Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Sep 1.
Published in final edited form as: Curr Opin HIV AIDS. 2022 Jul 5;17(5):286–292. doi: 10.1097/COH.0000000000000755

HIV and cardiovascular disease: the role of inflammation

Sahera Dirajlal-Fargo 1,2, Nicholas Funderburg 3
PMCID: PMC9370832  NIHMSID: NIHMS1818900  PMID: 35938462

Abstract

Purpose of Review:

HIV and antiretroviral therapy (ART) use are linked to an increased incidence of atherosclerotic cardiovascular disease (ASCVD). Immune activation persists in ART-treated people with HIV (PWH), and markers of inflammation (i.e. IL-6, C-reactive protein) predict mortality in this population. This review discusses underlying mechanisms that likely contribute to inflammation and the development of ASCVD in PWH.

Recent Findings:

Persistent inflammation contributes to accelerated ASCVD in HIV and several new insights into the underlying immunologic mechanisms of chronic inflammation in PWH have been made (e.g. clonal hematopoiesis, trained immunity, lipidomics). We will also highlight potential pro-inflammatory mechanisms that may differ in vulnerable populations including women, minorities and children.

Summary:

Mechanistic studies into the drivers of chronic inflammation in PWH are ongoing and may aid in tailoring effective therapeutic strategies that can reduce ASCVD risk in this population. Focus should also include factors that lead to persistent disparities in HIV care and co-morbidities, including sex as a biological factor and social determinants of health. It remains unclear if ASCVD progression in HIV is driven by unique mediators (HIV itself, ART, immunodeficiency), or if it is an accelerated version of disease progression seen in the general population.

Keywords: cardiovascular disease, inflammation, HIV, immune activation

Introduction

Human immunodeficiency virus (HIV) infection is associated with increased atherosclerotic cardiovascular disease (ASCVD) risk, potentially due to chronic immune activation and inflammation(1, 2). Plasma markers of inflammation, including interleukin (IL)-6, high-sensitivity C-reactive protein (hs-CRP), D-dimer, tumor necrosis factor receptor (TNFR) 1 and 2, and soluble CD14 (sCD14) are independent predictors of mortality, including CVD-related deaths, in people with HIV (PWH)(3). Recent findings from the mechanistic substudy of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE), a large ongoing primary prevention trial in PWH, demonstrate that several biomarkers of innate immune activation and inflammation were significantly higher in participants with, compared to levels in participants without, coronary artery plaques as measured by Computed Tomography Angiography (CTA)(4). These findings highlight that even in virally suppressed PWH with low CVD risk scores, there is a high plaque burden that is associated with increased immune activation and arterial inflammation. Multiple mechanisms likely contribute to persistent immune activation in antiretroviral therapy (ART) treated PWH, including: HIV persistence, co-pathogen infection, microbial translocation, and pro-inflammatory lipids(3). Many of these drivers activate innate immune pathways and the production of cytokines (TNF, IL-6, IL-1β) that contribute to vascular inflammation and progression of atherosclerotic cardiovascular disease (ASCVD). In this article, we will focus on potential mediators of inflammation and the underlying biological mechanisms that may contribute to ASCVD progression in PWH.

Mechanisms contributing to inflammation in PWH

Clonal Hematopoiesis of indeterminate potential (CHIP) and CVD

Migration of immune cells into the blood vessel wall and the pro-inflammatory mediators these cells produce contribute to ASCVD progression(5, 6). Inflammation may modify hematopoietic stem and progenitor cells (HSPCs) in the bone marrow, resulting in the production of immune cells with altered functional and proliferative capacity; these cells may contribute to inflammation and ASCVD(7, 8). Recent studies have explored relationships among clonal hematopoiesis of indeterminate potential (CHIP), inflammation, and CVD risk in PWH. The prevalence of somatic mutations associated with CHIP are enriched among PWH and are related to ART duration, levels of IL-6 and hs-CRP in the Age-Related Clonal hematopoiesis in an HIV Evaluation cohort (ARCHIVE)(9). The increased prevalence of CHIP mutations in PWH has been confirmed in other studies; however, associations between CHIP and inflammation are inconsistent. While levels of inflammation and monocyte activation in PWH (e.g. IL-6, CRP, sCD14, sCD163) were not related to CHIP in a recent publication by van der Heijden et al., levels of von Willebrand factor and D-dimer were increased in CHIP carriers among PWH(10). The HIV reservoir, low nadir CD4 counts and an increased CD4/CD8 T cell ratio were also related to CHIP prevalence in this study(10). In the Swiss HIV Cohort study and the Atherosclerosis Risk in Communities study (ARIC), HIV was associated with a two-fold increase in CHIP prevalence, and there was a trend towards an association between CHIP and coronary artery disease in PWH(11). While age is a known contributor to CHIP mutations in the general population, multiple factors, including immunodeficiency, inflammation, coagulation, and ART, may contribute to CHIP prevalence in PWH, but further exploration into the underlying mechanisms related to CHIP, and whether systemic and vascular inflammation are cause or consequence of CHIP in PWH is needed.

