Thromb Haemost 2022; 122(04): 476-479
DOI: 10.1055/s-0041-1736445
Invited Mini Series: Novel Clinical Concepts in Thrombosis

Human Immunodeficiency Virus and Cardiovascular Disease: Revisiting the Inflammation–Thrombosis Axis

Keir McCutcheon
1   Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
,
1   Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
› Institutsangaben

Introduction

There are more than 37 million people living with the human immunodeficiency virus (PLWH) worldwide with an estimated 2 million new infections occurring annually. Antiretroviral medication has dramatically prolonged the life expectancy of PLWH resulting in a growing population of older PLWH. Based on recent data from the Netherlands, by 2030, 73% of human immunodeficiency virus (HIV)-positive patients will be older than 50 years of age.[1] Despite this increasing life expectancy, there is a twofold increased risk of atherosclerotic cardiovascular disease (ASCVD) and a two- to 10-fold increased risk of venous thromboembolism (VTE). The reasons for the increased risk of cardiovascular disease among PLWH are complex and multifactorial. In addition to a high frequency of traditional risk factors in PLWH, persistent inflammation and coagulopathy appear to contribute to the increased risk of ASCVD and VTE.

HIV-1 infects CD4+ cells, mainly T helper cells, but also macrophages and dendritic cells, and directly or indirectly destroys these cells leading to immune dysfunction and deficiency.[2] HIV is able to activate pattern recognition receptors (PRRs) that promote inflammasome activation in circulating monocyte-derived dendritic cells, human monocytes, and monocyte-derived macrophages.[3] Although combined antiretroviral therapy (cART) suppresses circulating viral counts, there is no complete elimination of viral latency, chronic inflammation, or immune activation. This chronic inflammation in PLWH, despite cART with viral suppression, is probably related to increased traditional risk factors and proinflammatory lipids, certain antiretroviral combinations, low levels of viral replication, coinfection with other pathogens such as cytomegalovirus, and microbial translocation ([Fig. 1]).[4]

Zoom Image
Fig. 1 Bidirectional relationship between inflammation and thrombosis in HIV-infected patients. CRP, C-reactive protein; IL-1β, interleukin-1β; IL-6, interleukin 6; sCD163, soluble CD163; TNF-α, tumor necrosis factor-α; vWF, von Willebrand factor.

Contributing to the proinflammatory phenotype in PLWH is lipodystrophy and adipocyte tissue dysfunction.[5] Lipodystrophy (abnormal fat redistribution) is associated with metabolic derangements, such as dyslipidemia and insulin resistance, and is well described following cART initiation.[5] [6] It is not clear whether a particular antiretroviral therapy drug or class of drugs is responsible for the abnormal fat accumulation since lipodystrophy appears to occur to some degree with any combination, even in patients with no prior exposure to protease inhibitors.[7] [8] Adipocyte tissue dysfunction, which is caused by both the HIV infection and certain antiretroviral agents, promotes systemic inflammation through secretion of adipokines.[9] In addition, due to chronic viral load suppression with effective cART among PLWH, there is an increasing frequency of obesity, which promotes insulin resistance and chronic inflammation through adipocyte dysfunction and lipotoxicity.[10]



