CC BY-NC-ND 4.0 · Thromb Haemost 2022; 122(09): 1435-1442
DOI: 10.1055/a-1830-2147
Invited Mini Series: Novel Clinical Concepts in Thrombosis

Hematopoiesis of Indeterminate Potential and Atherothrombotic Risk

Andrew J. Murphy
1   Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Dragana Dragoljevic
1   Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Pradeep Natarajan
2   Cardiology Division, Department of Medicine, Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
3   Department of Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States
,
Nan Wang
4   Division of Molecular Medicine, Department of Medicine, Columbia University Medical Center, New York, United States
› Author Affiliations
Funding A.J.M is supported by project (APP1142938) and investigator (APP1194329) grants from the National Health and Medical Research Council and a CSL Centenary Award. D.D is supported by a grant from the Jack Brockhoff Foundation (JBF 4867–2021) and Diabetes Australia. P.N. is supported by grants from the National Heart, Lung, and Blood Institute (R01HL142711, R01HL148050, R01HL151283, R01HL127564, R01HL148565, R01HL135242, R01HL151152), National Institute of Diabetes and Digestive and Kidney Diseases (R01DK125782), and Massachusetts General Hospital (Paul and Phyllis Fireman Endowed Chair in Vascular Medicine) and by a grant from Fondation Leducq (TNE-18CVD04). N.W. is supported by grants from the National Heart, Lung, and Blood Institute (R01HL118567, R01HL148071).

Abstract

Hematopoiesis is the process of blood production, essential for the continued supply of immune cells and red blood cells. However, the proliferative nature of hematopoietic stem cells (HSCs) renders them susceptible to developing somatic mutations. HSCs carrying a mutation can gain a selective advantage over normal HSCs and result in hematological disorders. One such disorder is termed clonal hematopoiesis of indeterminate potential (CHIP), a premalignant state associated with aging, where the mutant HSCs are responsible for producing a small portion of mature immune cells in the circulation and subsequently in tissues. People with CHIP have been shown to have an increased risk of mortality due to cardiovascular disease (CVD). Why this occurs is under rigorous investigation, but the majority of the studies to date have suggested that increased atherosclerosis is due to heightened inflammatory cytokine release from mutant lesional macrophages. However, given CHIP is driven by several mutations, other hematopoietic lineages can be altered to promote CVD. In this review we explore the relationship between mutations in genes causing CHIP and atherothrombotic disorders, along with potential mechanisms of enhanced clonal outgrowth and potential therapies and strategies to slow CHIP progression.



