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:

TP53-mediated clonal hematopoiesis confers increased risk for incident atherosclerotic disease

Abstract

Somatic mutations in blood indicative of clonal hematopoiesis of indeterminate potential (CHIP) are associated with an increased risk of hematologic malignancy, coronary artery disease and all-cause mortality. Here we analyze the relation between CHIP status and incident peripheral artery disease (PAD) and atherosclerosis, using whole-exome sequencing and clinical data from the UK Biobank and the Mass General Brigham Biobank. CHIP associated with incident PAD and atherosclerotic disease across multiple beds, with increased risk among individuals with CHIP driven by mutation in DNA damage repair (DDR) genes, such as TP53 and PPM1D. To model the effects of DDR-induced CHIP on atherosclerosis, we used a competitive bone marrow transplantation strategy and generated atherosclerosis-prone Ldlr−/− chimeric mice carrying 20% p53-deficient hematopoietic cells. The chimeric mice were analyzed 13 weeks after grafting and showed increased aortic plaque size and accumulation of macrophages within the plaque, driven by increased proliferation of p53-deficient plaque macrophages. In summary, our findings highlight the role of CHIP as a broad driver of atherosclerosis across the entire arterial system beyond the coronary arteries and provide genetic and experimental support for a direct causal contribution of TP53-mutant CHIP to 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: CHIP and incident PAD risk.
Fig. 2: CHIP and incident pan-arterial atherosclerosis risk.
Fig. 3: Gene-specific association of CHIP with incident PAD.
Fig. 4: Accelerated atherosclerosis in a murine model of TP53 mutation-driven CHIP.
Fig. 5: Expansion of p53-deficient macrophages within atherosclerotic plaques.
Fig. 6: Increased proliferation of p53-deficient macrophages.

Similar content being viewed by others

Data availability

UKB individual-level data are available by request via application (https://www.ukbiobank.ac.uk). Individual-level MGBB data are available from https://personalizedmedicine.partners.org/Biobank/Default.aspx, only to Partners HealthCare investigators with appropriate approval from the Partners institutional review board. RNA-seq data are available at the Gene Expression Omnibus (GSE184420). The present article includes all other data generated or analyzed during this study.

Code availability

Our CHIP-calling Terra pipeline using Mutect2 (version 1) is available at https://app.terra.bio/#workspaces/terra-outreach/CHIP-Detection-Mutect2. R-scripts for the observational epidemiologic associations are available at https://github.com/mzekavat/CHIP_PAD.

References

  1. Song, P. et al. Global, regional, and national prevalence and risk factors for peripheral artery disease in 2015: an updated systematic review and analysis. Lancet Glob. Health 7, e1020–e1030 (2019).

    Article  PubMed  Google Scholar 

  2. Conte, M. S. et al. Global vascular guidelines on the management of chronic limb-threatening ischemia. J. Vasc. Surg. 69, 3S–125S (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371, 2488–2498 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Jaiswal, S. et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N. Engl. J. Med. 377, 111–121 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Xie, M. et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 20, 1472–1478 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 371, 2477–2487 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Bick, A. G. et al. Genetic interleukin 6 signaling deficiency attenuates cardiovascular risk in clonal hematopoiesis. Circulation 141, 124–131 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Pascual-Figal, D. A. et al. Clonal hematopoiesis and risk of progression of heart failure with reduced left ventricular ejection fraction. J. Am. Coll. Cardiol. 77, 1747–1759 (2021).

    Article  PubMed  Google Scholar 

  9. Yu, B. et al. Supplemental association of clonal hematopoiesis with incident heart failure. J. Am. Coll. Cardiol. 78, 42–52 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bhattacharya, R. et al. Clonal hematopoiesis is associated with higher risk of stroke. Stroke 53, 788–797 (2022).

