Current traces and statistical source data.

Source Data Fig. 6

Uncropped gels.

Source Data Fig. 7

Fura2 imaging traces and statistical source data.

Source Data Fig. 7

Uncropped gels.

Source Data Fig. 8

Statistical source data.

Source Data Fig. 8

Uncropped gels.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical 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

Zong, P., Feng, J., Yue, Z. et al. TRPM2 deficiency in mice protects against atherosclerosis by inhibiting TRPM2–CD36 inflammatory axis in macrophages. Nat Cardiovasc Res 1, 344–360 (2022). https://doi.org/10.1038/s44161-022-00027-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-022-00027-7

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