Skip to main content
Erschienen in: Current HIV/AIDS Reports 1/2021

12.01.2021 | HIV Pathogenesis and Treatment (AL Landay and NS Utay, Section Editors)

Immune Dysregulation in Myocardial Fibrosis, Steatosis, and Heart Failure: Current Insights from HIV and the General Population

verfasst von: Arjun Sinha, Matthew J. Feinstein

Erschienen in: Current HIV/AIDS Reports | Ausgabe 1/2021

Einloggen, um Zugang zu erhalten

Abstract

Purpose of Review

HIV is an independent risk factor for heart
failure (HF). Cardiac imaging studies in people with HIV (PWH) have identified myocardial pathologies, namely fibrosis and steatosis, that likely contribute to the higher risk of HF. In this review, we survey existing epidemiological, clinical, and mechanistic literature to identify potential pathways that may contribute to the burden of myocardial fibrosis and steatosis among PWH.

Recent Findings

Multiple cohort studies over the past 20 years have demonstrated a roughly 2-fold higher risk of incident HF in PWH, as well as a disproportionate burden of myocardial fibrosis and steatosis in PWH without HF. Both myocardial fibrosis and steatosis are known contributors to HF in adults without HIV. Pathways involving the NLRP3 inflammasome, TGF-β1, and adipocyte dysfunction are known to play a crucial role in the development of myocardial fibrosis and steatosis. Upregulation of these pathways in HIV due to direct effects of viral proteins, persistent immune dysregulation, gut epithelial breakdown and dysbiosis, and toxicities from antiretroviral therapy may contribute to myocardial dysfunction in HIV. Understanding these pathways may lead to more precise diagnostic and therapeutic targets to curb HF in PWH.