    Article  CAS  PubMed  Google Scholar 

  11. Bick, A. G. et al. Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature 586, 763–768 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Klarin, D. et al. Genome-wide association study of peripheral artery disease in the Million Veteran Program. Nat. Med. 25, 1274–1279 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Denny, J. C. et al. Systematic comparison of phenome-wide association study of electronic medical record data and genome-wide association study data. Nat. Biotechnol. 31, 1102–1110 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Fuster, J. J. et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science 355, 842–847 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Visconte, V., M, O. N. & H, J. R. Mutations in splicing factor genes in myeloid malignancies: significance and impact on clinical features. Cancers (Basel) 11, 1844 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Boettcher, S. et al. A dominant-negative effect drives selection of TP53 missense mutations in myeloid malignancies. Science 365, 599–604 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bondar, T. & Medzhitov, R. p53-mediated hematopoietic stem and progenitor cell competition. Cell Stem Cell 6, 309–322 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Liu, Y. et al. p53 regulates hematopoietic stem cell quiescence. Cell Stem Cell 4, 37–48 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. TeKippe, M., Harrison, D. E. & Chen, J. Expansion of hematopoietic stem cell phenotype and activity in Trp53-null mice. Exp. Hematol. 31, 521–527 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Sano, S. et al. Tet2-mediated clonal hematopoiesis accelerates heart failure through a mechanism involving the IL-1β/NLRP3 inflammasome. J. Am. Coll. Cardiol. 71, 875–886 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fuster, J. J. et al. TET2-loss-of-function-driven clonal hematopoiesis exacerbates experimental insulin resistance in aging and obesity. Cell Rep. 33, 108326 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Abplanalp, W. T. et al. Clonal hematopoiesis–driver DNMT3A mutations alter immune cells in heart failure. Circ. Res. 128, 216–228 (2021).

    Article  CAS  PubMed  Google Scholar 

  23. Sinha, S. K. et al. Local M-CSF (macrophage colony-stimulating factor) expression regulates macrophage proliferation and apoptosis in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 41, 220–233 (2021).

    CAS  PubMed  Google Scholar 

  24. Fuster, J. J. Clonal hematopoiesis and cardiovascular disease in cancer patients and survivors. Thromb. Res. 213, S107–S112 (2022).

    Article  CAS  PubMed  Google Scholar 

  25. Boesten, L. S. et al. Macrophage p53 controls macrophage death in atherosclerotic lesions of apolipoprotein E deficient mice. Atherosclerosis 207, 399–404 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. Fuster, J. J. et al. Control of cell proliferation in atherosclerosis: insights from animal models and human studies. Cardiovasc. Res. 86, 254–264 (2010).

    Article  CAS  PubMed  Google Scholar 

  27. Guevara, N. V., Kim, H. S., Antonova, E. I. & Chan, L. The absence of p53 accelerates atherosclerosis by increasing cell proliferation in vivo. Nat. Med. 5, 335–339 (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Mercer, J., Figg, N., Stoneman, V., Braganza, D. & Bennett, M. R. Endogenous p53 protects vascular smooth muscle cells from apoptosis and reduces atherosclerosis in ApoE knockout mice. Circ. Res. 96, 667–674 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Merched, A. J., Williams, E. & Chan, L. Macrophage-specific p53 expression plays a crucial role in atherosclerosis development and plaque remodeling. Arterioscler. Thromb. Vasc. Biol. 23, 1608–1614 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. Sanz-Gonzalez, S. M. et al. Increased p53 gene dosage reduces neointimal thickening induced by mechanical injury but has no effect on native atherosclerosis. Cardiovasc. Res. 75, 803–812 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. van Vlijmen, B. J. et al. Macrophage p53 deficiency leads to enhanced atherosclerosis in APOE*3-Leiden transgenic mice. Circ. Res. 88, 780–786 (2001).

    Article  PubMed  Google Scholar 

  32. Robbins, C. S. et al. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat. Med. 19, 1166–1172 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Min, K. D., Polizio, A. H., Kour, A., Thel, M. C. & Walsh, K. Experimental ASXL1-mediated clonal hematopoiesis promotes inflammation and accelerates heart failure. J. Am. Heart Assoc. 11, e026154 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Heyde, A. et al. Increased stem cell proliferation in atherosclerosis accelerates clonal hematopoiesis. Cell 184, 1348–1361 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Fidler, T. P. et al. The AIM2 inflammasome exacerbates atherosclerosis in clonal haematopoiesis. Nature 592, 296–301 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bycroft, C. et al. The UK Biobank resource with deep phenotyping and genomic data. Nature 562, 203–209 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Smoller, J. W. et al. An eMERGE clinical center at Partners Personalized Medicine. J. Pers. Med. 6, 5 (2016).

  38. Van Hout, C. V. et al. Exome sequencing and characterization of 49,960 individuals in the UK Biobank. Nature 586, 749–756 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Wu, P. et al. Mapping ICD-10 and ICD-10-CM codes to phecodes: workflow development and initial evaluation. JMIR Med. Inform. 7, e14325 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Zekavat, S. M. et al. Hematopoietic mosaic chromosomal alterations increase the risk for diverse types of infection. Nat. Med. 27, 1012–1024 (2021).