Microbial Translocation

The impaired intestinal mucosal barrier and the resultant translocation of microbial products into the circulation likely contribute to HIV-associated immune activation(1214). Recognition of microbial products by innate immune receptors results in cellular activation and production of inflammatory cytokines. Markers of microbial translocation and gut barrier dysfunction have been associated with biomarkers of inflammation and mortality in PWH(12, 1517). Previous attempts to reduce microbial translocation, however, have yielded mixed results: sevelamer, a phosphate-lowering drug that decreases circulating LPS, did not reduce microbial translocation or inflammation in PWH, however, it did decrease levels of the procoagulant molecule tissue factor (18). Additionally, prebiotics, symbiotics and probiotics, have been studied in HIV and the administration appears to be safe and able to modify microbiota composition, but with limited to no effect on systemic inflammatory markers, pathology or gut permeability (1926). None of these studies measured changes in CVD specific markers. Recent findings from a proof of concept trial found that teduglutide, a glucagon-like peptide-2 analog, decreased arterial inflammation after 6 months of treatment in PWH. The reduction in arterial inflammation was associated with a reduction in activated monocytes and CD8 T cells(27). These findings suggest that improving intestinal integrity may reduce inflammation and CVD risk in PWH.

The Lipidome

Alterations in lipid profiles may also contribute to chronic inflammation and exacerbate ASCVD in PWH. Technological advances have enabled investigators to examine the concentration and fatty acid composition of multiple lipid classes and individual lipid species (“the lipidome”). Lipid profiles have been associated with CVD risk and atherosclerosis in people with(28, 29) and without(30) HIV. Treatment with ART alters lipid levels, and various classes of ART drugs (e.g. protease inhibitors versus integrase inhibitors) may have differential effects on the lipidome(31). Several lipid classes and species that are associated with CVD in people without HIV(30) are increased in PWH and are related to levels of inflammatory biomarkers (e.g. sCD14, IL-6, D-dimer) that are predictive of CVD risk in this population(32, 33).

Lipids may activate inflammatory cascades and drive CVD progression(34, 35). Plasma concentrations of saturated fatty acids (SaFA) have been associated with ASCVD(36, 37), including non-calcified coronary plaque burden(38), in HIV- populations and these lipids likely contribute to inflammation. In vitro studies demonstrate that SaFAs can activate the inflammasome within myeloid cells(35), and induce NFκB signaling in macrophages(39). Exposure of monocytes from HIV- individuals to the SAFAs stearic (18:0) or palmitic acid (16:0), molecules that are increased in PWH, induced activation marker expression on monocyte subsets and the production of inflammatory cytokines (IL-6 and TNFα) in PBMCs(40). Monocytes from PWH also have altered lipid handling capabilities(41, 42), priming them for foam cell formation and contributing to atherosclerotic plaque progression.

While several lipids may contribute to inflammation and atherosclerosis, ceramides (CER) may be of particular interest in PWH. Ceramides can activate the inflammasome and increase IL-1β production and levels of CERs can be increased by inflammation (43). Elevated levels of CER species (CER16:0, CER22:0, CER24:0, and CER 24:1) are associated with ART use, carotid atherosclerosis, and levels of sCD14 in PWH(28). Increased CER levels were also directly related to pro-coagulant, pro-atherosclerotic gene profiles in monocyte derived macrophages (MDMs) in PWH(42). Recently, in a substudy of SATURN HIV(4446), we reported that treatment with rosuvastatin decreased levels of CERs associated with atherosclerosis in PWH and improved pro-inflammatory gene expression profiles in MDMs(47). Whether or not statin induced improvements in lipid and inflammatory profiles are related to a reduction in CVD events in PWH is being explored in the REPRIEVE trial (48).

