Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The AIM2 inflammasome exacerbates atherosclerosis in clonal haematopoiesis

Abstract

Clonal haematopoiesis, which is highly prevalent in older individuals, arises from somatic mutations that endow a proliferative advantage to haematopoietic cells. Clonal haematopoiesis increases the risk of myocardial infarction and stroke independently of traditional risk factors1. Among the common genetic variants that give rise to clonal haematopoiesis, the JAK2V617F (JAK2VF) mutation, which increases JAK–STAT signalling, occurs at a younger age and imparts the strongest risk of premature coronary heart disease1,2. Here we show increased proliferation of macrophages and prominent formation of necrotic cores in atherosclerotic lesions in mice that express Jak2VF selectively in macrophages, and in chimeric mice that model clonal haematopoiesis. Deletion of the essential inflammasome components caspase 1 and 11, or of the pyroptosis executioner gasdermin D, reversed these adverse changes. Jak2VF lesions showed increased expression of AIM2, oxidative DNA damage and DNA replication stress, and Aim2 deficiency reduced atherosclerosis. Single-cell RNA sequencing analysis of Jak2VF lesions revealed a landscape that was enriched for inflammatory myeloid cells, which were suppressed by deletion of Gsdmd. Inhibition of the inflammasome product interleukin-1β reduced macrophage proliferation and necrotic formation while increasing the thickness of fibrous caps, indicating that it stabilized plaques. Our findings suggest that increased proliferation and glycolytic metabolism in Jak2VF macrophages lead to DNA replication stress and activation of the AIM2 inflammasome, thereby aggravating atherosclerosis. Precise application of therapies that target interleukin-1β or specific inflammasomes according to clonal haematopoiesis status could substantially reduce cardiovascular risk.

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: Increased atherosclerosis in mice with macrophage Jak2VF expression and Jak2VF clonal haematopoiesis.
Fig. 2: Inflammasome activation promotes macrophage proliferation and features of plaque instability in mice with Jak2VF bone marrow.
Fig. 3: Jak2VF promotes pyroptosis and inflammation in lesions.
Fig. 4: Effects of anakinra or IL-1β antibodies on atherosclerotic plaque development.

Similar content being viewed by others

Data availability

The datasets generated and/or analysed during the current study are available in the Gene Expression Omnibus with the following accession numbers: mouse CD11b+ cells for bulk RNA-seq analysis (Extended Data Fig. 1) (GSE163689), mouse CD45+ aortic cells for scRNA-seq (GSE163536). All other relevant data are available from the corresponding authors upon reasonable request. Source data are provided with this paper.

Code availability

Gene ontology analysis used PANTHER (http://www.pantherdb.org/).

References

  1. 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 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Benjamin, E. J. et al. Heart disease and stroke statistics—2019 update: a report from the American Heart Association. Circulation 139, e56–e528 (2019).

    Article  PubMed  Google Scholar 

  4. Ridker, P. M. et al. Antiinflammatory therapy with Canakinumab for atherosclerotic disease. N. Engl. J. Med. 377, 1119–1131 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  Google Scholar 

  6. Landolfi, R. et al. Efficacy and safety of low-dose aspirin in polycythemia vera. N. Engl. J. Med. 350, 114–124 (2004).

    Article  CAS  PubMed  Google Scholar 

  7. Cordua, S. et al. Prevalence and phenotypes of JAK2 V617F and calreticulin mutations in a Danish general population. Blood 134, 469–479 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Wang, W. et al. Macrophage inflammation, erythrophagocytosis, and accelerated atherosclerosis in Jak2V617F mice. Circ. Res. 123, e35–e47 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mullally, A. et al. Physiological Jak2V617F expression causes a lethal myeloproliferative neoplasm with differential effects on hematopoietic stem and progenitor cells. Cancer Cell 17, 584–596 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. 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 

  11. Edelmann, B. et al. JAK2-V617F promotes venous thrombosis through β1/β2 integrin activation. J. Clin. Invest. 128, 4359–4371 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Snippert, H. J. et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143, 134–144 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Liu, D. J. et al. Exome-wide association study of plasma lipids in >300,000 individuals. Nat. Genet. 49, 1758–1766 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hansson, G. K., Libby, P. & Tabas, I. Inflammation and plaque vulnerability. J. Intern. Med. 278, 483–493 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Anderson, K. L. et al. PU.1 and the granulocyte- and macrophage colony-stimulating factor receptors play distinct roles in late-stage myeloid cell differentiation. Blood 94, 2310–2318 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Rajavashisth, T. et al. Heterozygous osteopetrotic (op) mutation reduces atherosclerosis in LDL receptor-deficient mice. J. Clin. Invest. 101, 2702–2710 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Celada, A. et al. The transcription factor PU.1 is involved in macrophage proliferation. J. Exp. Med. 184, 61–69 (1996).

