Clonal hematopoiesis of indeterminate potential (CHIP): Linking somatic mutations, hematopoiesis, chronic inflammation and cardiovascular disease

Christopher S Marnell, Alexander Bick, Pradeep Natarajan, Christopher S Marnell, Alexander Bick, Pradeep Natarajan

Abstract

Clonal hematopoiesis of indeterminate potential (CHIP) is the presence of a clonally expanded hematopoietic stem cell caused by a leukemogenic mutation in individuals without evidence of hematologic malignancy, dysplasia, or cytopenia. CHIP is associated with a 0.5-1.0% risk per year of leukemia. Remarkably, it confers a two-fold increase in cardiovascular risk independent of traditional risk factors. Roughly 80% of patients with CHIP have mutations in epigenetic regulators DNMT3A, TET2, ASXL1, DNA damage repair genes PPM1D, TP53, the regulatory tyrosine kinase JAK2, or mRNA spliceosome components SF3B1, and SRSF2. CHIP is associated with a pro-inflammatory state that has been linked to coronary artery disease, myocardial infarction, and venous thromboembolic disease, as well as prognosis among those with aortic stenosis and heart failure. Heritable and acquired risk factors are associated with increased CHIP prevalence, including germline variation, age, unhealthy lifestyle behaviors (i.e. smoking, obesity), inflammatory conditions, premature menopause, HIV and exposure to cancer therapies. This review aims to summarize emerging research on CHIP, the mechanisms underlying its important role in propagating inflammation and accelerating cardiovascular disease, and new studies detailing the role of associated risk factors and co-morbidities that increase CHIP prevalence.

Keywords: ASCVD; Aortic stenosis; CHIP; Cardio-oncology; Heart failure; Inflammation.

Copyright © 2021 Elsevier Ltd. All rights reserved.

Figures

Figure 1:. CHIP associates with an altered…
Figure 1:. CHIP associates with an altered inflammatory state, elevating cardiovascular risk
The acquisition of driver mutations (in DNMT3A, TET2, and JAK2 shown as a selection here) leads to clonal hematopoiesis of indeterminate potential (CHIP), which induces an altered inflammatory state that is associated with an increased risk of atherosclerosis, poorer outcomes in aortic stenosis and heart failure, and enhanced thrombogenesis. HSC= hematopoietic stem cell, NET = neutrophil extracellular traps (created with Biorender.com)
Figure 2:. Selected hazard ratios (HR) for…
Figure 2:. Selected hazard ratios (HR) for CHIP and incident coronary heart disease are of similar magnitude to traditional risk factors
(A) HR with 95% CI for incident coronary heart disease adjusted for age, sex, HTN, HLD, smoking, obesity, diabetes among Jackson Heart Study & FUSION participants, Summarized data from Jaiswal et al 2014 [4] (B) HR with 95% CI for incident coronary heart disease adjusted for age, sex, HTN, total and HDL cholesterol, triglycerides, smoking, and type 2 diabetes among BioImage, MDC (CHIP) and a fixed-effects meta-analysis for each gene (incorporating BioImage, MDC, JHS/Fusion/FHS). Summarized data from Jaiswal et al 2017 [7]