  41. Hernan, M. A., Brumback, B. & Robins, J. M. Marginal structural models to estimate the causal effect of zidovudine on the survival of HIV-positive men. Epidemiology 11, 561–570 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Walter, W., Sánchez-Cabo, F. & Ricote, M. GOplot: an R package for visually combining expression data with functional analysis. Bioinformatics 31, 2912–2914 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Adrover, J. M. et al. Programmed ‘disarming’ of the neutrophil proteome reduces the magnitude of inflammation. Nat. Immunol. 21, 135–144 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

D.K. is supported by the Veterans Administration (award 1IK2BX005759-01). P.N. is supported by a Hassenfeld Scholar Award from Massachusetts General Hospital; grants from the National Institutes of Health (NIH) National Heart, Lung, and Blood Institute (NHLBI) (R01HL1427, R01HL148565 and R01HL148050); and a grant from Fondation Leducq (TNE-18CVD04). S.M.Z is supported by the NIH NHLBI (1F30HL149180-01) and the NIH Medical Scientist Training Program Training Grant (T32GM136651). J.P.P. is supported by a John S. LaDue Memorial Fellowship. K.P. is supported by NIH grant 5-T32HL007208-43. P.L. receives funding support from the NHLBI (1R01HL134892), the American Heart Association (18CSA34080399), the RRM Charitable Fund and the Simard Fund. J.J.F. is supported by grant RYC-2016-20026 from the Spanish ‘Ministerio de Ciencia e Innovación’ (MICIN/AEI /10.13039/501100011033’ and ‘ESF Investing in your future’); a 2019 Leonardo Grant for Researchers and Cultural Creators from the BBVA Foundation; the European Research Area Network on Cardiovascular Diseases CHEMICAL (grant AC19/00133, funded by Instituto de Salud Carlos III and co-funded by the European Union, ERDF, ‘A way to make Europe’); and the Leducq Foundation (TNE-18CVD04). A.G.B. is supported by NIH grant DP5 OD029586, a Burroughs Wellcome Fund Career Award for Medical Scientists and a Pew-Stewart Scholar for Cancer Research award, supported by the Pew Charitable Trusts and the Alexander and Margaret Stewart Trust. C.G. is supported by the European Research Area Network on Cardiovascular Diseases CHEMICAL. A.H. is supported by the Leducq Foundation (TNE-18CVD04). C.M.B. is supported by NIH grant R01-HL148050. C.J.G. is supported by NIH/NCI K08CA263555. P.L. receives funding support from the NHLBI (1R01HL134892 and 1R01HL163099-01), the American Heart Association (18CSA34080399), the RRM Charitable Fund and the Simard Fund. M.A.-P. is supported by grant FPU18/02913 funded by MCIN/AEI /10.13039/501100011033 and by ‘ESF Investing in your future’. The project leading to these results also received funding from ‘la Caixa’ Foundation (ID 100010434), including fellowship code LCF/BQ/DR19/11740022 (to A.A.-C.) and agreement HR17-00267. Confocal microscopy was conducted at the Microscopy & Dynamic Imaging Unit at CNIC, ICTS-ReDib, co-funded by MCIN/AEI /10.13039/501100011033 and FEDER ‘Una manera de hacer Europa’ (ICTS-2018-04-CNIC-16). The CNIC is supported by the Instituto de Salud Carlos III (ISCIII), the MICIN and the Pro CNIC Foundation and is a Severo Ochoa Center of Excellence (grant CEX2020-001041-S funded by MICIN/AEI/10.13039/501100011033). We thank R. Moro for assistance with figure preparation and F. Sánchez-Cabo and M. Gómez for assistance with bioinformatics analysis of RNA-seq data. S.M.D. is supported by the Veterans Administration (award IK2-CX001780). We also thank participants and staff of the UK Biobank and the Mass General Brigham Biobank. UK Biobank analyses were conducted using application 7089.