Trained Immunity

Acute exposure of immune cells to microbial products and pro-inflammatory lipids contributes to low grade inflammation, persistent exposure to these molecules exacerbates the inflammatory response. Trained Immunity describes the mechanisms whereby cells of the innate immune system (i.e. monocytes, macrophages) respond more robustly to re-exposure of pathogen associated molecular patterns (PAMPs) or other “danger signals” (i.e. lipopolysaccharide, flagellin, oxidized LDL)(4953). Innate immune cells undergo transcriptional, epigenetic, and metabolic changes in response to Toll-like Receptor (TLR) ligation, resulting in innate immune memory and enhanced functional capabilities on repeated exposure(50, 53). Trained immunity of monocytes and macrophages has been proposed as a mediator of vascular inflammation and atherosclerosis in the ageing general population(51). Several studies have demonstrated increased responsiveness of immune cells from PWH to TLR ligands(54, 55). Recent work shows that monocytes, but not Tcells, from PWH have increased cytokine production in response to exposure to a number of TLR ligands, including bacterial and viral products. This enhanced monocyte response was related to priming of the IL-1β pathway and increased levels hsCRP, sCD14, and β-glucan, a known inducer of trained immunity(56). Chronic exposure of immune cells to TLR ligands, including molecules derived from HIV-1 itself, proinflammatory lipids, or bacterial products from the damaged GI-tract, may all exacerbate innate immune responses in PWH. How the myriad drivers of trained immunity contribute to inflammatory profiles and CVD risk in PWH requires further investigation.

ART and Mitochondrial Function

Treatment with ART has improved the lifespans of PWH, but ART may also decrease mitochondrial function and contribute directly to inflammation and immune cell activation. Mitochondrial dysfunction increases production of reactive oxygen species (ROS) and activation of redox sensitive transcription factors (including NFκB)(5759). Products derived from damaged mitochondria can activate the inflammasome and increase IL-1β production(60, 61). Singh et al reported that combination ART can induce a senescent phenotype (e.g., telomere shortening, increased ROS production and inflammatory cascade activation, and decreased efferocytosis) in monocytes and macrophages(62). Recent work from Korencak et al. demonstrated that exposure of peripheral blood mononuclear cells (PBMCs) to dolutegravir (DLG) or elvitegravir (EVG) resulted in decreased proliferation and polyfunctionality of CD4+ T cells and promoted a TNF-α focused stress response in these cells(63). We have also shown that exposure to tenofovir disoproxil fumerate (TDF) or emtricitibine (FTC) resulted in decreased mitochondrial function in PBMCs and increased ROS production from T cells and monocyte subsets and induced changes in the functional (i.e. lipid uptake, efferocytosis) and transcriptomic profiles of monocyte derived macrophages (MDMs)(64). Exposure to TDF or FTC resulted in profound changes in MDM gene expression, including changes in genes associated with immune signaling (i.e. STAT2, MyD88, NLRP3) and genes related to histones and chromatin/nucleosome structure. Exposure of MDMs to ART regimens containing TDF, FTC, and efavirenz (EFV) or abacavir (ABC), lamivudine (3TC) and DTG resulted in decreased proliferation of these cells and increased ROS and inflammatory cytokine production(65). ART induced mitochondrial dysfunction may contribute to persistent inflammation in PWH and as newer ART regimens are developed, an understanding of how these drugs influence immune cell function is needed.

Social Determinants of health and vulnerable populations

Sex as a biological factor

Racial and ethnic minority, sexual and gender minority, and low-income people continue to experience persistent disparities in HIV incidence, prevalence, access to care, and other HIV-related health outcomes(66). Specifically, large epidemiologic studies have highlighted increased CVD risk in women living with HIV (WLHIV) compared to men living with HIV (MLHIV)(6769). Several studies have identified that WLHIV have higher systemic immune activation and decreased gut integrity markers compared to MLHIV(7074). Similar results were found in a cohort of WLHIV in rural Uganda(75). Recent data presented from REPRIEVE(76), suggest that although WLHIV had lower prevalence of coronary artery plaques, and lower prevalence of plaques of non-calcified portions and/or vulnerable plaque features, they had higher levels of systemic inflammatory markers, including D-Dimer, and a higher percentage of inflammatory and patrolling monocytes. In addition, immune-plaque relationships differed by sex for D-dimer. Sex-related differences in immune activation in HIV may be influenced by hormonal effects on immune cell function, specifically related to sex-specific expression of TLRs(77, 78), sex-specificity of the microbiome(79) and behavioral factors which may not become relevant until puberty since these sex differences have not been found in pre-pubertal children(80). Similarly, the transgender population is disproportionately affected by HIV and is at higher risk of CVD due to HIV-related risks and potentially, due to hormone therapy(8183). The role of inflammation in the transgender population with HIV and how it may differ from the cisgender population has not been well studied. Social threats, minority stress and HIV may all be drivers of inflammation in this population(84).

Socioeconomic factors

Socio-economic inequality, structural racism and vulnerability may also influence chronic inflammation and downstream co-morbidities. In the Women’s Interagency HIV Study (WIHS), food insecurity was associated with inflammatory markers IL-6 and sTNFR1 in a cross-sectional analysis of WLHIV in the United States, even after adjusting for viral suppression and nutritional factors[26]. Similarly, we found that lack of electricity and clean water were associated with IL-6 and D-dimer in children with HIV, but not in those without HIV in Uganda even after adjusting for demographic and HIV-related factors(85). Careful consideration of biological and social factors are especially important when enrolling comparison cohorts of people with and without HIV in order to appreciate potential differences in inflammatory profiles and CVD risk in these populations.