Summary

During the past three decades, observational and mechanistic studies have provided important insights into risk factors and pathways that may contribute to the increased HF risk in PWH. Future work is needed to characterize these pathways more precisely in mechanistic studies of PWH, with the goal of ultimately deriving valuable targets for prevention, early diagnosis, and treatment of HF in PWH.
Literatur
1.
Zurück zum Zitat Cohen IS, Anderson DW, Virmani R, Reen BM, Macher AM, Sennesh J, et al. Congestive cardiomyopathy in association with the acquired immunodeficiency syndrome. N Engl J Med. 1986;315:628–30.PubMedCrossRef Cohen IS, Anderson DW, Virmani R, Reen BM, Macher AM, Sennesh J, et al. Congestive cardiomyopathy in association with the acquired immunodeficiency syndrome. N Engl J Med. 1986;315:628–30.PubMedCrossRef
2.
Zurück zum Zitat Calabrese LH, Proffitt MR, Yen-Lieberman B, Hobbs RE, Ratliff NB. Congestive cardiomyopathy and illness related to the acquired immunodeficiency syndrome (AIDS) associated with isolation of retrovirus from myocardium. Ann Intern Med. 1987;107:691–2.PubMedCrossRef Calabrese LH, Proffitt MR, Yen-Lieberman B, Hobbs RE, Ratliff NB. Congestive cardiomyopathy and illness related to the acquired immunodeficiency syndrome (AIDS) associated with isolation of retrovirus from myocardium. Ann Intern Med. 1987;107:691–2.PubMedCrossRef
3.
Zurück zum Zitat Acierno LJ. Cardiac complications in acquired immunodeficiency syndrome (AIDS): a review. J Am Coll Cardiol. 1989;13:1144–54.PubMedCrossRef Acierno LJ. Cardiac complications in acquired immunodeficiency syndrome (AIDS): a review. J Am Coll Cardiol. 1989;13:1144–54.PubMedCrossRef
4.
Zurück zum Zitat Herskowitz A, Willoughby SB, Baughman KL, Schulman SP, Bartlett JD. Cardiomyopathy associated with antiretroviral therapy in patients with HIV infection: a report of six cases. Ann Intern Med. 1992;116:311–3.PubMedCrossRef Herskowitz A, Willoughby SB, Baughman KL, Schulman SP, Bartlett JD. Cardiomyopathy associated with antiretroviral therapy in patients with HIV infection: a report of six cases. Ann Intern Med. 1992;116:311–3.PubMedCrossRef
6.
Zurück zum Zitat Lederman MM, Funderburg NT, Sekaly RP, Klatt NR, Hunt PW. Residual immune dysregulation syndrome in treated HIV infection. Adv Immunol. 2013;119:51–83.PubMedPubMedCentralCrossRef Lederman MM, Funderburg NT, Sekaly RP, Klatt NR, Hunt PW. Residual immune dysregulation syndrome in treated HIV infection. Adv Immunol. 2013;119:51–83.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Adamo L, Rocha-Resende C, Prabhu SD, Mann DL. Reappraising the role of inflammation in heart failure. Nat Rev Cardiol. 2020;17:269–85.PubMedCrossRef Adamo L, Rocha-Resende C, Prabhu SD, Mann DL. Reappraising the role of inflammation in heart failure. Nat Rev Cardiol. 2020;17:269–85.PubMedCrossRef
9.
Zurück zum Zitat Zhang Y, Bauersachs J, Langer HF. Immune mechanisms in heart failure. Eur J Heart Fail. 2017;19:1379–89.PubMedCrossRef Zhang Y, Bauersachs J, Langer HF. Immune mechanisms in heart failure. Eur J Heart Fail. 2017;19:1379–89.PubMedCrossRef
10.
Zurück zum Zitat Butt AA, Chang CC, Kuller L, et al. Risk of heart failure with human immunodeficiency virus in the absence of prior diagnosis of coronary heart disease. Arch Intern Med. 2011;171:737–43.PubMedPubMedCentral Butt AA, Chang CC, Kuller L, et al. Risk of heart failure with human immunodeficiency virus in the absence of prior diagnosis of coronary heart disease. Arch Intern Med. 2011;171:737–43.PubMedPubMedCentral
11.•
Zurück zum Zitat Freiberg MS, Chang CH, Skanderson M, et al. Association between HIV infection and the risk of heart failure with reduced ejection fraction and preserved ejection fraction in the antiretroviral therapy era: results from the Veterans Aging Cohort Study. JAMA Cardiol. 2017;2:536–46 This study extracted left ventricular ejection fractions from the electronic health records to determine the association of HIV infection with different HF subtypes, specifically HFrEF, HFpEF, and HFmrEF.PubMedPubMedCentralCrossRef Freiberg MS, Chang CH, Skanderson M, et al. Association between HIV infection and the risk of heart failure with reduced ejection fraction and preserved ejection fraction in the antiretroviral therapy era: results from the Veterans Aging Cohort Study. JAMA Cardiol. 2017;2:536–46 This study extracted left ventricular ejection fractions from the electronic health records to determine the association of HIV infection with different HF subtypes, specifically HFrEF, HFpEF, and HFmrEF.PubMedPubMedCentralCrossRef
13.•
Zurück zum Zitat Feinstein MJ, Steverson AB, Ning H, et al. Adjudicated Heart Failure in HIV-Infected and Uninfected Men and Women. J Am Heart Assoc. 2018;7:e009985 This is the only study to date to have evaluated the association of HIV infection with HF, where the HF diagnsoses were underwent physicain adjudication.PubMedPubMedCentralCrossRef Feinstein MJ, Steverson AB, Ning H, et al. Adjudicated Heart Failure in HIV-Infected and Uninfected Men and Women. J Am Heart Assoc. 2018;7:e009985 This is the only study to date to have evaluated the association of HIV infection with HF, where the HF diagnsoses were underwent physicain adjudication.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Yen YF, Ko MC, Yen MY, Hu BS, Wang TH, Chuang PH, et al. Human immunodeficiency virus increases the risk of incident heart failure. J Acquir Immune Defic Syndr. 2019;80:255–63.PubMedCrossRef Yen YF, Ko MC, Yen MY, Hu BS, Wang TH, Chuang PH, et al. Human immunodeficiency virus increases the risk of incident heart failure. J Acquir Immune Defic Syndr. 2019;80:255–63.PubMedCrossRef