Author information

Authors and Affiliations

Authors

Contributions

S.M.Z., S.D.J., M.M.U., M.T., K.P., S.D., J.P., G.G. and A.N. designed, performed and analyzed population-level data from the UKB and the MGBB and interpreted data. S.M.Z. and D.K. interpreted data, created the figures and assisted in writing the respective parts of the paper. N.M. designed, performed and analyzed experiments; interpreted experimental data; created the figures; and assisted in writing the manuscript. V.V.-H. designed, performed and analyzed experiments; interpreted experimental data; and contributed to figure preparation. J.J.F., M.A.Z., M.A.-P. and A.A.-C. performed and analyzed experiments and interpreted experimental data. V.Z. and A.F.-P. provided technical support and performed experiments. P.K., R.C. and C.M.G. conducted RNA-seq studies and analyzed transcriptomics data. A.H., V.F., B.L.E., P.L., C.J.G., C.M.B., H.Z., M.G. and D.K. discussed the research strategy and the results and provided conceptual advice. P.N., A.G.B., J.J.F. and D.K. conceived the study, analyzed and interpreted data and created the figures.

Corresponding authors

Correspondence to José J. Fuster or Derek Klarin.

Ethics declarations

Competing interests

D.K. is a scientific advisor for and accepts consulting fees from Bitterroot Bio. P.N. reports grant support from Amgen, Apple, AstraZeneca, Boston Scientific and Novartis; spousal employment and equity at Vertex; consulting income from Apple, AstraZeneca, Novartis, Genentech/Roche, Blackstone Life Sciences, Foresite Labs and TenSixteen Bio; and is a scientific advisory board member and shareholder of TenSixteen Bio and geneXwell, all unrelated to this work. P.L. is an unpaid consultant to, or involved in clinical trials for, Amgen, AstraZeneca, Baim Institute, Beren Therapeutics, Esperion Therapeutics, Genentech, Kancera, Kowa Pharmaceuticals, Medimmune, Merck, Novo Nordisk, Novartis, Pfizer and Sanofi-Regeneron. P.L. is a member of the scientific advisory board for Amgen, Caristo Diagnostics, Cartesian Therapeutics, CSL Behring, DalCor Pharmaceuticals, Dewpoint Therapeutics, Eulicid Bioimaging, Kancera, Kowa Pharmaceuticals, Olatec Therapeutics, Medimmune, Moderna, Novartis, PlaqueTec, TenSixteen Bio, Soley Therapeutics, and XBiotech. P.L.ʼs laboratory has received research funding in the last 2 years from Novartis. P.L. is on the board of directors of XBiotech. P.L. also has a financial interest in Xbiotech, a company developing therapeutic human antibodies; in TenSixteen Bio, a company targeting somatic mosaicism and CHIP to discover and develop novel therapeutics to treat age-related diseases; and in Soley Therapeutics, a biotechnology company that is combining artificial intelligence with molecular and cellular response detection for discovering and developing new drugs, currently focusing on cancer therapeutics. P.L.ʼs interests were reviewed and are managed by Brigham and Women’s Hospital and Mass General Brigham in accordance with their conflict of interest policies. B.L.E. has received research funding from Celgene, Deerfield, Novartis and Calico and consulting fees from GRAIL. B.L.E. is a member of the scientific advisory board and a shareholder for Neomorph, TenSixteen Bio, Skyhawk Therapeutics and Exo Therapeutics. A.G.B. is a paid advisor and holds equity in TenSixteen Bio. S.M.D. has received research support via the University of Pennsylvania from RenalytixAI and Novo Nordisk. The other authors do not have any competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks Alan Tall, Andrew Murphy, Gerard Pasterkamp and Steffen Massberg 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 Association of CHIP with blood counts among individuals without prevalent hematologic malignancy in the UK Biobank (n = 37,657).

Blood counts were acquired at time of blood draw for whole exome sequencing. a) Association of CHIP and Large CHIP with normalized blood counts (SD). Associations are adjusted for age, age2, sex, smoking status, and the first ten principal components of genetic ancestry. b) Association of CHIP variant allele frequency (VAF) with blood counts (in units of 10^9 cells/L). The gray horizontal dotted lines reflect average counts across non-CHIP carriers. The vertical black dotted line reflects the cutoff VAF for Large CHIP (VAF > 0.1). Error bands reflect the standard error of a generalized additive model with integrated smoothness fit to the data. CHIP = clonal hematopoiesis of indeterminate potential; VAF = variant allele fraction.

Extended Data Fig. 2 Association of CHIP (a) and VAF (b) with incident hematologic malignancy among individuals without prevalent hematological malignancy in the UK Biobank (n = 37,657).