Youth

ART scale-up has dramatically reduced rates of pediatric HIV mortality and morbidity. Recent findings suggest that children, despite contemporary ART, continue to be susceptible to subclinical cardiovascular and metabolic complications at an early age and that mechanisms may differ from adults. In a study conducted at our US site (Cleveland)(86), McComsey et al showed higher intima-media thickness (IMT) in children with HIV, with a subsequent follow up showing that increased hsCRP was independently associated with thicker IMT(87). On the other hand, other reports from children in Spain and Uganda have not found a relation between IMT and inflammation or immune activation(88, 89). These findings suggest that although youths with HIV also have persistent immune dysfunction (9092), the factors driving cardiometabolic complications in this population may differ from adults.

The microbiome:

Early childhood is a transitional period that is key for acquisition of human microbiome. HIV/ART exposure in utero, early HIV acquisition and ART initiation likely all play a role in alteration in the gut microbiome that has been described in children with perinatally acquired HIV (PHIV) (9395). Similarly, to adults with HIV, a decrease in alpha diversity (a measure of bacterial richness and evenness within a community), and a shift from Bacteroides to Prevotella predominance are associated with markers of immune and endothelial cell activation markers (94, 95).

The lipidome:

We have also highlighted difference in the lipidome in this population which may be distinct from adults living with HIV. Biomarkers associated with CVD risk including hsCRP, sCD163, and CD8+ T cell activation were directly correlated with lipid species in PHIV(89). When comparing serum fatty acids concentrations in PHIV to those in HIV+ adults on ART(40), we find that levels of SaFA are nearly two fold higher in PHIV, suggesting that this population likely has a unique lipidomic signature.

Mitochondrial function:

Mitochondrial function and substrate utilization are perturbed in young children with PHIV(96, 97). Despite early ART treatment and viral suppression, cross-sectional analyses suggest that young PHIV children (mean age= 6 years) have mitochondrial impairment as measured by complex IV, citrate synthase and mitochondrial DNA content(97). It is unclear, however, whether decades of ART will allow for recovery or further declines in mitochondrial function.

Conclusion

Cardiovascular disease is the leading cause of death in the United States; and there is ~2-fold greater risk for CVD events for PWH(1, 2). The mechanisms underlying increased CVD prevalence in PWH are not fully understood, but likely involve persistent systemic and vascular inflammation. Even when ART is initiated during acute infection, some, but not all, inflammatory biomarkers remain elevated in PWH compared to levels in people without HIV(98, 99). It will be interesting to assess whether early ART initiation improves CVD risk in PWH in the future. Multiple potential mediators of inflammation have been identified in PWH and recent studies have sought to link patterns of biomarkers to specific drivers of immune activation and to clinical outcomes in this population (100102). Potentially, certain “drivers of inflammation” or the patterns of inflammatory mediators they elicit will be more strongly associated with specific comorbidities (e.g. CVD, cancer, neurological impairments). Understanding potential mediators of inflammation in PWH and the biological processes they regulate, may inform future clinical interventions. Additionally, understanding the causal effect of HIV on co-morbidities in the context of age, sex, race and social determinants of health deserves special consideration. These factors may not only predict HIV infection, but also CVD risk, and they remain poorly measured and investigated in co-morbidity research. There is a need for research focused on identification of mechanistic pathways contributing to excess risk of CVD among HIV-infected women, sexual minorities, and the transgender population. In addition, considering the consequences of lifelong HIV, ART, and inflammation on children living with perinatally acquired HIV may provide insights into immune health and function in a vulnerable population with implications for treatment of comorbid conditions, even though they lack other traditional risk factors such as hypertension, smoking and substance use that contribute to CVD in adults with HIV. The complex mechanisms that underpin how chronic immune dysregulation and inflammation may contribute to ASCVD risk in PWH remain an important area of clinical and scientific investigation.

  • Cardiovascular disease is increased in people with HIV (PWH) and is likely driven by chronic inflammation.

  • Trained immunity may exacerbate inflammation in PWH and microbial products and pro-inflammatory lipids likely play a role in initiating pathways related to trained immunity.

  • Early treatment with antiretroviral therapy (ART) likely improves inflammatory profiles in PWH, but ART may also decrease mitochondrial function and alter immune cell function.

  • Cardiovascular disease is especially important in the aging population of PWH, but consideration of the cardiometabilic complications of lifelong exposure to HIV and ART in perinatally infected children is also warranted.