    Article  CAS  PubMed  Google Scholar 

  18. Shimada, K. et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 36, 401–414 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hu, B. et al. The DNA-sensing AIM2 inflammasome controls radiation-induced cell death and tissue injury. Science 354, 765–768 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Schwartz, D. M. et al. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat. Rev. Drug Discov. 16, 843–862 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Katherine, L. et al. Cloning a novel member of the human interferon-inducible gene family associated with control of tumorigenicity in a model of human melanoma. Oncogene 15, 453–457 (1997).

    Article  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Paulin, N. et al. Double-strand DNA sensing Aim2 inflammasome regulates atherosclerotic plaque vulnerability. Circulation 138, 321–323 (2018).

    Article  PubMed  Google Scholar 

  24. Kayagaki, N. et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526, 666–671 (2015).

    Article  ADS  CAS  PubMed  Google Scholar 

  25. Zernecke, A. et al. Meta-analysis of leukocyte diversity in atherosclerotic mouse aortas. Circ. Res. 127, 402–426 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mesa, R. A. et al. Effects of ruxolitinib treatment on metabolic and nutritional parameters in patients with myelofibrosis from COMFORT-I. Clin. Lymphoma Myeloma Leuk. 15, 214–221.e1 (2015).

    Article  PubMed  Google Scholar 

  27. Rao, T. N. et al. JAK2-mutant hematopoietic cells display metabolic alterations that can be targeted to treat myeloproliferative neoplasms. Blood 134, 1832–1846 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Choudhury, R. P. et al. Arterial effects of Canakinumab in patients with atherosclerosis and type 2 diabetes or glucose intolerance. J. Am. Coll. Cardiol. 68, 1769–1780 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Evavold, C. L. et al. The pore-forming protein gasdermin D regulates interleukin-1 secretion from living macrophages. Immunity 48, 35–44.e6 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. 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 

  31. Westerterp, M. et al. Cholesterol efflux pathways suppress inflammasome activation, NETosis, and atherogenesis. Circulation 138, 898–912 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Vromman, A. et al. Stage-dependent differential effects of interleukin-1 isoforms on experimental atherosclerosis. Eur. Heart J. 40, 2482–2491 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Mullally, A. et al. Distinct roles for long-term hematopoietic stem cells and erythroid precursor cells in a murine model of Jak2V617F-mediated polycythemia vera. Blood 120, 166–172 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Iacobucci, I. et al. Truncating erythropoietin receptor rearrangements in acute lymphoblastic leukemia. Cancer Cell 29, 186–200 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Maude, S. L. et al. Efficacy of JAK/STAT pathway inhibition in murine xenograft models of early T-cell precursor (ETP) acute lymphoblastic leukemia. Blood 125, 1759–1767 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Thomas, D. G. et al. LXR suppresses inflammatory gene expression and neutrophil migration through cis-repression and cholesterol efflux. Cell Rep. 25, 3774–3785.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Thomas, P. D. et al. PANTHER: a library of protein families and subfamilies indexed by function. Genome Res. 13, 2129–2141 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kotini, A. G. et al. Stage-specific human induced pluripotent stem cells map the progression of myeloid transformation to transplantable leukemia. Cell Stem Cell 20, 315–328.e7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Fidler, T. P. et al. Glucose metabolism is required for platelet hyperactivation in a murine model of type 1 diabetes. Diabetes 68, 932–938 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Durinck, S., Spellman, P. T., Birney, E. & Huber, W. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat. Protocols 4, 1184–1191 (2009).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Supported by the Leducq Foundation (TNE-18CVD04) and NIH grants HL-118567, HL-155431, HL-148071, HL-137663, CA108671, CA008748, HL-080472, HL-134892. T.P.F. was supported by a Columbia Clinical and Translational Science Award (CTSA) (TL1-TR-001875), and F32HL151051-01. D.G.T. was supported by F30HL137327. E.P.P. is supported by the US National Institutes of Health (NIH) grants R01HL137219 and R01CA225231, by the New York State Stem Cell Board, and by a Scholar Award from the Leukemia and Lymphoma Society (LLS). C.S. and S.M. were supported by the German Research Foundation (DFG), CRC 1123, grants A07 (C.S.) and B06 (S.M.), as well as the DZHK (German Centre for Cardiovascular Research) and the BMBF (German Ministry of Education and Research) (grant 81Z0600204 to C.S.). We thank M. Gonzalez Pisfil, S. Dietzel and the Core Facility Bioimaging at the Biomedical Center, Ludwig Maximilian University, Planegg-Martinsried, Germany, for technical support. M.W. was supported by a Rosalind Franklin Fellowship from the University Medical Center Groningen and by Netherlands Organization of Scientific Research VIDI grant 917.15.350. Research reported in this publication was performed in the CCTI Flow Cytometry Core, supported in part by the Office of the Director, National Institutes of Health under awards S10RR027050 and S10OD020056. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. Images were collected and/or image processing and analysis for this work was performed in the Confocal and Specialized Microscopy Shared Resource of the Herbert Irving Comprehensive Cancer Center at Columbia University, supported by NIH grant P30 CA013696.