Associations are adjusted for age, age2, sex, smoking status, Townsend deprivation index, and the first ten principal components of genetic ancestry. (a) Error bars are centered at the HR and show the 95% CI for estimates. (b) Error bands reflect the standard error of a generalized binomial additive model with integrated smoothness fit to the data. CHIP = clonal hematopoiesis of indeterminate potential; VAF = variant allele fraction.

Extended Data Fig. 3 Epidemiological causal inference analysis for CHIP on incident peripheral artery disease in the UK Biobank.

a) Propensity scores by CHIP and Large CHIP status in the UKB (n = 37,657). b) Propensity score adjustment and stabilized inverse probability treatment weighting (IPTW) for the CHIP and Large CHIP association with incident PAD in the UKB. Error bars are centered at the HR and show the 95% CI for estimates. CHIP = clonal hematopoiesis of indeterminate potential; VAF = variant allele fraction; PAD = peripheral artery disease.

Extended Data Fig. 4 Association of Large CHIP (VAF > 10%) with incident pan-arterial atherosclerosis, combined across peripheral artery disease, coronary artery disease, aneurysms, chronic and acute mesenteric ischemia, cerebral atherosclerosis, and renal artery stenosis.

Error bars are centered at the HR and show the 95% CI for estimates. CHIP = clonal hematopoiesis of indeterminate potential; VAF = variant allele fraction.

Extended Data Fig. 5 Association of a) CHIP and b) Large CHIP genes with incident pan-arterial atherosclerosis, combined across peripheral artery disease, coronary artery disease, aneurysms, chronic and acute mesenteric ischemia, cerebral atherosclerosis, and renal artery stenosis.

Error bars are centered at the HR and show the 95% CI for estimates. CHIP = clonal hematopoiesis of indeterminate potential; VAF = variant allele fraction.

Extended Data Fig. 6 Effects of experimental p53-deficient CHIP on atherosclerosis development in Ldlr-/- mice.

a-e) 20% KO-BMT male mice (n = 19 mice) and 20% WT-BMT controls (n = 20 mice) were fed a high-fat/high-cholesterol (HF/HC) diet for 9 weeks, starting 4 weeks after BMT. a) Percentage of CD45.2+ cells in different white blood cell (WBC) lineages in peripheral blood, evaluated by flow cytometry. Two-tailed unpaired t-tests were used for statistical analysis (mean ± SEM, ****p < 0.0001). b) Absolute counts of main WBC sub-populations in peripheral blood, evaluated by flow cytometry (mean ± SEM). c) Body weight (mean ± SEM). d) Spleen weight (mean ± SEM). e) Total cholesterol level in serum, evaluated by enzymatic methods (mean ± SEM). f-k) 20% KO-BMT female mice and 20% WT-BMT controls were fed a high-fat/high-cholesterol (HF/HC) diet for 9 weeks, starting 4 weeks after BMT. f) Percentage of CD45.2+ cells in white blood cells at different timepoints, evaluated by flow cytometry. A two-way ANOVA with Sidak’s multiple comparison test was used for statistical analysis (mean ± SEM,****p < 0.0001). g) Percentage of CD45.2+ cells in different WBC lineages in peripheral blood after 9 weeks on HF/HC diet (13 weeks post-BMT), evaluated by flow cytometry. Two-tailed unpaired t-tests were used for statistical analysis (mean ± SEM, n = 16 20% WT-BMT mice, n = 13 20% KO-BMT mice, ****p < 0.0001). h) Body weight (mean ± SEM, n = 17 20% WT-BMT mice, n = 13 20% KO-BMT mice). i) Spleen weight (mean ± SEM, n = 17 20% WT-BMT mice, n = 13 20% KO-BMT mice). j) Total cholesterol level in serum, evaluated by enzymatic methods (mean ± SEM, n = 17 20% WT-BMT mice, n = 12 20% KO-BMT mice) k) Aortic root plaque size. A two-tailed unpaired t-test was used for statistical analysis (mean ± SEM, n = 17 20% WT-BMT mice, n = 13 20% KO-BMT mice). Representative images of hematoxylin and eosin-stained sections are shown; atherosclerotic plaques are delineated by dashed lines. Scale bars, 100 μm.