Acknowledgements

Financial support and Sponsorship

Dr. Funderburg has received funding related to this work from the National Heart Lung and Blood institute (R01HL158592) and Dr. Funderburg and Dr. Dirajlal-Fargo have received funding from the National Institute of Allergy and Infectious Diseases (U01AI168630).

Footnotes

Conflicts of Interest

Dr. Funderburg has served a consultant and received support from Gilead.

References

  • 1. Shah ASV, Stelzle D, Lee KK, Beck EJ, Alam S, Clifford S, et al. Global Burden of Atherosclerotic Cardiovascular Disease in People Living With HIV. Circulation. 2018;138(11):1100–12. Papers of particular interest, published recently, have been highlighted as: • Of importance
  • 2.Triant VA, Lee H, Hadigan C, Grinspoon SK. Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease. The Journal of clinical endocrinology and metabolism. 2007;92(7):2506–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Gabuzda D, Jamieson BD, Collman RG, Lederman MM, Burdo TH, Deeks SG, et al. Pathogenesis of Aging and Age-related Comorbidities in People with HIV: Highlights from the HIV ACTION Workshop. Pathog Immun. 2020;5(1):143–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Udo Hoffmann MTL, Borek Foldyna, Markella V. Zanni, Tricia H. Burdo, Carl Fichtenbaum, Turner E. Overton, Judith A Aberg, Judith S. Currier, Craig A. Sponseller, Kathleen Melbourne, Pamela S. Douglas, Heather J. Ribaudo0, Thomas Mayrhofer, Steven Grinspoon. CORONARY ARTERY DISEASE, TRADITIONAL RISK, AND INFLAMMATION AMONG PWH IN REPRIEVE. Conference on Retroviruses and Opportunistic Infections; Virtual2022. [Google Scholar]
  • 5.Fernandez DM, Rahman AH, Fernandez NF, Chudnovskiy A, Amir ED, Amadori L, et al. Single-cell immune landscape of human atherosclerotic plaques. Nat Med. 2019;25(10):1576–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Williams JW, Zaitsev K, Kim KW, Ivanov S, Saunders BT, Schrank PR, et al. Limited proliferation capacity of aortic intima resident macrophages requires monocyte recruitment for atherosclerotic plaque progression. Nat Immunol. 2020. [DOI] [PMC free article] [PubMed]
  • 7.Lavine KJ, Pinto AR, Epelman S, Kopecky BJ, Clemente-Casares X, Godwin J, et al. The Macrophage in Cardiac Homeostasis and Disease: JACC Macrophage in CVD Series (Part 4). J Am Coll Cardiol. 2018;72(18):2213–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Dragoljevic D, Westerterp M, Veiga CB, Nagareddy P, Murphy AJ. Disordered haematopoiesis and cardiovascular disease: a focus on myelopoiesis. Clin Sci (Lond). 2018;132(17):1889–99. [DOI] [PubMed] [Google Scholar]
  • 9.Dharan NJ, Yeh P, Bloch M, Yeung MM, Baker D, Guinto J, et al. HIV is associated with an increased risk of age-related clonal hematopoiesis among older adults. Nat Med. 2021;27(6):1006–11. [DOI] [PubMed] [Google Scholar]
  • 10.van der Heijden WA, van Deuren RC, van de Wijer L, van den Munckhof ICL, Steehouwer M, Riksen NP, et al. Clonal Hematopoiesis Is Associated With Low CD4 Nadir and Increased Residual HIV Transcriptional Activity in Virally Suppressed Individuals With HIV. J Infect Dis. 2022;225(8):1339–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bick AG, Popadin K, Thorball CW, Uddin MM, Zanni MV, Yu B, et al. Increased prevalence of clonal hematopoiesis of indeterminate potential amongst people living with HIV. Sci Rep. 2022;12(1):577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12(12):1365–71. [DOI] [PubMed] [Google Scholar]
  • 13.Marchetti G, Tincati C, Silvestri G. Microbial translocation in the pathogenesis of HIV infection and AIDS. Clinical microbiology reviews. 2013;26(1):2–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Somsouk M, Estes JD, Deleage C, Dunham RM, Albright R, Inadomi JM, et al. Gut epithelial barrier and systemic inflammation during chronic HIV infection. AIDS. 2015;29(1):43–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Funderburg NT, Zidar DA, Shive C, Lioi A, Mudd J, Musselwhite LW, et al. Shared monocyte subset phenotypes in HIV-1 infection and in uninfected subjects with acute coronary syndromes. Blood. 2012. [DOI] [PMC free article] [PubMed]