15.
Zurück zum Zitat Sax PE, Erlandson KM, Lake JE, et al. Weight gain following initiation of antiretroviral therapy: risk factors in randomized comparative clinical trials. Clin Infect Dis. 2020;71:1379–89. Sax PE, Erlandson KM, Lake JE, et al. Weight gain following initiation of antiretroviral therapy: risk factors in randomized comparative clinical trials. Clin Infect Dis. 2020;71:1379–89.
16.
Zurück zum Zitat El-Sadr WM, Lundgren J, Neaton JD, et al. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med. 2006;355:2283–96.PubMedCrossRef El-Sadr WM, Lundgren J, Neaton JD, et al. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med. 2006;355:2283–96.PubMedCrossRef
17.
Zurück zum Zitat Lundgren JD, Babiker AG, Gordin F, et al. Initiation of antiretroviral therapy in early asymptomatic HIV infection. N Engl J Med. 2015;373:795–807.PubMedCrossRef Lundgren JD, Babiker AG, Gordin F, et al. Initiation of antiretroviral therapy in early asymptomatic HIV infection. N Engl J Med. 2015;373:795–807.PubMedCrossRef
18.
Zurück zum Zitat Saag MS, Benson CA, Gandhi RT, Hoy JF, Landovitz RJ, Mugavero MJ, et al. Antiretroviral drugs for treatment and prevention of HIV infection in adults: 2018 recommendations of the International Antiviral Society-USA Panel. Jama. 2018;320:379–96.PubMedPubMedCentralCrossRef Saag MS, Benson CA, Gandhi RT, Hoy JF, Landovitz RJ, Mugavero MJ, et al. Antiretroviral drugs for treatment and prevention of HIV infection in adults: 2018 recommendations of the International Antiviral Society-USA Panel. Jama. 2018;320:379–96.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Erqou S, Lodebo BT, Masri A, Altibi AM, Echouffo-Tcheugui JB, Dzudie A, et al. Cardiac dysfunction among people living with HIV: a systematic review and meta-analysis. JACC Heart Fail. 2019;7:98–108.PubMedCrossRef Erqou S, Lodebo BT, Masri A, Altibi AM, Echouffo-Tcheugui JB, Dzudie A, et al. Cardiac dysfunction among people living with HIV: a systematic review and meta-analysis. JACC Heart Fail. 2019;7:98–108.PubMedCrossRef
20.
Zurück zum Zitat Sinha A, Mystakelis H, Rivera AS, et al. Association of low CD4/CD8 ratio with adverse cardiac mechanics in lymphopenic HIV-infected adults. J Acquir Immune Defic Syndr. 2020;85:e73-e76. Sinha A, Mystakelis H, Rivera AS, et al. Association of low CD4/CD8 ratio with adverse cardiac mechanics in lymphopenic HIV-infected adults. J Acquir Immune Defic Syndr. 2020;85:e73-e76.
21.•
Zurück zum Zitat Holloway CJ, Ntusi N, Suttie J, et al. Comprehensive cardiac magnetic resonance imaging and spectroscopy reveal a high burden of myocardial disease in HIV patients. Circulation. 2013;128:814–22 This study used MRI/MRS to demonstrate that HIV patients without clinical cardiovascular disease had significantly greater myocardial fibrosis and steatosis than uninfected controls.PubMedCrossRef Holloway CJ, Ntusi N, Suttie J, et al. Comprehensive cardiac magnetic resonance imaging and spectroscopy reveal a high burden of myocardial disease in HIV patients. Circulation. 2013;128:814–22 This study used MRI/MRS to demonstrate that HIV patients without clinical cardiovascular disease had significantly greater myocardial fibrosis and steatosis than uninfected controls.PubMedCrossRef
22.••
Zurück zum Zitat Thiara DK, Liu CY, Raman F, et al. Abnormal myocardial function is related to myocardial Steatosis and diffuse myocardial fibrosis in HIV-infected adults. J Infect Dis. 2015;212:1544–51 This study used MRI/MRS to further corroborate that HIV patients had a higher burden of myocardial fibrosis and steatosis compared with uninfected controls and that LV function was negatively associated with inflammatory markers.PubMedPubMedCentralCrossRef Thiara DK, Liu CY, Raman F, et al. Abnormal myocardial function is related to myocardial Steatosis and diffuse myocardial fibrosis in HIV-infected adults. J Infect Dis. 2015;212:1544–51 This study used MRI/MRS to further corroborate that HIV patients had a higher burden of myocardial fibrosis and steatosis compared with uninfected controls and that LV function was negatively associated with inflammatory markers.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Feinstein MJ, Mitter SS, Yadlapati A, Achenbach CJ, Palella FJ Jr, Engel Gonzalez P, et al. HIV-related myocardial vulnerability to infarction and coronary artery disease. J Am Coll Cardiol. 2016;68:2026–7.PubMedPubMedCentralCrossRef Feinstein MJ, Mitter SS, Yadlapati A, Achenbach CJ, Palella FJ Jr, Engel Gonzalez P, et al. HIV-related myocardial vulnerability to infarction and coronary artery disease. J Am Coll Cardiol. 2016;68:2026–7.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Nelson MD, Szczepaniak LS, LaBounty TM, et al. Cardiac steatosis and left ventricular dysfunction in HIV-infected patients treated with highly active antiretroviral therapy. JACC Cardiovasc Imaging. 2014;7:1175–7.PubMedPubMedCentralCrossRef Nelson MD, Szczepaniak LS, LaBounty TM, et al. Cardiac steatosis and left ventricular dysfunction in HIV-infected patients treated with highly active antiretroviral therapy. JACC Cardiovasc Imaging. 2014;7:1175–7.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat González A, Schelbert EB, Díez J, Butler J. Myocardial interstitial fibrosis in heart failure: biological and translational perspectives. J Am Coll Cardiol. 2018;71:1696–706.PubMedCrossRef González A, Schelbert EB, Díez J, Butler J. Myocardial interstitial fibrosis in heart failure: biological and translational perspectives. J Am Coll Cardiol. 2018;71:1696–706.PubMedCrossRef
26.
