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:

Senescent cells suppress innate smooth muscle cell repair functions in atherosclerosis

Abstract

Senescent cells (SNCs) degenerate the fibrous cap that normally prevents atherogenic plaque rupture, a leading cause of myocardial infarction and stroke. Here we explore the underlying mechanism using pharmacological or transgenic approaches to clear SNCs in the Ldlr–/– mouse model of atherosclerosis. SNC clearance reinforced fully deteriorated fibrous caps in highly advanced lesions, as evidenced by restored vascular smooth muscle cell (VSMC) numbers, elastin content and overall cap thickness. We found that SNCs inhibit VSMC promigratory phenotype switching in the first interfiber space of the arterial wall directly beneath the atherosclerotic plaque, thereby limiting lesion entry of medial VSMCs for fibrous cap assembly or reinforcement. SNCs do so by antagonizing insulin-like growth factor (IGF)-1 through the secretion of IGF-binding protein-3. These data indicate that the intermittent use of senolytic agents or IGF-binding protein-3 inhibition in combination with lipid-lowering drugs may provide therapeutic benefit in 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: ABT263 blocks fibrous cap thinning through depletion of SNCs.
Fig. 2: Fibrous caps of advanced atheromas increase VSMC content and thicken with senolysis.
Fig. 3: SNCs inhibit promigratory switching and neointimal recruitment of medial VSMCs.
Fig. 4: SNCs suppress innate repair functions of medial VSMCs beginning with the earliest stages of atherogenesis.
Fig. 5: VSMC lineage tracing approaches reveal senolysis enhances medial VSMC phenotypic switching and migration into lesions.
Fig. 6: 3MR-mediated senolysis prompts rapid VSMC migration during early atherogenesis.
Fig. 7: Senolysis depletes lesional Igfbp3 to promote promigratory VSMC phenotype switching of medial VSMCs.
Fig. 8: Igfbp3 neutralization promotes promigratory switching of VSMC in mouse and human explant atheromas.

Similar content being viewed by others

Data availability

RNA-seq data have been deposited in the Gene Expression Omnibus under the accession number GSE130382 and GSE167003. All underlying data used for generation of figures are collated in the associated source files.

References

  1. Demaria, M. et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev. Cell 31, 722–733 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Chiche, A. et al. Injury-induced senescence enables in vivo reprogramming in skeletal muscle. Cell Stem Cell 20, 407–414 (2017).

    Article  PubMed  CAS  Google Scholar 

  3. Mosteiro, L., Pantoja, C., de Martino, A. & Serrano, M. Senescence promotes in vivo reprogramming through p16(INK)(4a) and IL-6. Aging Cell https://doi.org/10.1111/acel.12711 (2018).

  4. Jeon, O. H. et al. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat. Med. 23, 775–781 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Bussian, T. J. et al. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature https://doi.org/10.1038/s41586-018-0543-y (2018).

  6. Baker, D. J. et al. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature 530, 184–189 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Childs, B. G. et al. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science 354, 472–477 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Chang, J. et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat. Med. 22, 78–83 (2016).

    Article  PubMed  CAS  Google Scholar 

  9. Zhu, Y. et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 15, 428–435 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Bittencourt, M. S. & Cerci, R. J. Statin effects on atherosclerotic plaques: regression or healing? BMC Med. 13, 260 (2015).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  11. Reith, C. & Armitage, J. Management of residual risk after statin therapy. Atherosclerosis 245, 161–170 (2016).

    Article  PubMed  CAS  Google Scholar 

  12. Kloppenburg, G. T., Grauls, G. E., Bruggeman, C. A. & Stassen, F. R. Adenoviral activin A expression prevents vein graft intimal hyperplasia in a rat model. Interact. Cardiovasc. Thorac. Surg. 8, 31–34 (2009).

    Article  PubMed  Google Scholar 

  13. Engelse, M. A. et al. Adenoviral activin a expression prevents intimal hyperplasia in human and murine blood vessels by maintaining the contractile smooth muscle cell phenotype. Circ. Res. 90, 1128–1134 (2002).