  • 16.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(8):1228–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sandler NG, Wand H, Roque A, Law M, Nason MC, Nixon DE, et al. Plasma levels of soluble CD14 independently predict mortality in HIV infection. J Infect Dis. 2011;203(6):780–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sandler NG, Zhang X, Bosch RJ, Funderburg NT, Choi AI, Robinson JK, et al. 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. The Journal of Infectious Diseases. 2014;210(10):1549–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Happel AU, Barnabas SL, Froissart R, Passmore JS. Weighing in on the risks and benefits of probiotic use in HIV-infected and immunocompromised populations. Beneficial microbes. 2018;9(2):239–46. [DOI] [PubMed] [Google Scholar]
  • 20.d’Ettorre G, Ceccarelli G, Giustini N, Serafino S, Calantone N, De Girolamo G, et al. Probiotics Reduce Inflammation in Antiretroviral Treated, HIV-Infected Individuals: Results of the “Probio-HIV” Clinical Trial. PloS one. 2015;10(9):e0137200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Serrano-Villar S, Vázquez-Castellanos JF, Vallejo A, Latorre A, Sainz T, Ferrando-Martínez S, et al. The effects of prebiotics on microbial dysbiosis, butyrate production and immunity in HIV-infected subjects. Mucosal immunology. 2017;10(5):1279–93. [DOI] [PubMed] [Google Scholar]
  • 22.Stiksrud B, Nowak P, Nwosu FC, Kvale D, Thalme A, Sonnerborg A, et al. Reduced Levels of D-dimer and Changes in Gut Microbiota Composition After Probiotic Intervention in HIV-Infected Individuals on Stable ART. Journal of acquired immune deficiency syndromes (1999). 2015;70(4):329–37. [DOI] [PubMed] [Google Scholar]
  • 23.Yang OO, Kelesidis T, Cordova R, Khanlou H. Immunomodulation of antiretroviral drug-suppressed chronic HIV-1 infection in an oral probiotic double-blind placebo-controlled trial. AIDS research and human retroviruses. 2014;30(10):988–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Serrano-Villar S, de Lagarde M, Vázquez-Castellanos J, Vallejo A, Bernadino JI, Madrid N, et al. Effects of Immunonutrition in Advanced Human Immunodeficiency Virus Disease: A Randomized Placebo-controlled Clinical Trial (Promaltia Study). Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2019;68(1):120–30. [DOI] [PubMed] [Google Scholar]
  • 25.Santos A, Silveira EAD, Falco MO, Nery MW, Turchi MD. Effectiveness of nutritional treatment and synbiotic use on gastrointestinal symptoms reduction in HIV-infected patients: Randomized clinical trial. Clinical nutrition (Edinburgh, Scotland). 2017;36(3):680–5. [DOI] [PubMed] [Google Scholar]
  • 26.Presti RM, Yeh E, Williams B, Landay A, Jacobson JM, Wilson C, et al. A Randomized, Placebo-Controlled Trial Assessing the Effect of VISBIOME ES Probiotic in People With HIV on Antiretroviral Therapy. Open Forum Infect Dis. 2021;8(12):ofab550-ofab. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Caroline E. Diggins SM, Douglas S. Kwon, Charles F. Saylor, Lediya Cheru, Jae Sim, Meaghan Flagg, Bjoern Corleis, Emily Rudmann, Shady Abohashem, Ahmed Tawakol, Janet Lo, editor IMPROVING INTESTINAL BARRIER USING GLP-2 AGONIST REDUCES ARTERIAL INFLAMMATION IN PWH. Conference on Retroviruses and Opportunistic Infections; 2022; Virtual. [Google Scholar]
  • 28. Zhao W, Wang X, Deik AA, Hanna DB, Wang T, Haberlen SA, et al. Elevated Plasma Ceramides Are Associated With Antiretroviral Therapy Use and Progression of Carotid Artery Atherosclerosis in HIV Infection. Circulation. 2019. •This work provides a linkage between alterations in CER levels, inflammation and atherosclerosis in ART treated PWH.
  • 29.Chai JC, Deik AA, Hua S, Wang T, Hanna DB, Xue X, et al. Association of Lipidomic Profiles With Progression of Carotid Artery Atherosclerosis in HIV Infection. JAMA Cardiol. 2019;4(12):1239–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Toledo E, Wang DD, Ruiz-Canela M, Clish CB, Razquin C, Zheng Y, et al. Plasma lipidomic profiles and cardiovascular events in a randomized intervention trial with the Mediterranean diet. Am J Clin Nutr. 2017;106(4):973–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Chaudhary NS, Kind T, Willig AL, Saag MS, Shrestha S, Funderburg N, et al. Changes in lipidomic profile by anti-retroviral treatment regimen: An ACTG 5257 ancillary study. Medicine (Baltimore). 2021;100(30):e26588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bowman ER, Kulkarni M, Gabriel J, Mo X, Klamer B, Belury M, et al. Plasma lipidome abnormalities in people with HIV initiating antiretroviral therapy. Translational Medicine Communications. 2020;5(1):26. [Google Scholar]