Zurück zum Zitat Weber KT, Sun Y, Bhattacharya SK, Ahokas RA, Gerling IC. Myofibroblast-mediated mechanisms of pathological remodelling of the heart. Nat Rev Cardiol. 2013;10:15–26.PubMedCrossRef Weber KT, Sun Y, Bhattacharya SK, Ahokas RA, Gerling IC. Myofibroblast-mediated mechanisms of pathological remodelling of the heart. Nat Rev Cardiol. 2013;10:15–26.PubMedCrossRef
28.
Zurück zum Zitat Sun Y, Weber KT. Angiotensin converting enzyme and myofibroblasts during tissue repair in the rat heart. J Mol Cell Cardiol. 1996;28:851–8.PubMedCrossRef Sun Y, Weber KT. Angiotensin converting enzyme and myofibroblasts during tissue repair in the rat heart. J Mol Cell Cardiol. 1996;28:851–8.PubMedCrossRef
29.
Zurück zum Zitat Dobaczewski M, Bujak M, Li N, Gonzalez-Quesada C, Mendoza LH, Wang XF, et al. Smad3 signaling critically regulates fibroblast phenotype and function in healing myocardial infarction. Circ Res. 2010;107:418–28.PubMedPubMedCentralCrossRef Dobaczewski M, Bujak M, Li N, Gonzalez-Quesada C, Mendoza LH, Wang XF, et al. Smad3 signaling critically regulates fibroblast phenotype and function in healing myocardial infarction. Circ Res. 2010;107:418–28.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Schelbert EB, Piehler KM, Zareba KM, Moon JC, Ugander M, Messroghli DR, et al. Myocardial fibrosis quantified by extracellular volume is associated with subsequent hospitalization for heart failure, death, or both across the Spectrum of ejection fraction and heart failure stage. J Am Heart Assoc. 2015;4:e002613. Schelbert EB, Piehler KM, Zareba KM, Moon JC, Ugander M, Messroghli DR, et al. Myocardial fibrosis quantified by extracellular volume is associated with subsequent hospitalization for heart failure, death, or both across the Spectrum of ejection fraction and heart failure stage. J Am Heart Assoc. 2015;4:e002613.
31.
Zurück zum Zitat Schelbert EB, Fridman Y, Wong TC, Abu Daya H, Piehler KM, Kadakkal A, et al. Temporal relation between myocardial fibrosis and heart failure with preserved ejection fraction: association with baseline disease severity and subsequent outcome. JAMA Cardiol. 2017;2:995–1006.PubMedPubMedCentralCrossRef Schelbert EB, Fridman Y, Wong TC, Abu Daya H, Piehler KM, Kadakkal A, et al. Temporal relation between myocardial fibrosis and heart failure with preserved ejection fraction: association with baseline disease severity and subsequent outcome. JAMA Cardiol. 2017;2:995–1006.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Zheng D, Liwinski T, Elinav E. Inflammasome activation and regulation: toward a better understanding of complex mechanisms. Cell Discovery. 2020;6:36.PubMedPubMedCentralCrossRef Zheng D, Liwinski T, Elinav E. Inflammasome activation and regulation: toward a better understanding of complex mechanisms. Cell Discovery. 2020;6:36.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Mullis C, Swartz TH. NLRP3 Inflammasome signaling as a link between HIV-1 infection and atherosclerotic cardiovascular disease. Front Cardiovasc Med. 2020;7:95. Mullis C, Swartz TH. NLRP3 Inflammasome signaling as a link between HIV-1 infection and atherosclerotic cardiovascular disease. Front Cardiovasc Med. 2020;7:95.
34.
Zurück zum Zitat Bandera A, Masetti M, Fabbiani M, Biasin M, Muscatello A, Squillace N, et al. The NLRP3 Inflammasome is upregulated in HIV-infected antiretroviral therapy-treated individuals with defective immune recovery. Front Immunol. 2018;9:214.PubMedPubMedCentralCrossRef Bandera A, Masetti M, Fabbiani M, Biasin M, Muscatello A, Squillace N, et al. The NLRP3 Inflammasome is upregulated in HIV-infected antiretroviral therapy-treated individuals with defective immune recovery. Front Immunol. 2018;9:214.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Song J, Jiao Y, Zhang T, Zhang Y, Huang X, Li H, et al. Longitudinal changes in plasma Caspase-1 and Caspase-3 during the first 2 years of HIV-1 infection in CD4Low and CD4High patient groups. PLoS One. 2015;10:e0121011.PubMedPubMedCentralCrossRef Song J, Jiao Y, Zhang T, Zhang Y, Huang X, Li H, et al. Longitudinal changes in plasma Caspase-1 and Caspase-3 during the first 2 years of HIV-1 infection in CD4Low and CD4High patient groups. PLoS One. 2015;10:e0121011.PubMedPubMedCentralCrossRef
36.•
Zurück zum Zitat Chattergoon MA, Latanich R, Quinn J, et al. HIV and HCV activate the inflammasome in monocytes and macrophages via endosomal Toll-like receptors without induction of type 1 interferon. PLoS Pathog. 2014;10:e1004082 This in vitro study demonstrated that HIV could activate the NLRP3 inflammasome in monocytes and macrophages via an infection-independent process.PubMedPubMedCentralCrossRef Chattergoon MA, Latanich R, Quinn J, et al. HIV and HCV activate the inflammasome in monocytes and macrophages via endosomal Toll-like receptors without induction of type 1 interferon. PLoS Pathog. 2014;10:e1004082 This in vitro study demonstrated that HIV could activate the NLRP3 inflammasome in monocytes and macrophages via an infection-independent process.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Guo H, Gao J, Taxman DJ, Ting JP, Su L. HIV-1 infection induces interleukin-1β production via TLR8 protein-dependent and NLRP3 inflammasome mechanisms in human monocytes. J Biol Chem. 2014;289:21716–26.PubMedPubMedCentralCrossRef Guo H, Gao J, Taxman DJ, Ting JP, Su L. HIV-1 infection induces interleukin-1β production via TLR8 protein-dependent and NLRP3 inflammasome mechanisms in human monocytes. J Biol Chem. 2014;289:21716–26.PubMedPubMedCentralCrossRef
38.
39.