Author information

Authors and Affiliations

Authors

Contributions

T.P.F. designed and performed experiments, analysed data and wrote the manuscript. C.X. aided in the design of scRNA-seq studies and conducted scRNA-seq analysis. M.Y. and B.H. aided in mouse studies, immunohistochemistry and analysis of data. S.A. and T.X. processed tissue and conducted histology experiments and analysis. W.L. aided in mouse studies. D.G.T. designed and conducted RNA-seq analysis of CD11b cells. M.A.H., J.P., C.S., and S.M. designed, conducted, and analysed data from studies of Confetti mice. C.S.-R. and O.S. aided in studies relating to neutrophils. A.G.K. and E.P.P. designed and conducted experiments related to human iPS-derived macrophages. L.L.L. and H.-W.S. aided in the design and analysis of studies related to metabolism, including Seahorse studies. Y.W. aided in statistical analysis of data. M.W. provided scientific feedback thorought the project, including specifically designing and conducting studies on wild-type versus Gsdmd−/− mice. N.W. designed, performed, and analysed data related to atherosclerosis experiments. B.E., R.L.L. and P.L. provided reagents, designed experiments, and wrote the manuscript. M.P.R. designed experiments and aided in analysis of studies related to scRNA-seq. A.R.T. designed experiments, analysed data, and wrote the manuscript.

Corresponding authors

Correspondence to Trevor P. Fidler, Nan Wang or Alan R. Tall.

Ethics declarations

Competing interests

R.L.L. is on the supervisory board of Qiagen and is a scientific advisor to Loxo, Imago, C4 Therapeutics and Isoplexis, which include equity interest. He receives research support from and consulted for Celgene and Roche and has consulted for Lilly, Janssen, Astellas, Morphosys and Novartis. He has received honoraria from Roche, Lilly and Amgen for invited lectures and from Gilead for grant reviews. P.L. is an unpaid consultant to, or involved in clinical trials for Amgen, AstraZeneca, Esperion Therapeutics, Ionis Pharmaceuticals, Kowa Pharmaceuticals, Novartis, Pfizer, Sanofi-Regeneron, and XBiotech, Inc. P.L. is a member of the scientific advisory board for Amgen, Corvidia Therapeutics, DalCor Pharmaceuticals, IFM Therapeutics, Kowa Pharmaceuticals, Olatec Therapeutics, Medimmune, Novartis, and XBiotech, Inc. P.L.’s laboratory has received research funding in the last two years from Novartis. A.R.T. is a scientific advisory board member for Amgen, Staten Biotech and Fortico Biotech and is a consultant for Janssen, CSL and the Medicines Company. B.E. has research funding from Celgene and Deerfield, and consulting fees from Grail. E.P.P. has received honoraria from Celgene and Merck and research support from Incyte for research not related to this study.