Publikationsverlauf

Eingereicht: 03. August 2021

Angenommen: 08. September 2021

Artikel online veröffentlicht:
24. Oktober 2021

© 2021. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Smit M, Brinkman K, Geerlings S. et al; ATHENA observational cohort. Future challenges for clinical care of an ageing population infected with HIV: a modelling study. Lancet Infect Dis 2015; 15 (07) 810-818
  • 2 Deeks SG, Overbaugh J, Phillips A, Buchbinder S. HIV infection. Nat Rev Dis Primers 2015; 1: 15035
  • 3 Leal VNC, Reis EC, Pontillo A. Inflammasome in HIV infection: lights and shadows. Mol Immunol 2020; 118: 9-18
  • 4 Vachiat A, McCutcheon K, Tsabedze N, Zachariah D, Manga P. HIV and ischemic heart disease. J Am Coll Cardiol 2017; 69 (01) 73-82
  • 5 McComsey GA, Moser C, Currier J. et al. Body composition changes after initiation of raltegravir or protease inhibitors: ACTG A5260s. Clin Infect Dis 2016; 62 (07) 853-862
  • 6 Worm SW, Friis-Møller N, Bruyand M. et al; D:A:D study group. High prevalence of the metabolic syndrome in HIV-infected patients: impact of different definitions of the metabolic syndrome. AIDS 2010; 24 (03) 427-435
  • 7 Mulligan K, Tai VW, Algren H. et al. Altered fat distribution in HIV-positive men on nucleoside analog reverse transcriptase inhibitor therapy. J Acquir Immune Defic Syndr 2001; 26 (05) 443-448
  • 8 Lake JE, McComsey GA, Hulgan T. et al. Switch to raltegravir from protease inhibitor or nonnucleoside reverse-transcriptase inhibitor does not reduce visceral fat in human immunodeficiency virus-infected women with central adiposity. Open Forum Infect Dis 2015; 2 (02) ofv059
  • 9 Bourgeois C, Gorwood J, Olivo A. et al. Contribution of adipose tissue to the chronic immune activation and inflammation associated with HIV infection and its treatment. Front Immunol 2021; 12: 670566
  • 10 Glesby MJ, Hanna DB, Hoover DR. et al. Abdominal fat depots and subclinical carotid artery atherosclerosis in women with and without HIV infection. J Acquir Immune Defic Syndr 2018; 77 (03) 308-316
  • 11 d'Alessandro E, Becker C, Bergmeier W. et al; Scientific Reviewer Committee. Thrombo-inflammation in cardiovascular disease: an expert consensus document from the third maastricht consensus conference on thrombosis. Thromb Haemost 2020; 120 (04) 538-564
  • 12 Verhamme P, Hoylaerts MF. The pivotal role of the endothelium in haemostasis and thrombosis. Acta Clin Belg 2006; 61 (05) 213-219
  • 13 Levi M, van der Poll T, Büller HR. Bidirectional relation between inflammation and coagulation. Circulation 2004; 109 (22) 2698-2704
  • 14 Davalos D, Akassoglou K. Fibrinogen as a key regulator of inflammation in disease. Semin Immunopathol 2012; 34 (01) 43-62
  • 15 Pretorius E. Platelets in HIV: a guardian of host defence or transient reservoir of the virus?. Front Immunol 2021; 12: 649465
  • 16 Funderburg NT, Lederman MM. Coagulation and morbidity in treated HIV infection. Thromb Res 2014; 133 (Suppl. 01) S21-S24
  • 17 Bahrami H, Budoff M, Haberlen SA. et al. Inflammatory markers associated with subclinical coronary artery disease: the multicenter AIDS cohort study. J Am Heart Assoc 2016; 5 (06) 5
  • 18 Fitch KV, Srinivasa S, Abbara S. et al. Noncalcified coronary atherosclerotic plaque and immune activation in HIV-infected women. J Infect Dis 2013; 208 (11) 1737-1746
  • 19 Funderburg NT, Zidar DA, Shive C. et al. Shared monocyte subset phenotypes in HIV-1 infection and in uninfected subjects with acute coronary syndrome. Blood 2012; 120 (23) 4599-4608
  • 20 Ford ES, Greenwald JH, Richterman AG. et al. Traditional risk factors and D-dimer predict incident cardiovascular disease events in chronic HIV infection. AIDS 2010; 24 (10) 1509-1517