Publication History

Received: 08 November 2021

Accepted: 23 February 2022

Accepted Manuscript online:
20 April 2022

Article published online:
18 June 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Geiger H, de Haan G, Florian MC. The ageing haematopoietic stem cell compartment. Nat Rev Immunol 2013; 13 (05) 376-389
  • 2 Challen GA, Goodell MA. Clonal hematopoiesis: mechanisms driving dominance of stem cell clones. Blood 2020; 136 (14) 1590-1598
  • 3 Lee-Six H, Øbro NF, Shepherd MS. et al. Population dynamics of normal human blood inferred from somatic mutations. Nature 2018; 561 (7724): 473-478
  • 4 Osorio FG, Rosendahl Huber A, Oka R. et al. Somatic mutations reveal lineage relationships and age-related mutagenesis in human hematopoiesis. Cell Rep 2018; 25 (09) 2308-2316.e4
  • 5 Busque L, Patel JP, Figueroa ME. et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat Genet 2012; 44 (11) 1179-1181
  • 6 Shlush LI, Zandi S, Mitchell A. et al; HALT Pan-Leukemia Gene Panel Consortium. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 2014; 506 (7488): 328-333
  • 7 Welch JS, Ley TJ, Link DC. et al. The origin and evolution of mutations in acute myeloid leukemia. Cell 2012; 150 (02) 264-278
  • 8 Watson CJ, Papula AL, Poon GYP. et al. The evolutionary dynamics and fitness landscape of clonal hematopoiesis. Science 2020; 367 (6485): 1449-1454
  • 9 Natarajan P, Jaiswal S, Kathiresan S. Clonal hematopoiesis: somatic mutations in blood cells and atherosclerosis. Circ Genom Precis Med 2018; 11 (07) e001926
  • 10 Steensma DP, Bejar R, Jaiswal S. et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood 2015; 126 (01) 9-16
  • 11 Bick AG, Pirruccello JP, Griffin GK. et al. Genetic interleukin 6 signaling deficiency attenuates cardiovascular risk in clonal hematopoiesis. Circulation 2020; 141 (02) 124-131
  • 12 Genovese G, Kähler AK, Handsaker RE. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 2014; 371 (26) 2477-2487
  • 13 Jaiswal S, Fontanillas P, Flannick J. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 2014; 371 (26) 2488-2498
  • 14 Young AL, Challen GA, Birmann BM, Druley TE. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat Commun 2016; 7: 12484
  • 15 Jaiswal S, Libby P. Clonal haematopoiesis: connecting ageing and inflammation in cardiovascular disease. Nat Rev Cardiol 2020; 17 (03) 137-144
  • 16 Veiga CB, Lawrence EM, Murphy AJ, Herold MJ, Dragoljevic D. Myelodysplasia syndrome, clonal hematopoiesis and cardiovascular disease. Cancers (Basel) 2021; 13 (08) 13
  • 17 Chambers SM, Boles NC, Lin KY. et al. Hematopoietic fingerprints: an expression database of stem cells and their progeny. Cell Stem Cell 2007; 1 (05) 578-591
  • 18 Izzo F, Lee SC, Poran A. et al. DNA methylation disruption reshapes the hematopoietic differentiation landscape. Nat Genet 2020; 52 (04) 378-387
  • 19 Zhang X, Su J, Jeong M. et al. DNMT3A and TET2 compete and cooperate to repress lineage-specific transcription factors in hematopoietic stem cells. Nat Genet 2016; 48 (09) 1014-1023
  • 20 Dorsheimer L, Assmus B, Rasper T. et al. Association of mutations contributing to clonal hematopoiesis with prognosis in chronic ischemic heart failure. JAMA Cardiol 2019; 4 (01) 25-33
  • 21 Jaiswal S, Natarajan P, Silver AJ. et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N Engl J Med 2017; 377 (02) 111-121
  • 22 Pascual-Figal DA, Bayes-Genis A, Díez-Díez M. et al. Clonal hematopoiesis and risk of progression of heart failure with reduced left ventricular ejection fraction. J Am Coll Cardiol 2021; 77 (14) 1747-1759
  • 23 Fuster JJ, MacLauchlan S, Zuriaga MA. et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science 2017; 355 (6327): 842-847