  • 33.Belury MA, Bowman E, Gabriel J, Snyder B, Kulkarni M, Palettas M, et al. Prospective Analysis of Lipid Composition Changes with Antiretroviral Therapy and Immune Activation in Persons Living with HIV. Pathog Immun. 2017;2(3):376–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Akerele OA, Cheema SK. Fatty acyl composition of lysophosphatidylcholine is important in atherosclerosis. Med Hypotheses. 2015. [DOI] [PubMed]
  • 35.Robblee MM, Kim CC, Porter Abate J, Valdearcos M, Sandlund KL, Shenoy MK, et al. Saturated Fatty Acids Engage an IRE1alpha-Dependent Pathway to Activate the NLRP3 Inflammasome in Myeloid Cells. Cell Rep. 2016;14(11):2611–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Martinelli N, Girelli D, Malerba G, Guarini P, Illig T, Trabetti E, et al. FADS genotypes and desaturase activity estimated by the ratio of arachidonic acid to linoleic acid are associated with inflammation and coronary artery disease. Am J Clin Nutr. 2008;88(4):941–9. [DOI] [PubMed] [Google Scholar]
  • 37.Fernandez C, Sandin M, Sampaio JL, Almgren P, Narkiewicz K, Hoffmann M, et al. Plasma lipid composition and risk of developing cardiovascular disease. PloS one. 2013;8(8):e71846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ueeda M, Doumei T, Takaya Y, Shinohata R, Katayama Y, Ohnishi N, et al. Serum N-3 polyunsaturated fatty acid levels correlate with the extent of coronary plaques and calcifications in patients with acute myocardial infarction. Circ J. 2008;72(11):1836–43. [DOI] [PubMed] [Google Scholar]
  • 39.Suganami T, Tanimoto-Koyama K, Nishida J, Itoh M, Yuan X, Mizuarai S, et al. Role of the Toll-like receptor 4/NF-kappaB pathway in saturated fatty acid-induced inflammatory changes in the interaction between adipocytes and macrophages. Arterioscler Thromb Vasc Biol. 2007;27(1):84–91. [DOI] [PubMed] [Google Scholar]
  • 40.Bowman ER, Kulkarni M, Gabriel J, Cichon MJ, Riedl K, Belury MA, et al. Altered Lipidome Composition Is Related to Markers of Monocyte and Immune Activation in Antiretroviral Therapy Treated Human Immunodeficiency Virus (HIV) Infection and in Uninfected Persons. Frontiers in Immunology. 2019;10(785). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Maisa A, Hearps AC, Angelovich TA, Pereira CF, Zhou J, Shi MD, et al. Monocytes from HIV-infected individuals show impaired cholesterol efflux and increased foam cell formation after transendothelial migration. AIDS (London, England). 2015;29(12):1445–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Bowman ER, Cameron CM, Richardson B, Kulkarni M, Gabriel J, Cichon MJ, et al. Macrophage maturation from blood monocytes is altered in people with HIV, and is linked to serum lipid profiles and activation indices: A model for studying atherogenic mechanisms. PLoS Pathog. 2020;16(10):e1008869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chaurasia B, Talbot CL, Summers SA. Adipocyte Ceramides-The Nexus of Inflammation and Metabolic Disease. Front Immunol. 2020;11:576347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Funderburg NT, Jiang Y, Debanne SM, Labbato D, Juchnowski S, Ferrari B, et al. Rosuvastatin Reduces Vascular Inflammation and T-cell and Monocyte Activation in HIV-Infected Subjects on Antiretroviral Therapy. Journal of acquired immune deficiency syndromes (1999). 2015;68(4):396–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Funderburg NT, Jiang Y, Debanne SM, Storer N, Labbato D, Clagett B, et al. Rosuvastatin Treatment Reduces Markers of Monocyte Activation in HIV-Infected Subjects on Antiretroviral Therapy. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2014;58(4):588–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hileman CO, Turner R, N TF, Semba RD, McComsey GA. Changes in oxidized lipids drive the improvement in monocyte activation and vascular disease after statin therapy in HIV. AIDS. 2016;30(1):65–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ismail JC C. Kumar M. Tamilselvan B. Riedl K. Cannon. Bowman E. McComsey GA. Funderburg N. Cameron MJ. STATIN USE IN ART-TREATED HIV REVEALS AN ALTERED MACROPHAGE TRANSCRIPTOMIC PROFILE. Conference on Retroviruses and Opportunistic Infecitons; February 12–16, 2022; Virtual2022.