    Article  PubMed  CAS  Google Scholar 

  14. Almehmadi, A. et al. VWC2 increases bone formation through inhibiting activin signaling. Calcified Tissue Int. 103, 663–674 (2018).

    Article  CAS  Google Scholar 

  15. Lepore, J. J., Cappola, T. P., Mericko, P. A., Morrisey, E. E. & Parmacek, M. S. GATA-6 regulates genes promoting synthetic functions in vascular smooth muscle cells. Arter. Thromb. Vasc. Biol. 25, 309–314 (2005).

    Article  CAS  Google Scholar 

  16. Trovati, M. et al. Leptin and vascular smooth muscle cells. Curr. Pharm. Des. 20, 625–634 (2014).

    Article  PubMed  CAS  Google Scholar 

  17. Huang, K. et al. MicroRNA-33 protects against neointimal hyperplasia induced by arterial mechanical stretch in the grafted vein. Cardiovasc. Res. 113, 488–497 (2017).

    Article  PubMed  CAS  Google Scholar 

  18. Lagna, G. et al. Control of phenotypic plasticity of smooth muscle cells by bone morphogenetic protein signaling through the myocardin-related transcription factors. J. Biol. Chem. 282, 37244–37255 (2007).

    Article  PubMed  CAS  Google Scholar 

  19. Li, N. et al. Mutations in the histone modifier PRDM6 are associated with isolated nonsyndromic patent ductus arteriosus. Am. J. Hum. Genet. 98, 1082–1091 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Martin, E. et al. TSHZ3 and SOX9 regulate the timing of smooth muscle cell differentiation in the ureter by reducing myocardin activity. PLoS ONE 8, e63721 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Goettsch, C. et al. miR-125b regulates calcification of vascular smooth muscle cells. Am. J. Pathol. 179, 1594–1600 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. de Crombrugghe, B. et al. Transcriptional mechanisms of chondrocyte differentiation. Matrix Biol. 19, 389–394 (2000).

    Article  PubMed  Google Scholar 

  23. Gu, J. et al. Identification and characterization of the novel Col10a1 regulatory mechanism during chondrocyte hypertrophic differentiation. Cell Death Dis. 5, e1469 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Gong, Y. C. et al. Silencing of osterix expression by siRNA inhibits aldosteroneinduced calcification of vascular smooth muscle cells in mice. Mol. Med. Rep. 14, 2111–2118 (2016).

    Article  PubMed  CAS  Google Scholar 

  25. Lau, D. et al. The cartilage-specific lectin C-type lectin domain family 3 member A (CLEC3A) enhances tissue plasminogen activator-mediated plasminogen activation. J. Biol. Chem. 293, 203–214 (2018).

    Article  PubMed  CAS  Google Scholar 

  26. Beazley, K. E. et al. Transglutaminase inhibitors attenuate vascular calcification in a preclinical model. Arter. Thromb. Vasc. Biol. 33, 43–51 (2013).

    Article  CAS  Google Scholar 

  27. Hessle, L. et al. The skeletal phenotype of chondroadherin deficient mice. PLoS ONE 8, e63080 (2014).

    Article  PubMed  CAS  Google Scholar 

  28. Wu, M., Chen, G. & Li, Y. P. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Res. 4, 16009 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Gabbitas, B. & Canalis, E. Growth factor regulation of insulin-like growth factor binding protein-6 expression in osteoblasts. J. Cell. Biochem. 66, 77–86 (1997).

    Article  PubMed  CAS  Google Scholar 

  30. Ding, W. et al. miR-30e targets IGF2-regulated osteogenesis in bone marrow-derived mesenchymal stem cells, aortic smooth muscle cells, and ApoE-/- mice. Cardiovasc. Res. 106, 131–142 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Henein, M. et al. High dose and long-term statin therapy accelerate coronary artery calcification. Int. J. Cardiol. 184, 581–586 (2015).