Additional information

Peer review information Nature thanks Clinton Robbins, Eicke Latz and the other, anonymous, reviewer(s) 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 figures and tables

Extended Data Fig. 1 Bulk RNA-seq analysis of CD11b+ splenocytes.

a, b, Gene ontology analysis of CD11b+ splenocytes from wild-type (a; n = 6 control, n = 7 Jak2VF mice) and Ldlr−/− recipient mice (b; n = 6 control, n = 4 Jak2VF mice) containing bone marrow from control or Mx1-Jak2VF transgenic mice.

Source data

Extended Data Fig. 2 Monocyte/macrophage- and neutrophil-specific expression of Jak2VF.

a, Scheme of experimental procedures for atherosclerosis studies in mice with monocyte/macrophage-specific expression of Jak2VF. b, qPCR analysis of Jak2VF mRNA in blood monocytes (n = 4 samples (3 mice pooled per sample,12-total mice per genotype)) and neutrophils (n = 5 (3 mice pooled per sample, 15 total mice per genotype)). c, Serum cholesterol levels (n = 14, 18, 19 control, n = 13, 18, 19 Jak2VF mice for day 21, 56, 83, respectively). d, Scheme of experimental procedure for studies of mice with neutrophil-specific expression of Jak2VF. e, qPCR analysis of Jak2VF mRNA in blood monocytes and neutrophils. Each point indicates four mice pooled together (n = 3 pooled samples, 12 total mice per genotype). f, Serum cholesterol levels. gk, Quantification of lesion area (g), percentage necrotic core area (h), percentage macrophage area (i), percentage collagen area (j), and percentage smooth muscle cell (SMC) area (k; n = 12 control, n = 9 Jak2VF mice). Mean ± s.e.m.; two-tailed Mann–Whitney test (g), two-tailed t-test (f, hk), two-way ANOVA followed by Bonferroni multiple comparison post hoc test (b, c, e).

Source data

Extended Data Fig. 3 Jak2VF allele burden and blood cell counts in mice with Jak2VF clonal haematopoiesis.

a, Experimental design for Jak2VF clonal haematopoiesis mice. be, Fraction CD45.2:CD45.1 (Jak2VF/control:CD45.1) in blood monocytes (b), all CD45+ cells (c), neutrophils (d), and lymphocytes (e); black arrow, induction of Mx1-cre (n = 10, 19, 18, 9, 20 control, n = 9, 18, 19, 9, 17 Jak2VFmice, for day −7, 26, 40, 47, 53, respectively). fj, Blood cell counts of white blood cells (f; WBC), lymphocytes (g), monocytes (h), neutrophils (i), and red blood cells (j; RBCs) (n = 17, 19, 10, 21 control, n = 17, 16, 10, 13 Jak2VF mice, for day −7, 26, 40, 53, respectively). k, Serum cholesterol (n = 17, 18, 19 control, n = 17, 12, 18 Jak2VF mice, for day 26, 40, 53, respectively). l, Spleen weight (n = 19 mice). Mean ± s.e.m.; two-tailed Mann–Whitney test (l), two-way ANOVA followed by Bonferroni multiple comparison post hoc test (bk).

Source data

Extended Data Fig. 4 Jak2VF monocytes/macrophages have increased recruitment and proliferation in lesions.

a, Representative immunofluorescence images of aortic roots stained for latex beads (green), MAC2 (red), and DAPI (blue). White arrows, beads; scale bars, 100 μm. b, Quantification of beads in lesions. c, qPCR analysis of mRNA from CD11b+ aortic cells sorted for CD45.1 or CD45.2; PolE (n = 9 control, n = 11 Jak2VF mice). d, e, CD68+ cells isolated from aortas quantified for percentage EdU incorporation (d) and S/G2/M phase with propidium iodine staining (e; n = 11 control, n = 9 Jak2VF mice). f, Representative images of lesions from mice with Mx1-Confetti or Mx1-Jak2VF-Confetti expression in bone marrow. Cyan, cyan fluorescent protein (membrane-tethered CFP); yellow, membrane yellow fluorescent protein (cytoplasmic YFP); red, red fluorescent protein (cytoplasmic RFP). Yellow dashed lines, lesion boundary; scale bars, 25 μm. g, Quantification of the number of cells per clone in Confetti lesions (n = 149 control, n = 82 Jak2VF clones; four mice per group). h, Representative immunofluorescence images of aortic roots stained for MAC2 (green), IL-1β (red), and DAPI (blue). Scale bars, 50 μm. i, Quantification of total IL-1β fluorescent intensity normalized to area (n = 15 mice). Mean ± s.e.m.; two-tailed t-test (b), two-tailed Mann–Whitney test (d, e, g, i), Kruskal–Wallis two-tailed test with Dunn’s comparison (c).