  • 24 Abplanalp WT, Mas-Peiro S, Cremer S, John D, Dimmeler S, Zeiher AM. Association of clonal hematopoiesis of indeterminate potential with inflammatory gene expression in patients with severe degenerative aortic valve stenosis or chronic postischemic heart failure. JAMA Cardiol 2020; 5 (10) 1170-1175
  • 25 Bick AG, Weinstock JS, Nandakumar SK. et al; NHLBI Trans-Omics for Precision Medicine Consortium. Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature 2020; 586 (7831): 763-768
  • 26 Cordua S, Kjaer L, Skov V, Pallisgaard N, Hasselbalch HC, Ellervik C. Prevalence and phenotypes of JAK2 V617F and calreticulin mutations in a Danish general population. Blood 2019; 134 (05) 469-479
  • 27 Khetarpal SA, Qamar A, Bick AG. et al. Clonal hematopoiesis of indeterminate potential reshapes age-related CVD: JACC review topic of the week. J Am Coll Cardiol 2019; 74 (04) 578-586
  • 28 Liu DJ, Peloso GM, Yu H. et al; Charge Diabetes Working Group, EPIC-InterAct Consortium, EPIC-CVD Consortium, GOLD Consortium, VA Million Veteran Program. Exome-wide association study of plasma lipids in >300,000 individuals. Nat Genet 2017; 49 (12) 1758-1766
  • 29 Wang W, Liu W, Fidler T. et al. Macrophage inflammation, erythrophagocytosis, and accelerated atherosclerosis in Jak V617F mice. Circ Res 2018; 123 (11) e35-e47
  • 30 Fidler TP, Xue C, Yalcinkaya M. et al. The AIM2 inflammasome exacerbates atherosclerosis in clonal haematopoiesis. Nature 2021; 592 (7853): 296-301
  • 31 Murphy AJ, Febbraio MA. Immune-based therapies in cardiovascular and metabolic diseases: past, present and future. Nat Rev Immunol 2021; 21 (10) 669-679
  • 32 Ridker PM, Everett BM, Thuren T. et al; CANTOS Trial Group. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med 2017; 377 (12) 1119-1131
  • 33 Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 2006; 440 (7081): 237-241
  • 34 Misawa T, Takahama M, Kozaki T. et al. Microtubule-driven spatial arrangement of mitochondria promotes activation of the NLRP3 inflammasome. Nat Immunol 2013; 14 (05) 454-460
  • 35 Tardif JC, Kouz S, Waters DD. et al. Efficacy and safety of low-dose colchicine after myocardial infarction. N Engl J Med 2019; 381 (26) 2497-2505
  • 36 Newby LK. Inflammation as a treatment target after acute myocardial infarction. N Engl J Med 2019; 381 (26) 2562-2563
  • 37 Svensson EC, Madar A, Campbell CD. et al. TET2-driven clonal hematopoiesis and response to canakinumab: an exploratory analysis of the CANTOS randomized clinical trial. JAMA Cardiol 2022; 7 (05) 521-528
  • 38 Interleukin-6 Receptor Mendelian Randomisation Analysis (IL6R MR) Consortium. Swerdlow DI, Holmes MV. et al. The interleukin-6 receptor as a target for prevention of coronary heart disease: a Mendelian randomisation analysis. Lancet 2012; 379: 1214-1224
  • 39 Cai T, Zhang Y, Ho YL. et al; VA Million Veteran Program. Association of interleukin 6 receptor variant with cardiovascular disease effects of interleukin 6 receptor blocking therapy: a phenome-wide association study. JAMA Cardiol 2018; 3 (09) 849-857
  • 40 Veninga A, De Simone I, Heemskerk JWM, Cate HT, van der Meijden PEJ. Clonal hematopoietic mutations linked to platelet traits and the risk of thrombosis or bleeding. Haematologica 2020; 105 (08) 2020-2031
  • 41 Panova-Noeva M, Marchetti M, Russo L. et al. ADP-induced platelet aggregation and thrombin generation are increased in Essential Thrombocythemia and Polycythemia Vera. Thromb Res 2013; 132 (01) 88-93
  • 42 Panova-Noeva M, Marchetti M, Spronk HM. et al. Platelet-induced thrombin generation by the calibrated automated thrombogram assay is increased in patients with essential thrombocythemia and polycythemia vera. Am J Hematol 2011; 86 (04) 337-342