    Article  PubMed  Google Scholar 

  32. Nakazato, R. et al. Statins use and coronary artery plaque composition: results from the International Multicenter CONFIRM Registry. Atherosclerosis 225, 148–153 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Braunersreuther, V. et al. A novel RANTES antagonist prevents progression of established atherosclerotic lesions in mice. Arter. Thromb. Vasc. Biol. 28, 1090–1096 (2008).

    Article  CAS  Google Scholar 

  34. Hiebert, P. R., Boivin, W. A., Zhao, H., McManus, B. M. & Granville, D. J. Perforin and granzyme B have separate and distinct roles during atherosclerotic plaque development in apolipoprotein E knockout mice. PLoS ONE 8, e78939 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Grandoch, M. et al. Deficiency in lymphotoxin β receptor protects from atherosclerosis in apoE-deficient mice. Circ. Res. 116, e57–e68 (2015).

    Article  PubMed  CAS  Google Scholar 

  36. Wessling-Resnick, M. Iron homeostasis and the inflammatory response. Ann. Rev. Nutr. 30, 105–122 (2010).

    Article  CAS  Google Scholar 

  37. Mo, Y. et al. Epithelial SERPINB10, a novel marker of airway eosinophilia in asthma, contributes to allergic airway inflammation. Am. J. Physiol. Lung Cell. Mol. Physiol. 316, L245–L254 (2019).

    Article  PubMed  CAS  Google Scholar 

  38. He, R. et al. IL-33 improves wound healing through enhanced M2 macrophage polarization in diabetic mice. Mol. Immunol. 90, 42–49 (2017).

    Article  PubMed  CAS  Google Scholar 

  39. Miller, A. M. et al. IL-33 reduces the development of atherosclerosis. J. Exp. Med. 205, 339–346 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. McLaren, J. E. et al. IL-33 reduces macrophage foam cell formation. J. Immunol. 185, 1222–1229 (2010).

    Article  PubMed  CAS  Google Scholar 

  41. Gundra, U. M. et al. Alternatively activated macrophages derived from monocytes and tissue macrophages are phenotypically and functionally distinct. Blood 123, e110–e122 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Cho, C. H. et al. Angiogenic role of LYVE-1-positive macrophages in adipose tissue. Circ. Res. 100, e47–e57 (2007).

    PubMed  CAS  Google Scholar 

  43. Tabas, I. & Lichtman, A. H. Monocyte-macrophages and T cells in atherosclerosis. Immunity 47, 621–634 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Sage, A. P., Tsiantoulas, D., Binder, C. J. & Mallat, Z. The role of B cells in atherosclerosis. Nat. Rev. Cardiol. 16, 180–196 (2019).

    Article  PubMed  CAS  Google Scholar 

  45. Tang, C. Y. et al. Runx1 up-regulates chondrocyte to osteoblast lineage commitment and promotes bone formation by enhancing both chondrogenesis and osteogenesis. Biochem. J. 477, 2421–2438 (2020).

    Article  PubMed  CAS  Google Scholar 

  46. Kimura, A. et al. Runx1 and Runx2 cooperate during sternal morphogenesis. Development 137, 1159–1167 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. von der Thusen, J. H. et al. IGF-1 has plaque-stabilizing effects in atherosclerosis by altering vascular smooth muscle cell phenotype. Am. J. Pathol. 178, 924–934 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Cheng, J. & Du, J. Mechanical stretch simulates proliferation of venous smooth muscle cells through activation of the insulin-like growth factor-1 receptor. Arter. Thromb. Vasc. Biol. 27, 1744–1751 (2007).

    Article  CAS  Google Scholar 

  49. Li, K., Wang, Y., Zhang, A., Liu, B. & Jia, L. miR-379 inhibits cell proliferation, invasion, and migration of vascular smooth muscle cells by targeting insulin-like factor-1. Yonsei Med. J. 58, 234–240 (2017).

    Article  PubMed  CAS  Google Scholar 

  50. Elzi, D. J. et al. Plasminogen activator inhibitor 1-insulin-like growth factor binding protein 3 cascade regulates stress-induced senescence. Proc. Natl Acad. Sci. USA 109, 12052–12057 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Kim, K. S. et al. Regulation of replicative senescence by insulin-like growth factor-binding protein 3 in human umbilical vein endothelial cells. Aging Cell 6, 535–545 (2007).