Zurück zum Zitat Galloway NL, Doitsh G, Monroe KM, et al. Cell-to-cell transmission of HIV-1 is required to trigger pyroptotic death of lymphoid-tissue-derived CD4 T cells. Cell Rep. 2015;12:1555–63.PubMedPubMedCentralCrossRef Galloway NL, Doitsh G, Monroe KM, et al. Cell-to-cell transmission of HIV-1 is required to trigger pyroptotic death of lymphoid-tissue-derived CD4 T cells. Cell Rep. 2015;12:1555–63.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Hunt PW, Sinclair E, Rodriguez B, Shive C, Clagett B, Funderburg N, et al. Gut epithelial barrier dysfunction and innate immune activation predict mortality in treated HIV infection. J Infect Dis. 2014;210:1228–38.PubMedPubMedCentralCrossRef Hunt PW, Sinclair E, Rodriguez B, Shive C, Clagett B, Funderburg N, et al. Gut epithelial barrier dysfunction and innate immune activation predict mortality in treated HIV infection. J Infect Dis. 2014;210:1228–38.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Assimakopoulos SF, Dimitropoulou D, Marangos M, Gogos CA. Intestinal barrier dysfunction in HIV infection: pathophysiology, clinical implications and potential therapies. Infection. 2014;42:951–9.PubMedCrossRef Assimakopoulos SF, Dimitropoulou D, Marangos M, Gogos CA. Intestinal barrier dysfunction in HIV infection: pathophysiology, clinical implications and potential therapies. Infection. 2014;42:951–9.PubMedCrossRef
43.
Zurück zum Zitat Sankaran S, George MD, Reay E, Guadalupe M, Flamm J, Prindiville T, et al. Rapid onset of intestinal epithelial barrier dysfunction in primary human immunodeficiency virus infection is driven by an imbalance between immune response and mucosal repair and regeneration. J Virol. 2008;82:538–45.PubMedCrossRef Sankaran S, George MD, Reay E, Guadalupe M, Flamm J, Prindiville T, et al. Rapid onset of intestinal epithelial barrier dysfunction in primary human immunodeficiency virus infection is driven by an imbalance between immune response and mucosal repair and regeneration. J Virol. 2008;82:538–45.PubMedCrossRef
44.
Zurück zum Zitat Ribeiro A, Heimesaat MM, Bereswill S. Changes of the intestinal microbiome-host homeostasis in HIV-infected individuals - a focus on the bacterial gut microbiome. Eur J Microbiol Immunol (Bp). 2017;7:158–67.CrossRef Ribeiro A, Heimesaat MM, Bereswill S. Changes of the intestinal microbiome-host homeostasis in HIV-infected individuals - a focus on the bacterial gut microbiome. Eur J Microbiol Immunol (Bp). 2017;7:158–67.CrossRef
45.
Zurück zum Zitat Vujkovic-Cvijin I, Somsouk M. HIV and the gut microbiota: composition, consequences, and avenues for amelioration. Curr HIV/AIDS Rep. 2019;16:204–13.PubMedPubMedCentralCrossRef Vujkovic-Cvijin I, Somsouk M. HIV and the gut microbiota: composition, consequences, and avenues for amelioration. Curr HIV/AIDS Rep. 2019;16:204–13.PubMedPubMedCentralCrossRef
46.•
Zurück zum Zitat Colaco NA, Ma Y, Scherzer R, et al. Abstract 15318: transmethylamine-N-oxide (TMAO) is associated with diffuse cardiac fibrosis in persons living with HIV. Circulation. 2019;140:A15318-A15318 This study showed a strong association between TMAO, a metabolite produced by gut bacteria, and myocardial fibrosis in HIV, thus providing a link between gut microbiota dysbiosis and heart failure in this population. Colaco NA, Ma Y, Scherzer R, et al. Abstract 15318: transmethylamine-N-oxide (TMAO) is associated with diffuse cardiac fibrosis in persons living with HIV. Circulation. 2019;140:A15318-A15318 This study showed a strong association between TMAO, a metabolite produced by gut bacteria, and myocardial fibrosis in HIV, thus providing a link between gut microbiota dysbiosis and heart failure in this population.
47.
Zurück zum Zitat Li X, Geng J, Zhao J, Ni Q, Zhao C, Zheng Y, et al. Trimethylamine N-oxide exacerbates cardiac fibrosis via activating the NLRP3 inflammasome. Front Physiol. 2019;10:866.PubMedPubMedCentralCrossRef Li X, Geng J, Zhao J, Ni Q, Zhao C, Zheng Y, et al. Trimethylamine N-oxide exacerbates cardiac fibrosis via activating the NLRP3 inflammasome. Front Physiol. 2019;10:866.PubMedPubMedCentralCrossRef
48.••
Zurück zum Zitat Hsue PY, Li D, Ma Y, et al. IL-1β inhibition reduces atherosclerotic inflammation in HIV infection. J Am Coll Cardiol. 2018;72:2809–11 This study showed that IL-1β inhibition reduced vascular inflammation in HIV.PubMedPubMedCentralCrossRef Hsue PY, Li D, Ma Y, et al. IL-1β inhibition reduces atherosclerotic inflammation in HIV infection. J Am Coll Cardiol. 2018;72:2809–11 This study showed that IL-1β inhibition reduced vascular inflammation in HIV.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Allen JB, Wong HL, Guyre PM, Simon GL, Wahl SM. Association of circulating receptor Fc gamma RIII-positive monocytes in AIDS patients with elevated levels of transforming growth factor-beta. J Clin Invest. 1991;87:1773–9.PubMedPubMedCentralCrossRef Allen JB, Wong HL, Guyre PM, Simon GL, Wahl SM. Association of circulating receptor Fc gamma RIII-positive monocytes in AIDS patients with elevated levels of transforming growth factor-beta. J Clin Invest. 1991;87:1773–9.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Reinhold D, Wrenger S, Kähne T, Ansorge S. HIV-1 Tat: immunosuppression via TGF-beta1 induction. Immunol Today. 1999;20:384–5.PubMedCrossRef Reinhold D, Wrenger S, Kähne T, Ansorge S. HIV-1 Tat: immunosuppression via TGF-beta1 induction. Immunol Today. 1999;20:384–5.PubMedCrossRef
51.
Zurück zum Zitat Hu R, Oyaizu N, Than S, Kalyanaraman VS, Wang XP, Pahwa S. HIV-1 gp160 induces transforming growth factor-beta production in human PBMC. Clin Immunol Immunopathol. 1996;80:283–9.PubMedCrossRef Hu R, Oyaizu N, Than S, Kalyanaraman VS, Wang XP, Pahwa S. HIV-1 gp160 induces transforming growth factor-beta production in human PBMC. Clin Immunol Immunopathol. 1996;80:283–9.PubMedCrossRef