  • 21 Musselwhite LW, Sheikh V, Norton TD. et al. Markers of endothelial dysfunction, coagulation and tissue fibrosis independently predict venous thromboembolism in HIV. AIDS 2011; 25 (06) 787-795
  • 22 Funderburg NT, Mayne E, Sieg SF. et al. Increased tissue factor expression on circulating monocytes in chronic HIV infection: relationship to in vivo coagulation and immune activation. Blood 2010; 115 (02) 161-167
  • 23 Jackson SP, Darbousset R, Schoenwaelder SM. Thromboinflammation: challenges of therapeutically targeting coagulation and other host defense mechanisms. Blood 2019; 133 (09) 906-918
  • 24 Torriani FJ, Komarow L, Parker RA. et al; ACTG 5152s Study Team. Endothelial function in human immunodeficiency virus-infected antiretroviral-naive subjects before and after starting potent antiretroviral therapy: The ACTG (AIDS Clinical Trials Group) Study 5152s. J Am Coll Cardiol 2008; 52 (07) 569-576
  • 25 Rethy L, Feinstein MJ, Sinha A, Achenbach C, Shah SJ. Coronary microvascular dysfunction in HIV: a review. J Am Heart Assoc 2020; 9 (01) e014018
  • 26 Mahtab S, Zar HJ, Ntusi NAB. et al. Endothelial dysfunction in South African youth living with perinatally acquired human immunodeficiency virus on antiretroviral therapy. Clin Infect Dis 2020; 71 (10) e672-e679
  • 27 Graham SM, Rajwans N, Jaoko W. et al. Endothelial activation biomarkers increase after HIV-1 acquisition: plasma vascular cell adhesion molecule-1 predicts disease progression. AIDS 2013; 27 (11) 1803-1813
  • 28 Vachiat A, Dix-Peek T, Duarte R, Manga P. Endothelial dysfunction in HIV-positive patients with acute coronary syndromes. Cardiovasc J Afr 2020; 31 (04) 58-64
  • 29 Konst RE, Guzik TJ, Kaski JC, Maas AHEM, Elias-Smale SE. The pathogenic role of coronary microvascular dysfunction in the setting of other cardiac or systemic conditions. Cardiovasc Res 2020; 116 (04) 817-828
  • 30 Hauguel-Moreau M, Boccara F, Boyd A. et al. Platelet reactivity in human immunodeficiency virus infected patients on dual antiplatelet therapy for an acute coronary syndrome: the EVERE2ST-HIV study. Eur Heart J 2017; 38 (21) 1676-1686
  • 31 Green SA, Smith M, Hasley RB. et al. Activated platelet-T-cell conjugates in peripheral blood of patients with HIV infection: coupling coagulation/inflammation and T cells. AIDS 2015; 29 (11) 1297-1308
  • 32 Baker JV, Huppler Hullsiek K, Bradford RL, Prosser R, Tracy RP, Key NS. Circulating levels of tissue factor microparticle procoagulant activity are reduced with antiretroviral therapy and are associated with persistent inflammation and coagulation activation among HIV-positive patients. J Acquir Immune Defic Syndr 2013; 63 (03) 367-371
  • 33 Titanji B, Gavegnano C, Hsue P, Schinazi R, Marconi VC. Targeting inflammation to reduce atherosclerotic cardiovascular risk in people with HIV infection. J Am Heart Assoc 2020; 9 (03) e014873
  • 34 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
  • 35 Hsue PY, Li D, Ma Y. et al. IL-1β inhibition reduces atherosclerotic inflammation in HIV infection. J Am Coll Cardiol 2018; 72 (22) 2809-2811
  • 36 Hsue PY, Ribaudo HJ, Deeks SG. et al. Safety and impact of low-dose methotrexate on endothelial function and inflammation in individuals with treated human immunodeficiency virus: AIDS Clinical Trials Group Study A5314. Clin Infect Dis 2019; 68 (11) 1877-1886
  • 37 O'Brien MP, Zafar MU, Rodriguez JC. et al. Targeting thrombogenicity and inflammation in chronic HIV infection. Sci Adv 2019; 5 (06) eaav5463
  • 38 Sandler NG, Zhang X, Bosch RJ. et al; AIDS Clinical Trials Group A5296 Team. Sevelamer does not decrease lipopolysaccharide or soluble CD14 levels but decreases soluble tissue factor, low-density lipoprotein (LDL) cholesterol, and oxidized LDL cholesterol levels in individuals with untreated HIV infection. J Infect Dis 2014; 210 (10) 1549-1554