    Article  PubMed  CAS  Google Scholar 

  52. Ozcan, S. et al. Unbiased analysis of senescence associated secretory phenotype (SASP) to identify common components following different genotoxic stresses. Aging 8, 1316–1329 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Vassilieva, I. et al. Paracrine senescence of human endometrial mesenchymal stem cells: a role for the insulin-like growth factor binding protein 3. Aging 12, 1987–2004 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Sukhanov, S. et al. IGF-1 reduces inflammatory responses, suppresses oxidative stress, and decreases atherosclerosis progression in ApoE-deficient mice. Arter. Thromb. Vasc. Biol. 27, 2684–2690 (2007).

    Article  CAS  Google Scholar 

  55. Shai, S. Y. et al. Smooth muscle cell-specific insulin-like growth factor-1 overexpression in Apoe-/- mice does not alter atherosclerotic plaque burden but increases features of plaque stability. Arter. Thromb. Vasc. Biol. 30, 1916–1924 (2010).

    Article  CAS  Google Scholar 

  56. Zhang, C. et al. Regulation of vascular smooth muscle cell proliferation and migration by human sprouty 2. Arter. Thromb. Vasc. Biol. 25, 533–538 (2005).

    Article  CAS  Google Scholar 

  57. Yang, K. & Proweller, A. Vascular smooth muscle Notch signals regulate endothelial cell sensitivity to angiogenic stimulation. J. Biol. Chem. 286, 13741–13753 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Nicosia, R. F. The aortic ring model of angiogenesis: a quarter century of search and discovery. J. Cell. Mol. Med. 13, 4113–4136 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Lebedeva, A. et al. Ex vivo culture of human atherosclerotic plaques: a model to study immune cells in atherogenesis. Atherosclerosis 267, 90–98 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Higashi, Y., Gautam, S., Delafontaine, P. & Sukhanov, S. IGF-1 and cardiovascular disease. Growth Horm. IGF Res. 45, 6–16 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Basatemur, G. L., Jorgensen, H. F., Clarke, M. C. H., Bennett, M. R. & Mallat, Z. Vascular smooth muscle cells in atherosclerosis. Nat. Rev. Cardiol. 16, 727–744 (2019).

    Article  PubMed  Google Scholar 

  62. Jacobsen, K. et al. Diverse cellular architecture of atherosclerotic plaque derives from clonal expansion of a few medial SMCs. JCI Insight https://doi.org/10.1172/jci.insight.95890 (2017).

  63. Misra, A. et al. Integrin-β3 regulates clonality and fate of smooth muscle-derived atherosclerotic plaque cells. Nat. Commun. 9, 2073 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Chappell, J. et al. Extensive proliferation of a subset of differentiated, yet plastic, medial vascular smooth muscle cells contributes to neointimal formation in mouse injury and atherosclerosis models. Circ. Res. 119, 1313–1323 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Pan, H. et al. Single-cell genomics reveals a novel cell state during smooth muscle cell phenotypic switching and potential therapeutic targets for atherosclerosis in mouse and human. Circulation 142, 2060–2075 (2020).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  66. Alencar, G. F. et al. Stem cell pluripotency genes Klf4 and Oct4 regulate complex SMC phenotypic changes critical in late-stage atherosclerotic lesion pathogenesis. Circulation 142, 2045–2059 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Shankman, L. S. et al. KLF4-dependent phenotypic modulation of smooth muscle cells has a key role in atherosclerotic plaque pathogenesis. Nat. Med. 21, 628–637 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Newman, A. A. C. Multiple cell types contribute to the atherosclerotic lesion fibrous cap by PDGFRβ and bioenergetic mechanisms. Nat. Metab. 3, 166–181 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Baker, D. J. et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479, 232–236 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Gomez, D., Shankman, L. S., Nguyen, A. T. & Owens, G. K. Detection of histone modifications at specific gene loci in single cells in histological sections. Nat. Methods 10, 171–177 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank C. Conover for helpful discussions. This work was supported by the Glenn Foundation for Medical Research (J.M.v.D., D.J.B. and H.L.), the Keck Foundation (J.M.v.D.), National Institute of Aging grants AG57493 (J.M.v.D.) and AG049672 (B.G.C.) and the Mayo Clinic Kendall Fellowship (B.G.C).