References

    1. Watson CJ, et al. The evolutionary dynamics and fitness landscape of clonal hematopoiesis. Science, 2020. 367(6485): p. 1449–1454.
    1. Bowman RL, Busque L, and Levine RL, Clonal Hematopoiesis and Evolution to Hematopoietic Malignancies. Cell Stem Cell, 2018. 22(2): p. 157–170.
    1. Young AL, et al. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat Commun, 2016. 7: p. 12484.
    1. Genovese G, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med, 2014. 371(26): p. 2477–87.
    1. Jaiswal S, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med, 2014. 371(26): p. 2488–98.
    1. Steensma DP, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood, 2015. 126(1): p. 9–16.
    1. Jaiswal S, et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N Engl J Med, 2017. 377(2): p. 111–121.
    1. Dorsheimer L, et al. Association of Mutations Contributing to Clonal Hematopoiesis With Prognosis in Chronic Ischemic Heart Failure. JAMA Cardiol, 2019. 4(1): p. 25–33.
    1. Mas-Peiro S, et al. Clonal haematopoiesis in patients with degenerative aortic valve stenosis undergoing transcatheter aortic valve implantation. Eur Heart J, 2020. 41(8): p. 933–939.
    1. Cancer Genome Atlas Research, N., et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med, 2013. 368(22): p. 2059–74.
    1. Lindsley RC, et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood, 2015. 125(9): p. 1367–76.
    1. Bejar R, et al. Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med, 2011. 364(26): p. 2496–506.
    1. Papaemmanuil E, et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood, 2013. 122(22): p. 3616–27; quiz 3699.
    1. Jeong M, et al. Loss of Dnmt3a Immortalizes Hematopoietic Stem Cells In Vivo. Cell Rep, 2018. 23(1): p. 1–10.
    1. Challen GA, et al. Dnmt3a is essential for hematopoietic stem cell differentiation. Nat Genet, 2011. 44(1): p. 23–31.
    1. Hormaechea-Agulla D, et al. Chronic infection drives Dnmt3a-loss-of-function clonal hematopoiesis via IFNgamma signaling. Cell Stem Cell, 2021.
    1. Sano S, et al. CRISPR-Mediated Gene Editing to Assess the Roles of Tet2 and Dnmt3a in Clonal Hematopoiesis and Cardiovascular Disease. Circ Res, 2018. 123(3): p. 335–341.
    1. Abplanalp WT, et al. Clonal Hematopoiesis-Driver DNMT3A Mutations Alter Immune Cells in Heart Failure. Circ Res, 2021. 128(2): p. 216–228.
    1. Buscarlet M, et al. Lineage restriction analyses in CHIP indicate myeloid bias for TET2 and multipotent stem cell origin for DNMT3A. Blood, 2018. 132(3): p. 277–280.
    1. Cull AH, et al. Tet2 restrains inflammatory gene expression in macrophages. Exp Hematol, 2017. 55: p. 56–70 e13.
    1. Fuster JJ, et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science, 2017. 355(6327): p. 842–847.
    1. Zhang Q, et al. Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6. Nature, 2015. 525(7569): p. 389–393.
    1. Sano S, et al. Tet2-Mediated Clonal Hematopoiesis Accelerates Heart Failure Through a Mechanism Involving the IL-1beta/NLRP3 Inflammasome. J Am Coll Cardiol, 2018. 71(8): p. 875–886.
    1. Bick AG, et al. Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature, 2020. 586(7831): p. 763–768.
    1. Bick AG, et al. Genetic Interleukin 6 Signaling Deficiency Attenuates Cardiovascular Risk in Clonal Hematopoiesis. Circulation, 2020. 141(2): p. 124–131.
    1. Asada S and Kitamura T, Aberrant histone modifications induced by mutant ASXL1 in myeloid neoplasms. Int J Hematol, 2019. 110(2): p. 179–186.
    1. Dawoud AAZ, Tapper WJ, and Cross NCP, Clonal myelopoiesis in the UK Biobank cohort: ASXL1 mutations are strongly associated with smoking. Leukemia, 2020. 34(10): p. 2660–2672.
    1. Bick AG, et al. Increased CHIP Prevalence Amongst People Living with HIV. medRxiv, 2020.
    1. Fujino T and Kitamura T, ASXL1 mutation in clonal hematopoiesis. Exp Hematol, 2020. 83: p. 74–84.
    1. Jeong JJ, et al. Cytokine-Regulated Phosphorylation and Activation of TET2 by JAK2 in Hematopoiesis. Cancer Discov, 2019. 9(6): p. 778–795.
    1. Wolach O, et al. Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci Transl Med, 2018. 10(436).
    1. Wang W, et al. Macrophage Inflammation, Erythrophagocytosis, and Accelerated Atherosclerosis in Jak2 (V617F) Mice. Circ Res, 2018. 123(11): p. e35–e47.