Source data

Extended Data Fig. 5 IL-1β promotes increased ERK/AKT-driven proliferation of Jak2VF macrophages associated with increased glycolytic metabolism and mitochondrial ROS generation.

a, b, Quantification of serum IL-18 (a; n = 7 control, n = 9 Casp1/11−/−, n = 9 Jak2VF, n = 9 Casp1/11−/−Jak2VF mice) and cholesterol (b; n = 16 control, n = 11 Casp1/11−/−, n = 7 Jak2VF, n = 11 Casp1/11−/−Jak2VF mice) following 12-week WTD. c, Macrophage proliferation marked by 3H-thymidine incorporation into BMDMs during 16-h incubations (n = 4 biological replicates, replicated twice). d, Representative immunoblot analysis of BMDMs co-incubated with anakinra. e, f, Densitometric quantification of pERK1/2 (e; n = 4 control, n = 5 control + anakinra, n = 5 Jak2VF, n = 4 Jak2VF + anakinra; biological replicates from three mice pooled, representative of two experiments) and pAKT (f; n = 5 biological replicates from three mice pooled, representative of two experiments). g, Incorporation of BrdU into BMDMs co-incubated for 16 h with 100 ng ml−1 M-CSF and the indicated inhibitors (n = 6 biological replicates from three mice pooled, n = 5 M-CSF + FR180204). h, LDH release from non-stimulated BMDMs following 7-day culture (n = 24 control, n = 24 Casp1/11−/−, n = 22 Jak2VF, n = 19 Jak2VFCasp1/11−/− biological replicates from three mice repeated four times). i, j, Glycolysis rate indicated by the extracellular acidification rate (ECAR) (i) and mitochondrial respiration marked by oxygen consumption rate (OCR) (j; n = 15 control, n = 18 Jak2VF biological replicates repeated twice). k, Mitochondrial potential in CD11b+ splenocytes measured by tetramethylrhodamine, methyl ester, perchlorate (TMRM) geometric mean fluorescence intensity (geo. MFI) (n = 6 mice). l, MitoSOX geo. MFI in BMDMs (n = 3 mice). m, Quantification of mitochondrial localized 8-OHdG in BMDMs (n = 6 mice). n, o, Total cellular ROS in BMDMs marked by relative fluorescence units (RFU) of DCFDA (n) and Cell ROX (o; n = 6 biological replicates from three mice pooled). Mean ± s.e.m.; one-way ANOVA followed by Tukey’s post hoc test (a, b, e, o), Kruskal–Wallis two-tailed test with Dunn’s comparison (f, h, n), two-way ANOVA followed by Bonferroni’s multiple comparison post hoc test (c, g, i, j), two-tailed t-test (km).

Source data

Extended Data Fig. 6 JAK2VF macrophages display increased NLRP3 and AIM2 inflammasome activation.