Author information

Authors and Affiliations

Authors

Contributions

J.M.v.D. led the study. B.G.C. and J.M.v.D. designed experiments, interpreted data and wrote the manuscript. All authors contributed to manuscript writing and figure preparations. B.G.C., I.S., C.Z. and H.L. performed RNA-seq and bioinformatics data processing. F.S. collected and provided human endarterectomy specimens and helped design experiments on human explants. S.T. helped with assessments of proliferation and apoptosis rates. R.F.V. established mouse cohorts and helped administer senolytics. B.G.C. performed all other experiments with support of D.J.B.

Corresponding author

Correspondence to Jan M. van Deursen.

Ethics declarations

Competing interests

J.M.v.D. is a cofounder of Unity Biotechnology. J.M.v.D., D.J.B. and B.G.C. are inventors on Mayo Clinic patents licensed to Unity Biotechnology. J.M.v.D., D.J.B. H.L. and B.G.C. are current Unity Biotechnology shareholders. All other authors declare no competing interests.

Additional information

Peer review information Nature Aging thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 ABT263 depletes SNCs and reduces foam cell macrophage content in intermediate-stage lesions.

a, Representative SA β-Gal+ cells in H-E stained aortic arch plaques from indicated groups and quantified in b (left). b, (Left) Quantification of SA β-Gal+ cell counts in mice from indicated groups. (Right) En face quantification of SA β-Gal+ lesional surface area in mice from indicated groups. c, Representative foam cell macrophage pockets (black traced areas; quantified in d) in mice from 12-week HFD-fed Ldlr –/– mice and 12-week HFD-fed Ldlr –/– switched to LFD for 9 weeks with either 3x ABT263 or 3x Veh administration. d, Quantification of foam cell macrophage content per section (legend as in b). ‘n’ refers in all panels to number of mice. Statistics in panel b (left) and d were performed by ordinary one-way ANOVA with Holm-Sidak multiple comparison correction for the indicated comparisons. Panel b (right) was analyzed by unpaired, two-tailed t-test with Welch’s correction. Error bars represent s.e.m. Scale bars are 50 µm (a) and 100 µm (c).

Source data

Extended Data Fig. 2 Senolysis blocks fibrous cap thinning in the brachiocephalic artery.

a, Representative H-E staining of brachiocephalic artery sections from 12-week HFD-fed Ldlr –/– mice and 12-week HFD-fed Ldlr –/– switched to LFD for 9 weeks with either administration of 3 cycles ABT263 or 3 cycles Veh (3x Veh); see Fig. 1a for experimental design. Insets show a region of fibrous cap, outlined in black dashed lines and quantified in b. b, Quantification of fibrous cap thickness in brachiocephalic arteries from indicated groups. c, Quantification of average brachiocephalic plaque cross-sectional area per section in mice of indicated groups. ‘n’ refers in all panels to number of mice. All statistics were performed by ordinary one-way ANOVA with Holm-Sidak multiple comparison correction for the indicated comparisons. Error bars represent s.e.m. Scale bars for a are 400 µm (main) and 100 µm (inset).