    1. Fidler TP, et al. The AIM2 inflammasome exacerbates atherosclerosis in clonal haematopoiesis. Nature, 2021.
    1. Sano S, et al. JAK2 (V617F)-Mediated Clonal Hematopoiesis Accelerates Pathological Remodeling in Murine Heart Failure. JACC Basic Transl Sci, 2019. 4(6): p. 684–697.
    1. Hsu JI, et al. PPM1D Mutations Drive Clonal Hematopoiesis in Response to Cytotoxic Chemotherapy. Cell Stem Cell, 2018. 23(5): p. 700–713 e6.
    1. Chen S, et al. Mutant p53 drives clonal hematopoiesis through modulating epigenetic pathway. Nat Commun, 2019. 10(1): p. 5649.
    1. Zink F, et al. Clonal hematopoiesis, with and without candidate driver mutations, is common in the elderly. Blood, 2017. 130(6): p. 742–752.
    1. Heyde A, et al. Increased stem cell proliferation in atherosclerosis accelerates clonal hematopoiesis. Cell, 2021. 184(5): p. 1348–1361 e22.
    1. Terao C, et al. Chromosomal alterations among age-related haematopoietic clones in Japan. Nature, 2020. 584(7819): p. 130–135.
    1. Loh PR, et al. Insights into clonal haematopoiesis from 8,342 mosaic chromosomal alterations. Nature, 2018. 559(7714): p. 350–355.
    1. Loh PR, Genovese G, and McCarroll SA, Monogenic and polygenic inheritance become instruments for clonal selection. Nature, 2020. 584(7819): p. 136–141.
    1. Zekavat SM, et al. Hematopoietic mosaic chromosomal alterations and risk for infection among 767,891 individuals without blood cancer. medRxiv, 2020.
    1. Hinds DA, et al. Germ line variants predispose to both JAK2 V617F clonal hematopoiesis and myeloproliferative neoplasms. Blood, 2016. 128(8): p. 1121–8.
    1. Wang JC and Bennett M, Aging and atherosclerosis: mechanisms, functional consequences, and potential therapeutics for cellular senescence. Circ Res, 2012. 111(2): p. 245–59.
    1. Welch JS, et al. The origin and evolution of mutations in acute myeloid leukemia. Cell, 2012. 150(2): p. 264–78.
    1. Xie M, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med, 2014. 20(12): p. 1472–8.
    1. Perdigones N, et al. Clonal hematopoiesis in patients with dyskeratosis congenita. Am J Hematol, 2016. 91(12): p. 1227–1233.
    1. Ojha J, et al. Genetic Variation Associated with Longer Telomere Length Increases Risk of Chronic Lymphocytic Leukemia. Cancer Epidemiol Biomarkers Prev, 2016. 25(7): p. 1043–9.
    1. Telomeres Mendelian Randomization, C., et al. Association Between Telomere Length and Risk of Cancer and Non-Neoplastic Diseases: A Mendelian Randomization Study. JAMA Oncol, 2017. 3(5): p. 636–651.
    1. Li C, et al. Genome-wide Association Analysis in Humans Links Nucleotide Metabolism to Leukocyte Telomere Length. Am J Hum Genet, 2020. 106(3): p. 389–404.
    1. Nakao T, et al. Bidirectional Mendelian randomization supports bidirectional causality between telomere length and clonal hematopoiesis of intermediate potential. medRxiv, 2021: p. 2021.02.26.21252199.
    1. Aviv A and Levy D, Hemothelium, Clonal Hematopoiesis of Indeterminate Potential, and Atherosclerosis. Circulation, 2019. 139(1): p. 7–9.
    1. Oguro H, The Roles of Cholesterol and Its Metabolites in Normal and Malignant Hematopoiesis. Front Endocrinol (Lausanne), 2019. 10: p. 204.
    1. Morgan PK, et al. Hematopoiesis is regulated by cholesterol efflux pathways and lipid rafts: connections with cardiovascular diseases. J Lipid Res, 2020. 61(5): p. 667–675.
    1. Yvan-Charvet L, et al. ATP-binding cassette transporters and HDL suppress hematopoietic stem cell proliferation. Science, 2010. 328(5986): p. 1689–93.
    1. Murphy AJ, et al. ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte accumulation in atherosclerotic lesions in mice. J Clin Invest, 2011. 121(10): p. 4138–49.
    1. Liu DJ, et al. Exome-wide association study of plasma lipids in >300,000 individuals. Nat Genet, 2017. 49(12): p. 1758–1766.
    1. Haring B, et al. Healthy Lifestyle and Clonal Hematopoiesis of Indeterminate Potential: Results From the Women’s Health Initiative. J Am Heart Assoc, 2021. 10(5): p. e018789.
    1. Bolton KL, et al. Cancer therapy shapes the fitness landscape of clonal hematopoiesis. Nat Genet, 2020. 52(11): p. 1219–1226.