a, b, IL-1β was quantified in medium from mouse BMDMs treated with pdAdT for 6 h (a) or LPS (20 ng ml−1; b) followed by a 1-h incubation with ATP (n = 6 biological replicates, representative of five experiments). Vertical P values are relative to LPS within the same genotype. c, d, IL-1β release from human iPSC-macrophages stimulated with pdAdT (c; n = 12 baseline, n = 6 treatments biological replicates representative of two experiments) and nigericin (d; n = 6 biological replicates, representative of two experiments). Vertical P values are relative to baseline within the same genotype. e, Immunoblot analysis of NLRP3 and AIM2 in protein lysates from BMDMs. f, g, Densiometric quantification of AIM2 (f) and NLRP3 (g; n = 8 biological replicates from four mice per group pooled together). h, Aim2 mRNA expression in BMDMs incubated in the presence of IFNγ-neutralizing antibodies for 24 h (n = 6 biological replicates). i, Experimental scheme of atherosclerosis studies conducted in mice with Jak2VF bone marrow deficient in Nlrp3 or Aim2. j, k, Plasma cholesterol following 12-week WTD in Jak2VFNlrp3−/− (j; n = 15 Jak2VF, n = 19 Jak2VFNlrp3−/− mice) and Jak2VFAim2−/− mice (k; n = 24 Jak2VF, n = 25 Jak2VFAim2−/− mice). l. Representative immunofluorescence image of aortic roots stained for DAPI (blue) and AIM2 (magenta). Yellow dashed lines, lesions; scale bars, 50 μm. m, Quantification of AIM2 mean fluorescence intensity in lesions (n = 16 control, n = 18 Jak2VF mice). Mean ± s.e.m.; two-way ANOVA followed by Tukey’s post hoc test (ad), one-way ANOVA followed by Tukey’s post hoc test (h), two-tailed t-test (j, k), two-tailed Mann–Whitney test (f, g, m).

Source data

Extended Data Fig. 7 Haematological parameters and atherosclerosis in Ldlr−/− mice transplanted with Gsdmd−/− bone marrow.

a, WBCs; b, lymphocytes (n = 14 Jak2VF, n = 13 Jak2VFGsdmd−/− mice); c, monocytes (n = 14 Jak2VF, n = 12 Jak2VFGsdmd−/− mice); d, neutrophils (n = 14 Jak2VF, n = 13 Jak2VFGsdmd−/− mice); e, RBCs (n = 13 mice). fh, Jak2VF burden in blood lymphocytes (f), neutrophils (g), and monocytes (h; n = 14 Jak2VF, n = 13 Jak2VFGsdmd−/− mice). i, Serum cholesterol (n = 13 mice). j, Representative H&E images of aortic root lesions from Ldlr−/− mice with wild-type (WT) or Gsdmd−/− bone marrow fed a WTD for 12 weeks. Dashed lines, necrotic core; scale bars 200 μm. k, Quantification of lesion area. l, Percentage necrotic core area (n = 12 wild-type, n = 14 Gsdmd−/− mice). Mean ± s.e.m.; two-tailed t-test (a, b, d, e, i, k, l), two-tailed Mann–Whitney test (c, fh).

Source data

Extended Data Fig. 8 scRNA-seq reveals increased proliferation and inflammatory macrophages.

a, Heat map of top five differentially expressed genes enriched in each cluster. b, Table showing top ten differentially expressed genes in each cluster with cell annotations (each sample pooled from eight mice). c, Cell cycle classification based on cell cycle gene expression using Seurat analysis; one-tailed χ2 test, P = 1.415 × 10−10 indicates G2M cells enriched in Jak2VF lesions. d, UMAP plot of cluster 13 enriched Alox15. Blue indicates relative gene expression. e, Representative immunofluorescence images of aortic root lesions stained for MAC2 (green), ALOX15 (magenta) and DAPI (blue). White dashed lines, plaque; arrows, ALOX15+ cells; scale bars, 50 μm. f, Quantification of ALOX15+ cells per aortic root section (n = 14 Jak2VF, n = 13 Jak2VFGsdmd−/− mice). Mean ± s.e.m.; *P < 0.05 with respect to control genotype, two-tailed Mann–Whitney test (f).

Source data

Extended Data Fig. 9 Ruxolitinib reduces IL-18 and increases plasma cholesterol in individuals with MPN and mice with JAK2VF mutations.