Source data

Extended Data Fig. 3 ABT263 blocks fibrous cap thinning in aortic arch lesions by maintaining elastogenic VSMC numbers.

a, Immunohistochemical staining for Sma (quantified in b) in fibrous caps (dashed red lines) from 12-week HFD baseline mice, as well as those remodeling for 9 weeks on LFD with three 7-day cycles of ABT263 or Veh (3x ABT263 and 3x Veh; LFD weeks 1,4, and 7), or one 7-day cycle of ABT263 (1x ABT; LFD week 1). b, Quantification of Sma+ cells per µm of fibrous cap length from the indicated mice. c, Representative Verhoeff-Van Gieson staining of fibrous caps from the indicated groups and quantified in d. d, Quantification of Verhoeff-Van Gieson-positive fine fibers in fibrous caps of the indicated groups (legend as in b). e, Representative Vimentin and Sma costaining in fibrous caps of mice of the indicated groups (quantified in f). Yellow arrowheads indicated Vim/Sma+ fibrous cap cells. f, Quantification of the percentage of Sma+/Vim (left), Sma/Vim+ (middle), and Sma+/Vim+ (right) cells in fibrous caps from indicated groups (legend as in b). Arrowheads: Sma+/Vim fibrous cap cells. ‘n’ refers in all panels to number of mice. Statistics in panels b and d were performed by ordinary one-way ANOVA with Holm-Sidak multiple comparison correction for the indicated comparisons. Panel b includes data presented in Fig. 2f for context (12 w HFD baseline); here, ANOVA includes all indicated comparisons across Fig. 2f and Extended Data Fig. 3b. Panel f was analyzed by unpaired, two-tailed t-test with Welch’s correction. Error bars represent s.e.m.. Scale bar for a is 50 µm; c, 40 µm; and, e, 20 µm.

Source data

Extended Data Fig. 4 Transcriptome analysis indicates that ABT263 induces multiple favorable changes in plaque pathogenesis.

a, Heat map of differentially expressed genes upregulated (left) and downregulated (right) by ABT263 treatment in aortic arch plaque (see Fig. 3a for experimental schematic). b, Heat-map of pro-synthetic VSMC phenotype and muscle function genes upregulated with ABT263 treatment. c, Heat-map of anti-calcification changes in gene expression produced by ABT263 treatment among DEGs. d, Heat-map of pro-inflammatory and macrophage polarization DEGs modulated with ABT263 treatment in aortic arch plaque. e, Percentage of Sma+ cells showing TUNEL+ nuclei in fibrous caps (left) and total plaque (right) of indicated groups. Panel e (left and right) were assessed by unpaired, two-tailed Student’s t-test with Welch’s correction. Error bars represent s.e.m. ‘n’ in all cases represents number of individual mice.

Source data

Extended Data Fig. 5 Contractile to promigratory phenotype switching of medial VSMCs is restricted to IFS1 underneath lesions.

a, Representative images of IFS1-4 beneath plaque of the indicated groups stained for Sma and Vim (quantified in c). b, Representative images of plaque-flanking IFS1 VSMCs stained for Sma and Vim (quantified in c). c, VSMC expression profile of Vim and Sma in cells crossing the first elastic fiber, interfiber spaces (IFS) 1 to 3 beneath plaques, and IFS1 flanking plaques separated by treatment group (ABT or Vehicle administration during LFD feeding weeks 1, 4, and 7, following 24 weeks HFD feeding. *, p < 0.05; **, p < 0.01; ***, p < 0.001. Exact p-values are indicated in the raw data source file. d, VSMC expression profile of Vim and Sma in cells crossing the first elastic fiber versus interfiber space (IFS) 1 aggregated across treatment group (ABT or Vehicle administration during LFD feeding weeks 1, 4, and 7, following 12 weeks HFD feeding). Panels in c and d were analyzed by global x2, followed up by individual two-tailed Fischer’s exact tests. ‘n’ represents individual cells analyzed per aortic compartment. Scale bars in a and b are 20 µm.

Source data

Extended Data Fig. 6 Fibrous cap repair is characterized by enhanced migration of medial VSMCs.

a, (Left) Representative H/E-stained examples of medial cells crossing the first elastic lamina (blue arrowheads) from indicated groups. (Right) Quantification of medial cells crossing the first elastic lamina normalized to elastic fiber break length. b, Quantification of Sma/Vim+ cells in IFS1 of indicated groups (legend as in a). c, Quantification of Sma+/Vim cells in IFS1 of indicated groups (legend as in a). d, (Left) Representative H/E-stained examples of medial cells crossing the first elastic lamina (blue arrowheads) from indicated groups. (Right) Quantification of medial cells crossing the first elastic lamina normalized to elastic fiber break length. e, Quantification of Sma/Vim+ cells in IFS1 of indicated groups (legend as in d). f, Quantification of Sma+/Vim cells in IFS1 of indicated groups (legend as in a). Statistics in all panels were performed by unpaired, two-tailed t-test with Welch’s correction. Error bars represent s.e.m. ‘n’ in all cases refers to number of individual mice. Scale bars in a and d are 60 µm.