Extended Data Fig. 7 No effect of p53-deficient CHIP on plaque lipid content or aortic expression of the proinflammatory cytokines IL-1b and IL-6 or the NLRP3 inflammasome.

a) Aortic arch samples were obtained from HF/HC-fed 20% KO-BMT male mice and 20% WT-BMT controls, and gene expression was analyzed by qPCR (mean ± SEM, n = 11 20% WT-BMT mice, n = 10 20% KO-BMT mice). b) 20% KO-BMT male mice and 20% WT-BMT controls were fed a high-fat/high-cholesterol (HF/HC) diet for 9 weeks. Plaque lipid content was analyzed through Oil Red O (ORO) staining of cryostat sections (mean ± SEM, n = 12 20% WT-BMT mice, n = 15 20% KO-BMT mice). Representative images of ORO-stained sections are shown; atherosclerotic plaques are delineated by dashed lines. Scale bars, 200 μm.

Extended Data Fig. 8 Increased cell proliferation in conditions of p53-deficient CHIP.

a) 20% KO-BMT female mice and 20% WT-BMT controls were fed a high-fat/high-cholesterol (HF/HC) diet for 9 weeks, starting 4 weeks after BMT. Plaque cell proliferation was estimated based on immunohistochemical staining of the Ki-67 proliferation marker (mean ± SEM, n = 17 20% WT-BMT mice, n = 14 20% KO-BMT mice). A two-tailed unpaired t-test was used for statistical analysis. Representative images of Ki-67-stained sections are shown; color deconvolution was applied to show separately the staining of hematoxylin (nuclei) and Ki-67. Atherosclerotic plaques are delineated by dashed lines. Scale bars, 50 μm. b) Cell cycle phase distribution of cultured Trp53-/- and + /+ bone marrow-derived macrophages proliferating asynchronously in the presence of 100 ng/ml MCSF, evaluated by propidium iodide staining of cellular DNA content and flow cytometry (mean ± SEM, n = 6 Trp53 + /+ mice, n = 6 Trp53-/- mice). A two-way ANOVA with Sidak’s multiple comparison test was used for statistical analysis. c) Analysis of the proliferation of cultured Trp53-/- and + /+ bone marrow-derived macrophages through immunostaining of BrdU incorporation into the DNA. Quiescent G0-synchronized macrophages were treated with MCSF to induce proliferation (mean ± SEM, n = 3 Trp53 + /+ mice, n = 3 Trp53-/- mice). A two-tailed unpaired t-test was used for statistical analysis. A representative experiment is shown; three separate experiments were conducted. Scale bars, 100 μm.

Extended Data Fig. 9 Transcriptomic profiling of MCSF-stimulated p53-deficient macrophages.

An 18h-treatment with MCSF was used to induce cell cycle entry and progression in quiescent G0-synchronized Trp53-/- (KO) and + /+ (WT) murine macrophages in culture (n = 3 per genotype). mRNA sequencing was used for transcriptomic profiling. a) Heatmap of differentially expressed genes with fold change (FC) ≥ 1.5. b) Functional categories enriched in the set of genes differentially expressed between Trp53-/- and + /+ macrophages, based on Ingenuity Pathway Analysis (cancer-related pathways are not shown). c) Heatmap of the most upregulated genes within selected functional categories that are enriched in genes differentially expressed in Trp53-/- macrophages with fold change ≥ 1.5. d) GoPlot of selected functional categories (right hand-side) and the logFC values of the most differentially expressed genes included in these categories (left hand-side).

Extended Data Fig. 10 Association of CHIP and Large CHIP (variant allele fraction > 10%) with PAD in the UKB (N = 37,657).

under 1) unadjusted, 2) sparsely adjusted, and 3) fully adjusted models, where sparsely adjusted refers to the following covariates: age, age2, sex, smoking status, Townsend deprivation index, and the first ten principal components of genetic ancestry, and the fully adjusted model additionally includes normalized BMI, prevalent hypertension, hyperlipidemia, and type 2 diabetes as covariates. Given the minimal difference between the sparsely adjusted and fully adjusted model, the sparsely adjusted model was moved forward for use in analysis. Error bars are centered at the HR and show the 95% CI for estimates.

Supplementary information

Supplementary Information

Supplementary Figs. 1–6 and Supplementary Tables 1–6, 10 and 11

Reporting Summary

Supplementary Tables 7–9

Source data

Source Data Fig. 6

Western blot for Fig. 6d

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

Zekavat, S.M., Viana-Huete, V., Matesanz, N. et al. TP53-mediated clonal hematopoiesis confers increased risk for incident atherosclerotic disease. Nat Cardiovasc Res 2, 144–158 (2023). https://doi.org/10.1038/s44161-022-00206-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s44161-022-00206-6

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