a, b, Plasma from patients with MPN isolated before and after ruxolitinib (Rux) therapy were analysed for IL-18 (a) and total cholesterol (b; n = 17 patients). c, d, Serum from mice with chimeric 20% Jak2VF bone marrow was analysed for IL-18 (c) and total cholesterol (d; n = 20 Jak2VF, n = 22 Jak2VF + Rux mice). e, Body weight following 12 weeks WTD (n = 19 Jak2VF, n = 20 Jak2VF + Rux mice). fj, Blood counts of WBCs (f), lymphocytes (g), neutrophils (h), monocytes (i) and RBCs (j; n = 18 Jak2VF, n = 22 Jak2VF + Rux mice). k, l, Jak2VF burden in blood neutrophils (k) and monocytes (l; n = 17 Jak2VF, n = 20 Jak2VF + Rux mice). m, Representative H&E images of aortic root lesions from mice with 20% Jak2VF chimeric bone marrow treated with Rux. Dashed lines, necrotic core; scale bars, 200 μm. n, o, Quantification of lesion area (n; n = 17 Jak2VF, n = 18 Jak2VF + Rux mice) and percentage necrotic core area (o; n = 18 mice). p, Representative picrosirius red-stained lesions (black lines indicate cap thickness). Scale bars, 200 μm. q, Quantification of cap thickness (n = 18 mice). Mean ± s.e.m.; Wilcoxon paired two-tailed test (a), two-tailed Student’s paired t-test (b); two-tailed Mann–Whitney test (c, ej, np), two-tailed t-test (d), two-way ANOVA followed by Tukey’s post hoc test (k, l).

Source data

Extended Data Fig. 10 Inhibition of IL-1 reduces macrophage proliferation and density in plaques.

a, Terminal serum cholesterol levels in mice on a WTD and treated with anakinra for 7 weeks (n = 9 control, n = 8 control + anakinra, n = 7 Jak2VF, n = 9 Jak2VF +anakinra mice). b, Representative images of aortic roots following 7-week administration of anakinra or vehicle. MAC2 (green), phalloidin (cyan), Ki67 (red), DAPI (blue); arrows, Ki67+ nuclei; scale bars, 50 μm. ce, Quantification of percentage Ki67+ cells (c), macrophage density (d) and lesion area (e; n = 8 control, n = 8 control + anakinra, n = 6 Jak2VF, n = 9 Jak2VF + anakinra mice). f, Serum cholesterol following 12 weeks WTD, related to late lesions in Fig. 4 (n = 24 Jak2VF, n = 25 Jak2VF + anakinra mice). g, Representative picrosirius red-stained lesions from mice treated with anakinra for 12 weeks. Black lines indicate cap thickness; scale bars, 200 μm. h, Experimental design for Jak2VF clonal haematopoiesis mice using GFP+ cells. i, Terminal serum cholesterol (n = 18 control IgG, n = 19 control IL-1, n = 19 Jak2VF IgG, n = 17 Jak2VF IL-1 mice). j, G2-M phase monocytes from blood identified with propidium iodine staining (n = 18 control IgG, n = 20 control IL-1, n = 18 Jak2VF IgG, n = 17 Jak2VF IL-1 mice). k, Representative picrosirius red-stained aortic root lesions from mice as in j. Black lines indicate cap thickness; scale bars, 200 μm. l, Representative H&E images of aortic roots. Dashed lines, necrotic core; scale bars, 200 μm. m, Overall summary scheme. Mean ± s.e.m.; one-way ANOVA followed by Tukey’s multiple comparison post hoc test (a, ce), two-tailed t-test (f); Kruskal–Wallis two-tailed test with Dunn’s comparison (i, j).

Source data

Extended Data Table 1 Characteristics of scRNA-seq analysis

Supplementary information

Supplementary Figures

This file contains Supplementary Figures 1-3.

Reporting Summary

Supplementary Table 1

Complete Blood Counts. Cells concentrations are k/mL (RBC concentrations are 106/mL). Data are mean±Standard Deviation. * p<0.05, **p<0.01, ***p<0.001, ****p<0.0001 with respect to genotype same treatment. # p<0.05, ##p<0.01, ###p<0.001, ####p<0.0001 with respect to treatment same genotype. All statistical tests were Two-way ANOVA with Bonferroni multiple comparison post hoc test.

Supplementary Table 2

scRNA-Sequencing Gene Expression Profile. Statistics derived from a Likelihood ratio test, one-tailed P-value. Bonferroni corrected.

Video 1

Z-Stack of Mx1-Confetti Lesions.

Video 2

Z-Stack of Mx1-Jak2VF-Confetti Lesions.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fidler, T.P., Xue, C., Yalcinkaya, M. et al. The AIM2 inflammasome exacerbates atherosclerosis in clonal haematopoiesis. Nature 592, 296–301 (2021). https://doi.org/10.1038/s41586-021-03341-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-03341-5

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research