Source data

Extended Data Fig. 7 Impact on senolysis on proliferation, viability and osteogenic transdifferentiation of media-derived VSMCs in lesions.

a, Images of early-stage aortic arch lesions after various lengths of HFD feeding (14 and 21 day images were from Ldlr–/– females and 33 day images from Myh11-CreERT2;CAG-LSL-tdTomato-WPRE Ldlr–/– males). Note that 33-day streaks (n = 7 mice +Veh/ n = 8 mice +ABT) cover a broad spectrum of lesion sizes, including the small-sized foci developing in streaks of 14 (n = 4 mice) and 21 day (n = 4 mice) HFD-fed mice. b, Quantification of EdU+ percentage of tdTomato+ in fibrous caps and plaque cores for mice of the indicated groups. c, Percentage of Tom+/Sma cells with EdU among plaque fibrous cap and core cells, with or without ABT263 treatment. d, Percentage of Tom+/Sma+ cells with EdU among plaque fibrous cap and core cells, with or without ABT263 treatment. e, Quantification of TUNEL+ percentage of tdTomato+ cells in fibrous caps and plaque cores for mice of indicated groups. f, Percentage of Tom+/Sma cells with TUNEL among plaque fibrous cap and core cells, with or without ABT263 treatment. g, Percentage of Tom+/Sma+ cells with TUNEL among plaque fibrous cap and core cells, with or without ABT263 treatment. h, Representative images of Myh11/Sma+ cells among all tdTomato+ IFS1 and IFS2 cells (quantified in i). i, Quantification of indicated groups from h. j, Representative images (left) and quantification (right) of Runx1 and tdTomato colocalization in mice of indicated groups. Orange dashed line indicates the first elastic fiber. Panels b-g and j (right) were analyzed by unpaired, two-tailed t-tests with Welch’s correction. Panel h was analyzed by ordinary one-way ANOVA with Holm-Sidak multiple comparison correction for the indicated comparisons. ‘n’ in all cases refers to number of mice. Error bars represent s.e.m. Scale bars are 0.5 mm in a and 20 µm in h and j.

Source data

Extended Data Fig. 8 ABT263 accelerates formation of a fibrous cap structure by stimulating migration of medial VSMC.

a, Schematic of experiments designed to study the impact of ABT263-mediated senolysis on fibrous cap formation in early inner aortic arch lesions of Ldlr–/ mice after HFD-to-LFD switching. b, Total plaque burden as measured by en face scoring in mice from indicated groups. c, Neointimal cross sectional area as measured by TEM in lesions of indicated mice (legend is as in b). d, Macrophages per neointimal area in mice of indicated groups (legend is as in b). e, Representative image illustrating VSMCs traversing the first elastic lamina (red masks) in a lesion of the indicated Ldlr–/ mouse (quantified in f). f, Quantification of VSMCs crossing the first elastic lamina in mice of indicated groups (legend is as in b). g, VSMCs per µm of neointima in mice of indicated groups (legend is as in b). h, Percentage of elastin-producing VSMCs in mice from indicated groups (legend is as in b). i, (Left) Representative electron micrographs of 4-week HFD simple foam cell lesions and lesions remodeling during 6-week LFD feeding with Veh or ABT263 administration (quantified in right panel). EC, Endothelial cell. (Right) Quantification of fibrous cap thickness (legend is as in b). ‘n’ refers in all panels to number of mice. All analyses were performed by ordinary one-way ANOVA with Holm-Sidak multiple comparison correction for the indicated comparisons. Error bars represent s.e.m. Scale bars in e and i are 5 µm.