52.
Zurück zum Zitat Laurence J, Elhadad S, Robison T, Terry H, Varshney R, Woolington S, et al. HIV protease inhibitor-induced cardiac dysfunction and fibrosis is mediated by platelet-derived TGF-β1 and can be suppressed by exogenous carbon monoxide. PLoS One. 2017;12:e0187185.PubMedPubMedCentralCrossRef Laurence J, Elhadad S, Robison T, Terry H, Varshney R, Woolington S, et al. HIV protease inhibitor-induced cardiac dysfunction and fibrosis is mediated by platelet-derived TGF-β1 and can be suppressed by exogenous carbon monoxide. PLoS One. 2017;12:e0187185.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Sakaguchi S. Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol. 2004;22:531–62.PubMedCrossRef Sakaguchi S. Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol. 2004;22:531–62.PubMedCrossRef
54.
Zurück zum Zitat Phetsouphanh C, Xu Y, Zaunders J. CD4 T cells mediate both positive and negative regulation of the immune response to HIV infection: complex role of T follicular helper cells and regulatory T cells in pathogenesis. Front Immunol. 2015;5:681. Phetsouphanh C, Xu Y, Zaunders J. CD4 T cells mediate both positive and negative regulation of the immune response to HIV infection: complex role of T follicular helper cells and regulatory T cells in pathogenesis. Front Immunol. 2015;5:681.
55.
Zurück zum Zitat Nilsson J, Boasso A, Velilla PA, Zhang R, Vaccari M, Franchini G, et al. HIV-1-driven regulatory T-cell accumulation in lymphoid tissues is associated with disease progression in HIV/AIDS. Blood. 2006;108:3808–17.PubMedPubMedCentralCrossRef Nilsson J, Boasso A, Velilla PA, Zhang R, Vaccari M, Franchini G, et al. HIV-1-driven regulatory T-cell accumulation in lymphoid tissues is associated with disease progression in HIV/AIDS. Blood. 2006;108:3808–17.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Maina EK, Abana CZ, Bukusi EA, Sedegah M, Lartey M, Ampofo WK. Plasma concentrations of transforming growth factor beta 1 in non-progressive HIV-1 infection correlates with markers of disease progression. Cytokine. 2016;81:109–16.PubMedCrossRef Maina EK, Abana CZ, Bukusi EA, Sedegah M, Lartey M, Ampofo WK. Plasma concentrations of transforming growth factor beta 1 in non-progressive HIV-1 infection correlates with markers of disease progression. Cytokine. 2016;81:109–16.PubMedCrossRef
57.
Zurück zum Zitat Theron AJ, Anderson R, Rossouw TM, Steel HC. The role of transforming growth factor beta-1 in the progression of HIV/AIDS and development of non-AIDS-defining fibrotic disorders. Front Immunol. 2017;8:1461.PubMedPubMedCentralCrossRef Theron AJ, Anderson R, Rossouw TM, Steel HC. The role of transforming growth factor beta-1 in the progression of HIV/AIDS and development of non-AIDS-defining fibrotic disorders. Front Immunol. 2017;8:1461.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Chiu HC, Kovacs A, Ford DA, Hsu FF, Garcia R, Herrero P, et al. A novel mouse model of lipotoxic cardiomyopathy. J Clin Invest. 2001;107:813–22.PubMedPubMedCentralCrossRef Chiu HC, Kovacs A, Ford DA, Hsu FF, Garcia R, Herrero P, et al. A novel mouse model of lipotoxic cardiomyopathy. J Clin Invest. 2001;107:813–22.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Zhou YT, Grayburn P, Karim A, Shimabukuro M, Higa M, Baetens D, et al. Lipotoxic heart disease in obese rats: implications for human obesity. Proc Natl Acad Sci U S A. 2000;97:1784–9.PubMedPubMedCentralCrossRef Zhou YT, Grayburn P, Karim A, Shimabukuro M, Higa M, Baetens D, et al. Lipotoxic heart disease in obese rats: implications for human obesity. Proc Natl Acad Sci U S A. 2000;97:1784–9.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Atkinson LL, Kozak R, Kelly SE, Onay Besikci A, Russell JC, Lopaschuk GD. Potential mechanisms and consequences of cardiac triacylglycerol accumulation in insulin-resistant rats. Am J Physiol Endocrinol Metab. 2003;284:E923–30.PubMedCrossRef Atkinson LL, Kozak R, Kelly SE, Onay Besikci A, Russell JC, Lopaschuk GD. Potential mechanisms and consequences of cardiac triacylglycerol accumulation in insulin-resistant rats. Am J Physiol Endocrinol Metab. 2003;284:E923–30.PubMedCrossRef
62.
Zurück zum Zitat McGavock JM, Lingvay I, Zib I, et al. Cardiac steatosis in diabetes mellitus: a 1H-magnetic resonance spectroscopy study. Circulation. 2007;116:1170–5.PubMedCrossRef McGavock JM, Lingvay I, Zib I, et al. Cardiac steatosis in diabetes mellitus: a 1H-magnetic resonance spectroscopy study. Circulation. 2007;116:1170–5.PubMedCrossRef
63.
Zurück zum Zitat Rijzewijk LJ, van der Meer RW, Smit JW, et al. Myocardial steatosis is an independent predictor of diastolic dysfunction in type 2 diabetes mellitus. J Am Coll Cardiol. 2008;52:1793–9.PubMedCrossRef Rijzewijk LJ, van der Meer RW, Smit JW, et al. Myocardial steatosis is an independent predictor of diastolic dysfunction in type 2 diabetes mellitus. J Am Coll Cardiol. 2008;52:1793–9.PubMedCrossRef
64.
Zurück zum Zitat Korosoglou G, Humpert PM, Ahrens J, Oikonomou D, Osman NF, Gitsioudis G, et al. Left ventricular diastolic function in type 2 diabetes mellitus is associated with myocardial triglyceride content but not with impaired myocardial perfusion reserve. J Magn Reson Imaging. 2012;35:804–11.PubMedCrossRef Korosoglou G, Humpert PM, Ahrens J, Oikonomou D, Osman NF, Gitsioudis G, et al. Left ventricular diastolic function in type 2 diabetes mellitus is associated with myocardial triglyceride content but not with impaired myocardial perfusion reserve. J Magn Reson Imaging. 2012;35:804–11.PubMedCrossRef
65.