  • 43 Spivak JL. Myeloproliferative neoplasms. N Engl J Med 2017; 376 (22) 2168-2181
  • 44 Wolach O, Sellar RS, Martinod K. et al. Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci Transl Med 2018; 10 (436) 10
  • 45 Verstovsek S, Silver RT, Cross NC, Tefferi A. JAK2V617F mutational frequency in polycythemia vera: 100%, >90%, less?. Leukemia 2006; 20 (11) 2067
  • 46 Kwaan HC, Wang J. Hyperviscosity in polycythemia vera and other red cell abnormalities. Semin Thromb Hemost 2003; 29 (05) 451-458
  • 47 Brinkmann V, Reichard U, Goosmann C. et al. Neutrophil extracellular traps kill bacteria. Science 2004; 303 (5663): 1532-1535
  • 48 Martinod K, Demers M, Fuchs TA. et al. Neutrophil histone modification by peptidylarginine deiminase 4 is critical for deep vein thrombosis in mice. Proc Natl Acad Sci U S A 2013; 110 (21) 8674-8679
  • 49 Borissoff JI, Joosen IA, Versteylen MO. et al. Elevated levels of circulating DNA and chromatin are independently associated with severe coronary atherosclerosis and a prothrombotic state. Arterioscler Thromb Vasc Biol 2013; 33 (08) 2032-2040
  • 50 Hurtado-Nedelec M, Csillag-Grange MJ, Boussetta T. et al. Increased reactive oxygen species production and p47phox phosphorylation in neutrophils from myeloproliferative disorders patients with JAK2 (V617F) mutation. Haematologica 2013; 98 (10) 1517-1524
  • 51 Kushnir M, Cohen HW, Billett HH. Persistent neutrophilia is a marker for an increased risk of venous thrombosis. J Thromb Thrombolysis 2016; 42 (04) 545-551
  • 52 Marin Oyarzún CP, Carestia A, Lev PR. et al. Neutrophil extracellular trap formation and circulating nucleosomes in patients with chronic myeloproliferative neoplasms. Sci Rep 2016; 6: 38738
  • 53 Hobbs CM, Manning H, Bennett C. et al. JAK2V617F leads to intrinsic changes in platelet formation and reactivity in a knock-in mouse model of essential thrombocythemia. Blood 2013; 122 (23) 3787-3797
  • 54 Lamrani L, Lacout C, Ollivier V. et al. Hemostatic disorders in a JAK2V617F-driven mouse model of myeloproliferative neoplasm. Blood 2014; 124 (07) 1136-1145
  • 55 Etheridge SL, Roh ME, Cosgrove ME. et al. JAK2V617F-positive endothelial cells contribute to clotting abnormalities in myeloproliferative neoplasms. Proc Natl Acad Sci U S A 2014; 111 (06) 2295-2300
  • 56 Bersenev A, Wu C, Balcerek J, Tong W. Lnk controls mouse hematopoietic stem cell self-renewal and quiescence through direct interactions with JAK2. J Clin Invest 2008; 118 (08) 2832-2844
  • 57 Wang W, Tang Y, Wang Y. et al. LNK/SH2B3 loss of function promotes atherosclerosis and thrombosis. Circ Res 2016; 119 (06) e91-e103
  • 58 Dou H, Kotini A, Liu W. et al. Oxidized phospholipids promote NETosis and arterial thrombosis in LNK(SH2B3) deficiency. Circulation 2021; 144 (24) 1940-1954
  • 59 Deloukas P, Kanoni S, Willenborg C. et al; CARDIoGRAMplusC4D Consortium, DIAGRAM Consortium, CARDIOGENICS Consortium, MuTHER Consortium, Wellcome Trust Case Control Consortium. Large-scale association analysis identifies new risk loci for coronary artery disease. Nat Genet 2013; 45 (01) 25-33
  • 60 Hinds DA, Barnholt KE, Mesa RA. et al. Germ line variants predispose to both JAK2 V617F clonal hematopoiesis and myeloproliferative neoplasms. Blood 2016; 128 (08) 1121-1128
  • 61 Cremer S, Kirschbaum K, Berkowitsch A. et al. Multiple somatic mutations for clonal hematopoiesis are associated with increased mortality in patients with chronic heart failure. Circ Genom Precis Med 2020; 13 (04) e003003