    1. Buscarlet M, et al. DNMT3A and TET2 dominate clonal hematopoiesis and demonstrate benign phenotypes and different genetic predispositions. Blood, 2017. 130(6): p. 753–762.
    1. Fuster JJ, et al. TET2-Loss-of-Function-Driven Clonal Hematopoiesis Exacerbates Experimental Insulin Resistance in Aging and Obesity. Cell Rep, 2020. 33(4): p. 108326.
    1. McAlpine CS, et al. Sleep modulates haematopoiesis and protects against atherosclerosis. Nature, 2019. 566(7744): p. 383–387.
    1. Honigberg MC, et al. Association of Premature Natural and Surgical Menopause With Incident Cardiovascular Disease. JAMA, 2019.
    1. Honigberg MC, et al. Premature Menopause, Clonal Hematopoiesis, and Coronary Artery Disease in Postmenopausal Women. Circulation, 2021. 143(5): p. 410–423.
    1. Ricard L, et al. Clonal haematopoiesis is increased in early onset in systemic sclerosis. Rheumatology (Oxford), 2020. 59(11): p. 3499–3504.
    1. Savola P, et al. Clonal hematopoiesis in patients with rheumatoid arthritis. Blood Cancer J, 2018. 8(8): p. 69.
    1. Liu Y and Kaplan MJ, Cardiovascular disease in systemic lupus erythematosus: an update. Curr Opin Rheumatol, 2018. 30(5): p. 441–448.
    1. Bekele DI and Patnaik MM, Autoimmunity, Clonal Hematopoiesis, and Myeloid Neoplasms. Rheum Dis Clin North Am, 2020. 46(3): p. 429–444.
    1. Card TR, Langan SM, and Chu TP, Extra-Gastrointestinal Manifestations of Inflammatory Bowel Disease May Be Less Common Than Previously Reported. Dig Dis Sci, 2016. 61(9): p. 2619–26.
    1. Kristensen SL, et al. Disease activity in inflammatory bowel disease is associated with increased risk of myocardial infarction, stroke and cardiovascular death--a Danish nationwide cohort study. PLoS One, 2013. 8(2): p. e56944.
    1. Singh S, et al. Inflammatory bowel disease is associated with an increased risk of melanoma: a systematic review and meta-analysis. Clin Gastroenterol Hepatol, 2014. 12(2): p. 210–8.
    1. Zhang CRC, et al. Inflammatory cytokines promote clonal hematopoiesis with specific mutations in ulcerative colitis patients. Exp Hematol, 2019. 80: p. 36–41 e3.
    1. Jiang W, et al. Elevated levels of Th17 cells and Th17-related cytokines are associated with disease activity in patients with inflammatory bowel disease. Inflamm Res, 2014. 63(11): p. 943–50.
    1. Kaner JD, et al. HIV portends a poor prognosis in myelodysplastic syndromes. Leuk Lymphoma, 2019. 60(14): p. 3529–3535.
    1. Dharan NJ, et al. Age-related clonal haematopoiesis is more prevalent in older adults with HIV: the ARCHIVE study. medRxiv, 2020: p. 2020.11.19.20235069.
    1. Gibson CJ, et al. Clonal Hematopoiesis Associated With Adverse Outcomes After Autologous Stem-Cell Transplantation for Lymphoma. J Clin Oncol, 2017. 35(14): p. 1598–1605.
    1. Miller PG, et al. Fitness Landscape of Clonal Hematopoiesis Under Selective Pressure of Immune Checkpoint Blockade. JCO Precision Oncology, 2020(4): p. 1027–1033.
    1. Okamura R, et al. High prevalence of clonal hematopoiesis-type genomic abnormalities in cell-free DNA in invasive gliomas after treatment. Int J Cancer, 2021.
    1. Snyder MW, et al. Cell-free DNA Comprises an In Vivo Nucleosome Footprint that Informs Its Tissues-Of-Origin. Cell, 2016. 164(1-2): p. 57–68.
    1. Moss J, et al. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat Commun, 2018. 9(1): p. 5068.
    1. Lui YY, Chik KW, and Lo YM, Does centrifugation cause the ex vivo release of DNA from blood cells? Clin Chem, 2002. 48(11): p. 2074–6.
    1. Svensson EC, et al. Abstract 15111: TET2-Driven Clonal Hematopoiesis Predicts Enhanced Response to Canakinumab in the CANTOS Trial: An Exploratory Analysis. Circulation, 2018. 138(Suppl_1): p. A15111–A15111.
    1. Cimmino L, et al. Restoration of TET2 Function Blocks Aberrant Self-Renewal and Leukemia Progression. Cell, 2017. 170(6): p. 1079–1095 e20.
    1. Tang Y, et al. Inhibition of JAK2 Suppresses Myelopoiesis and Atherosclerosis in Apoe(−/−) Mice. Cardiovasc Drugs Ther, 2020. 34(2): p. 145–152.
    1. Fuster JJ and Walsh K, Somatic Mutations and Clonal Hematopoiesis: Unexpected Potential New Drivers of Age-Related Cardiovascular Disease. Circ Res, 2018. 122(3): p. 523–532.

Source: PubMed

3
Subscribe