Zurück zum Zitat Wei J, Nelson MD, Szczepaniak EW, Smith L, Mehta PK, Thomson LEJ, et al. Myocardial steatosis as a possible mechanistic link between diastolic dysfunction and coronary microvascular dysfunction in women. Am J Physiol Heart Circ Physiol. 2016;310:H14–9.PubMedCrossRef Wei J, Nelson MD, Szczepaniak EW, Smith L, Mehta PK, Thomson LEJ, et al. Myocardial steatosis as a possible mechanistic link between diastolic dysfunction and coronary microvascular dysfunction in women. Am J Physiol Heart Circ Physiol. 2016;310:H14–9.PubMedCrossRef
66.
Zurück zum Zitat Tsiodras S, Mantzoros C, Hammer S, Samore M. Effects of protease inhibitors on hyperglycemia, hyperlipidemia, and lipodystrophy: a 5-year cohort study. Arch Intern Med. 2000;160:2050–6.PubMedCrossRef Tsiodras S, Mantzoros C, Hammer S, Samore M. Effects of protease inhibitors on hyperglycemia, hyperlipidemia, and lipodystrophy: a 5-year cohort study. Arch Intern Med. 2000;160:2050–6.PubMedCrossRef
67.
Zurück zum Zitat Carr A, Samaras K, Thorisdottir A, Kaufmann GR, Chisholm DJ, Cooper DA. Diagnosis, prediction, and natural course of HIV-1 protease-inhibitor-associated lipodystrophy, hyperlipidaemia, and diabetes mellitus: a cohort study. Lancet. 1999;353:2093–9.PubMedCrossRef Carr A, Samaras K, Thorisdottir A, Kaufmann GR, Chisholm DJ, Cooper DA. Diagnosis, prediction, and natural course of HIV-1 protease-inhibitor-associated lipodystrophy, hyperlipidaemia, and diabetes mellitus: a cohort study. Lancet. 1999;353:2093–9.PubMedCrossRef
68.
Zurück zum Zitat Caron M, Auclairt M, Vissian A, Vigouroux C, Capeau J. Contribution of mitochondrial dysfunction and oxidative stress to cellular premature senescence induced by antiretroviral thymidine analogues. Antivir Ther. 2008;13:27–38.PubMedCrossRef Caron M, Auclairt M, Vissian A, Vigouroux C, Capeau J. Contribution of mitochondrial dysfunction and oxidative stress to cellular premature senescence induced by antiretroviral thymidine analogues. Antivir Ther. 2008;13:27–38.PubMedCrossRef
69.
Zurück zum Zitat McComsey GA, Moser C, Currier J, et al. Body composition changes after initiation of raltegravir or protease inhibitors: ACTG A5260s. Clin Infect Dis. 2016;62:853–62.PubMedPubMedCentralCrossRef McComsey GA, Moser C, Currier J, et al. Body composition changes after initiation of raltegravir or protease inhibitors: ACTG A5260s. Clin Infect Dis. 2016;62:853–62.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Godfrey C, Bremer A, Alba D, Apovian C, Koethe JR, Koliwad S, et al. Obesity and fat metabolism in human immunodeficiency virus–infected individuals: immunopathogenic mechanisms and clinical implications. J Infect Dis. 2019;220:420–31.PubMedPubMedCentralCrossRef Godfrey C, Bremer A, Alba D, Apovian C, Koethe JR, Koliwad S, et al. Obesity and fat metabolism in human immunodeficiency virus–infected individuals: immunopathogenic mechanisms and clinical implications. J Infect Dis. 2019;220:420–31.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Mori MA, Thomou T, Boucher J, Lee KY, Lallukka S, Kim JK, et al. Altered miRNA processing disrupts brown/white adipocyte determination and associates with lipodystrophy. J Clin Invest. 2014;124:3339–51.PubMedPubMedCentralCrossRef Mori MA, Thomou T, Boucher J, Lee KY, Lallukka S, Kim JK, et al. Altered miRNA processing disrupts brown/white adipocyte determination and associates with lipodystrophy. J Clin Invest. 2014;124:3339–51.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Torriani M, Srinivasa S, Fitch KV, Thomou T, Wong K, Petrow E, et al. Dysfunctional subcutaneous fat with reduced dicer and brown adipose tissue gene expression in HIV-infected patients. J Clin Endocrinol Metab. 2016;101:1225–34.PubMedPubMedCentralCrossRef Torriani M, Srinivasa S, Fitch KV, Thomou T, Wong K, Petrow E, et al. Dysfunctional subcutaneous fat with reduced dicer and brown adipose tissue gene expression in HIV-infected patients. J Clin Endocrinol Metab. 2016;101:1225–34.PubMedPubMedCentralCrossRef
73.•
Zurück zum Zitat Koethe JR, McDonnell W, Kennedy A, et al. Adipose Tissue is Enriched for Activated and Late-Differentiated CD8+ T Cells and Shows Distinct CD8+ Receptor Usage, Compared With Blood in HIV-Infected Persons. J Acquir Immune Defic Syndr. 2018;77:e14–21 This small study showed that adipose tissue in treated adults with HIV had a higher percentage of activated and late-differentiated CD8 T cells compared with peripheral blood.PubMedPubMedCentralCrossRef Koethe JR, McDonnell W, Kennedy A, et al. Adipose Tissue is Enriched for Activated and Late-Differentiated CD8+ T Cells and Shows Distinct CD8+ Receptor Usage, Compared With Blood in HIV-Infected Persons. J Acquir Immune Defic Syndr. 2018;77:e14–21 This small study showed that adipose tissue in treated adults with HIV had a higher percentage of activated and late-differentiated CD8 T cells compared with peripheral blood.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15:914–20.PubMedCrossRef Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15:914–20.PubMedCrossRef
75.
Zurück zum Zitat Couturier J, Suliburk JW, Brown JM, Luke DJ, Agarwal N, Yu X, et al. Human adipose tissue as a reservoir for memory CD4+ T cells and HIV. Aids. 2015;29:667–74.PubMedCrossRef Couturier J, Suliburk JW, Brown JM, Luke DJ, Agarwal N, Yu X, et al. Human adipose tissue as a reservoir for memory CD4+ T cells and HIV. Aids. 2015;29:667–74.PubMedCrossRef
76.•