Source data

Extended Data Fig. 9 Senescent cell-derived IGFBP3 inhibits IGF1-mediated promigratory phenotype switching of VSMCs.

a, Expression of Igfbp3 in the indicated MEFs. b, (Left) Representative images of VSMC outgrowing from aortic rings (AR) of wildtype C57BL/6 mice treated with the indicated conditioned media (CM). (Right) Quantification of outgrowing VSMCs in the indicated treatment groups. c, (Left) Representative images of human aortic VSMCs emigrating into scratch wound space with indicated conditioned media (CM). Red dashed lines indicate cell monolayer/scratch wound boundary. (Right) Quantification of emigrating VSMCs in the indicated experimental groups. LR3 IGF-1, long R3 mutant stabilized recombinant IGF-1. d, Immunofluorescent staining of Vim and Sma in human aortic VSMC with the indicated treatments. CM, conditioned media (quantified in e). e, Quantification of average fluorescent signal intensity of SMA and VIM per cell in human aortic VSMC receiving indicated treatments. f, (Top) Representative images of SA β-Gal stained Ldlr–/– aortic arches from mice fed HFD for 33 d with concurrent administration of either LR3 IGF1 or vehicle control. (Bottom left) Quantification of total plaque burden in the aortic arch and (bottom right) percentage of aortic arch plaque with SA β-Gal positivity in indicated treatment groups. ‘n’ refers individual MEF lines (panel a); individual aortic rings in b; individual mice in panel f; and, individual lines of MEF conditioned media (panels c and e). Panel a was analysed with unpaired, two-tailed t-test. Panel f was analyzed by unpaired, two-tailed t-test with Welch’s correction. Analyses in panels b and e were performed by ordinary one-way ANOVA with Holm-Sidak multiple comparison correction for the indicated comparisons, and analysis of panel c was performed by RM one-way ANOVA with Holm-Sidak multiple comparison correction for the indicated comparisons. Error bars represent s.e.m. Scale bar in b is 500 µm; c, 200 µm; d, 100 µm; and, f, 1 mm.

Source data

Extended Data Fig. 10 Proposed model for how SNCs suppress the cap repair functions of VSMCs in advanced atherosclerotic lesions.

(Top) SNCs inhibit lesional IGF signaling by elevating Igfbp3 levels, thereby suppressing promigratory phenotype switching of contractile VSMCs in the media and their recruitment to the fibrous cap, as well as ECM deposition by VSMCs in the cap, resulting in fibrous cap erosion. (Bottom) SNC clearance reduces lesional Igfbp3 levels, thereby stimulating IGF-mediated promigratory phenotype switching of medial VSMC and their recruitment to the fibrous cap, as well as ECM deposition by VSMCs in the cap, restoring fibrous cap thickness and plaque stability.

Supplementary information

Supplementary Information

Supplementary Figs. 1–4.

Reporting Summary

Supplementary Data 1

Source file for Supplementary Figs. 1–4.

Supplementary Tables

Supplementary Tables 1–3.

Source data

Source Data Fig. 1

Statistical source file for Fig. 1.

Source Data Fig. 2

Statistical source file for Fig. 2.

Source Data Fig. 3

Statistical source file for Fig. 3.

Source Data Fig. 4

Statistical source file for Fig. 4.

Source Data Fig. 5

Statistical source file for Fig. 5.

Source Data Fig. 6

Statistical source file for Fig. 6.

Source Data Fig. 7

Statistical source file for Fig. 7.

Source Data Fig. 8

Statistical source file for Fig. 8.

Source Data Extended Data Fig. 1

Statistical source file for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Statistical source file for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical source file for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical source file for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Statistical source file for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical source file for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Statistical source file for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Statistical source file for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Statistical source file for Extended Data Fig. 9.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Childs, B.G., Zhang, C., Shuja, F. et al. Senescent cells suppress innate smooth muscle cell repair functions in atherosclerosis. Nat Aging 1, 698–714 (2021). https://doi.org/10.1038/s43587-021-00089-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00089-5

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