  • 62 Yu B, Roberts MB, Raffield LM. et al; National Heart, Lung, and Blood Institute TOPMed Consortium. Supplemental association of clonal hematopoiesis with incident heart failure. J Am Coll Cardiol 2021; 78 (01) 42-52
  • 63 Dharan NJ, Yeh P, Bloch M. et al; ARCHIVE Study Group. HIV is associated with an increased risk of age-related clonal hematopoiesis among older adults. Nat Med 2021; 27 (06) 1006-1011
  • 64 Heyde A, Rohde D, McAlpine CS. et al. Increased stem cell proliferation in atherosclerosis accelerates clonal hematopoiesis. Cell 2021; 184 (05) 1348-1361.e22
  • 65 van Deuren RC, Andersson-Assarsson JC, Kristensson FM. et al. Clone expansion of mutation-driven clonal hematopoiesis is associated with aging and metabolic dysfunction in individuals with obesity. bioRxiv 2021 2021.2005.2012.443095
  • 66 Castillo JJ, Mull N, Reagan JL, Nemr S, Mitri J. Increased incidence of non-Hodgkin lymphoma, leukemia, and myeloma in patients with diabetes mellitus type 2: a meta-analysis of observational studies. Blood 2012; 119 (21) 4845-4850
  • 67 Shu X, Ji J, Li X, Sundquist J, Sundquist K, Hemminki K. Cancer risk among patients hospitalized for Type 1 diabetes mellitus: a population-based cohort study in Sweden. Diabet Med 2010; 27 (07) 791-797
  • 68 Cai Z, Lu X, Zhang C. et al. Hyperglycemia cooperates with Tet2 heterozygosity to induce leukemia driven by proinflammatory cytokine-induced lncRNA Morrbid. J Clin Invest 2021; 131 (01) 131
  • 69 Fuster JJ, Zuriaga MA, Zorita V. et al. TET2-loss-of-function-driven clonal hematopoiesis exacerbates experimental insulin resistance in aging and obesity. Cell Rep 2020; 33 (04) 108326
  • 70 Bhattacharya R, Zekavat SM, Uddin MM. et al. Association of diet quality with prevalence of clonal hematopoiesis and adverse cardiovascular events. JAMA Cardiol 2021; 6 (09) 1069-1077
  • 71 Haring B, Reiner AP, Liu J. et al. Healthy lifestyle and clonal hematopoiesis of indeterminate potential: results from the women's health initiative. J Am Heart Assoc 2021; 10 (05) e018789
  • 72 Lee MKS, Dragoljevic D, Bertuzzo Veiga C, Wang N, Yvan-Charvet L, Murphy AJ. Interplay between clonal hematopoiesis of indeterminate potential and metabolism. Trends Endocrinol Metab 2020; 31 (07) 525-535
  • 73 Nagareddy PR, Kraakman M, Masters SL. et al. Adipose tissue macrophages promote myelopoiesis and monocytosis in obesity. Cell Metab 2014; 19 (05) 821-835
  • 74 Nagareddy PR, Murphy AJ, Stirzaker RA. et al. Hyperglycemia promotes myelopoiesis and impairs the resolution of atherosclerosis. Cell Metab 2013; 17 (05) 695-708
  • 75 Wu D, Hu D, Chen H. et al. Glucose-regulated phosphorylation of TET2 by AMPK reveals a pathway linking diabetes to cancer. Nature 2018; 559 (7715): 637-641
  • 76 Talpaz M, Kiladjian JJ. Fedratinib, a newly approved treatment for patients with myeloproliferative neoplasm-associated myelofibrosis. Leukemia 2021; 35 (01) 1-17
  • 77 Tang Y, Liu W, Wang W. et al. Inhibition of JAK2 suppresses myelopoiesis and atherosclerosis in Apoe-/- mice. Cardiovasc Drugs Ther 2020; 34 (02) 145-152
  • 78 McNeil JJ, Wolfe R, Woods RL. et al; ASPREE Investigator Group. Effect of aspirin on cardiovascular events and bleeding in the healthy elderly. N Engl J Med 2018; 379 (16) 1509-1518
  • 79 Alvarez-Larrán A, Pereira A, Guglielmelli P. et al. Antiplatelet therapy versus observation in low-risk essential thrombocythemia with a CALR mutation. Haematologica 2016; 101 (08) 926-931
  • 80 Marin Oyarzún CP, Heller PG. Platelets as mediators of thromboinflammation in chronic myeloproliferative neoplasms. Front Immunol 2019; 10: 1373
  • 81 Binder CJ, Papac-Milicevic N, Witztum JL. Innate sensing of oxidation-specific epitopes in health and disease. Nat Rev Immunol 2016; 16 (08) 485-497