Zurück zum Zitat Damouche A, Pourcher G, Pourcher V, et al. High proportion of PD-1-expressing CD4(+) T cells in adipose tissue constitutes an immunomodulatory microenvironment that may support HIV persistence. Eur J Immunol. 2017;47:2113–23 In HIV-infected adults on ART, there was high PD-1 expression on adipose-tissue-resident CD4 T cells, which inhibits immune activation and contributes to viral persistence.PubMedCrossRef Damouche A, Pourcher G, Pourcher V, et al. High proportion of PD-1-expressing CD4(+) T cells in adipose tissue constitutes an immunomodulatory microenvironment that may support HIV persistence. Eur J Immunol. 2017;47:2113–23 In HIV-infected adults on ART, there was high PD-1 expression on adipose-tissue-resident CD4 T cells, which inhibits immune activation and contributes to viral persistence.PubMedCrossRef
77.
Zurück zum Zitat Palmer CS, Anzinger JJ, Zhou J, Gouillou M, Landay A, Jaworowski A, et al. Glucose transporter 1-expressing proinflammatory monocytes are elevated in combination antiretroviral therapy-treated and untreated HIV+ subjects. J Immunol. 2014;193:5595–603.PubMedCrossRef Palmer CS, Anzinger JJ, Zhou J, Gouillou M, Landay A, Jaworowski A, et al. Glucose transporter 1-expressing proinflammatory monocytes are elevated in combination antiretroviral therapy-treated and untreated HIV+ subjects. J Immunol. 2014;193:5595–603.PubMedCrossRef
78.
79.
Zurück zum Zitat Kosmiski LA, Bacchetti P, Kotler DP, Heymsfield SB, Lewis CE, Shlipak MG, et al. Relationship of fat distribution with adipokines in human immunodeficiency virus infection. J Clin Endocrinol Metab. 2008;93:216–24.PubMedCrossRef Kosmiski LA, Bacchetti P, Kotler DP, Heymsfield SB, Lewis CE, Shlipak MG, et al. Relationship of fat distribution with adipokines in human immunodeficiency virus infection. J Clin Endocrinol Metab. 2008;93:216–24.PubMedCrossRef
80.••
Zurück zum Zitat Neilan TG, Nguyen K-L, Zaha VG, et al. Myocardial steatosis among antiretroviral therapy–treated people with human immunodeficiency virus participating in the REPRIEVE Trial. J Infect Dis. 2020;222:S63–9 This was an ancillary study from the REPRIEVE trial comprised of individuals who underwent MRI/MRS and demonstrated increased intramyocardial triglyceride content in individuals with HIV, which was associated with immune dysfunction.PubMedPubMedCentralCrossRef Neilan TG, Nguyen K-L, Zaha VG, et al. Myocardial steatosis among antiretroviral therapy–treated people with human immunodeficiency virus participating in the REPRIEVE Trial. J Infect Dis. 2020;222:S63–9 This was an ancillary study from the REPRIEVE trial comprised of individuals who underwent MRI/MRS and demonstrated increased intramyocardial triglyceride content in individuals with HIV, which was associated with immune dysfunction.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Prevention. CfDCa. Supplemental Report: Monitoring Selected National HIV Prevention and Care Objectives by Using HIV Surveillance Data United States and 6 Dependent Areas. 2018;24:3. Prevention. CfDCa. Supplemental Report: Monitoring Selected National HIV Prevention and Care Objectives by Using HIV Surveillance Data United States and 6 Dependent Areas. 2018;24:3.
82.
Zurück zum Zitat Prasada S, Rivera A, Nishtala A, Pawlowski AE, Sinha A, Bundy JD, et al. Differential associations of chronic inflammatory diseases with incident heart failure. JACC Heart Fail. 2020;8:489–98.PubMedPubMedCentralCrossRef Prasada S, Rivera A, Nishtala A, Pawlowski AE, Sinha A, Bundy JD, et al. Differential associations of chronic inflammatory diseases with incident heart failure. JACC Heart Fail. 2020;8:489–98.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Mann DL. Incident heart failure in chronic inflammatory diseases: is it time to rethink stage a heart failure? JACC Heart Fail. 2020;8:499–500.PubMedCrossRef Mann DL. Incident heart failure in chronic inflammatory diseases: is it time to rethink stage a heart failure? JACC Heart Fail. 2020;8:499–500.PubMedCrossRef
84.
Zurück zum Zitat Sivanandham R, Kleinman AJ, Sette P, Brocca-Cofano E, Kilapandal Venkatraman SM, Policicchio BB, et al. Nonhuman primate testing of the impact of different Treg depletion strategies on reactivation and clearance of latent simian immunodeficiency virus. J Virol. 2020;94(19):e00533-20. Sivanandham R, Kleinman AJ, Sette P, Brocca-Cofano E, Kilapandal Venkatraman SM, Policicchio BB, et al. Nonhuman primate testing of the impact of different Treg depletion strategies on reactivation and clearance of latent simian immunodeficiency virus. J Virol. 2020;94(19):e00533-20.
85.
Zurück zum Zitat Ponte R, Mehraj V, Ghali P, Couëdel-Courteille A, Cheynier R, Routy J-P. Reversing gut damage in HIV infection: using non-human primate models to instruct clinical research. EBioMedicine. 2016;4:40–9.PubMedPubMedCentralCrossRef Ponte R, Mehraj V, Ghali P, Couëdel-Courteille A, Cheynier R, Routy J-P. Reversing gut damage in HIV infection: using non-human primate models to instruct clinical research. EBioMedicine. 2016;4:40–9.PubMedPubMedCentralCrossRef
Metadaten
Titel
Immune Dysregulation in Myocardial Fibrosis, Steatosis, and Heart Failure: Current Insights from HIV and the General Population
verfasst von
Arjun Sinha
Matthew J. Feinstein
Publikationsdatum
12.01.2021
Verlag
Springer US
Erschienen in
Current HIV/AIDS Reports / Ausgabe 1/2021
Print ISSN: 1548-3568
Elektronische ISSN: 1548-3576
DOI
https://doi.org/10.1007/s11904-020-00536-9

Weitere Artikel der Ausgabe 1/2021

Current HIV/AIDS Reports 1/2021 Zur Ausgabe

Implementation Science (E Geng, Section Editor)

Patient-Led Decision-Making for HIV Preexposure Prophylaxis

Complications of HIV and Antiretroviral Therapy (GA McComsey, Section Editor)

Gastrointestinal Dysfunction and HIV Comorbidities

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.