  • 48. Grinspoon SK, Fitch KV, Overton ET, Fichtenbaum CJ, Zanni MV, Aberg JA, et al. Rationale and design of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE). Am Heart J. 2019;212:23–35. • REPRIEVE is a prospective randomized, placebo-controlled trial of statin as a primary CVD prevention strategy in PWH.
  • 49.Saeed S, Quintin J, Kerstens HH, Rao NA, Aghajanirefah A, Matarese F, et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science. 2014;345(6204):1251086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Netea MG, Dominguez-Andres J, Barreiro LB, Chavakis T, Divangahi M, Fuchs E, et al. Defining trained immunity and its role in health and disease. Nat Rev Immunol. 2020;20(6):375–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Zhong C, Yang X, Feng Y, Yu J. Trained Immunity: An Underlying Driver of Inflammatory Atherosclerosis. Front Immunol. 2020;11:284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Lachmandas E, Boutens L, Ratter JM, Hijmans A, Hooiveld GJ, Joosten LA, et al. Microbial stimulation of different Toll-like receptor signalling pathways induces diverse metabolic programmes in human monocytes. Nat Microbiol. 2016;2:16246. [DOI] [PubMed] [Google Scholar]
  • 53.Owen AM, Fults JB, Patil NK, Hernandez A, Bohannon JK. TLR Agonists as Mediators of Trained Immunity: Mechanistic Insight and Immunotherapeutic Potential to Combat Infection. Front Immunol. 2020;11:622614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Merlini E, Tincati C, Biasin M, Saulle I, Cazzaniga FA, d’Arminio Monforte A, et al. Stimulation of PBMC and Monocyte-Derived Macrophages via Toll-Like Receptor Activates Innate Immune Pathways in HIV-Infected Patients on Virally Suppressive Combination Antiretroviral Therapy. Front Immunol. 2016;7:614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Jalbert E, Crawford TQ, D’Antoni ML, Keating SM, Norris PJ, Nakamoto BK, et al. IL-1Beta enriched monocytes mount massive IL-6 responses to common inflammatory triggers among chronically HIV-1 infected adults on stable anti-retroviral therapy at risk for cardiovascular disease. PloS one. 2013;8(9):e75500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. van der Heijden WA, Van de Wijer L, Keramati F, Trypsteen W, Rutsaert S, Horst RT, et al. Chronic HIV infection induces transcriptional and functional reprogramming of innate immune cells. JCI Insight. 2021;6(7). •This work highlights differential responsiveness of myeloid cells from people with and without HIV to microbial products, potentially identifying trained immunity as a contributor to chronic inflammation in PWH.
  • 57.Amma H, Naruse K, Ishiguro N, Sokabe M. Involvement of reactive oxygen species in cyclic stretch-induced NF-kappaB activation in human fibroblast cells. Br J Pharmacol. 2005;145(3):364–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chung HY, Cesari M, Anton S, Marzetti E, Giovannini S, Seo AY, et al. Molecular inflammation: underpinnings of aging and age-related diseases. Ageing Res Rev. 2009;8(1):18–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Cillero-Pastor B, Carames B, Lires-Dean M, Vaamonde-Garcia C, Blanco FJ, Lopez-Armada MJ. Mitochondrial dysfunction activates cyclooxygenase 2 expression in cultured normal human chondrocytes. Arthritis Rheum. 2008;58(8):2409–19. [DOI] [PubMed] [Google Scholar]
  • 60.Salminen A, Kaarniranta K, Kauppinen A. Inflammaging: disturbed interplay between autophagy and inflammasomes. Aging (Albany NY). 2012;4(3):166–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med. 2017;377(12):1119–31. [DOI] [PubMed] [Google Scholar]
  • 62. Singh MV, Kotla S, Le NT, Ae Ko K, Heo KS, Wang Y, et al. Senescent Phenotype Induced by p90RSK-NRF2 Signaling Sensitizes Monocytes and Macrophages to Oxidative Stress in HIV-Positive Individuals. Circulation. 2019;139(9):1199–216. •This work characterizes a novel pathway by which ART exposure may contribute to inflammation, immune cell senescence and CVD.
  • 63. Korencak M, Byrne M, Richter E, Schultz BT, Juszczak P, Ake JA, et al. Effect of HIV infection and antiretroviral therapy on immune cellular functions. JCI Insight. 2019;4(12). •This work demonstrates that certain ART drugs may induce an inflammatory/stress response in CD4+ Tcells, potentially altering immune cell function in PWH.
  • 64.Bowman ER, Cameron C, Richardson B, Kulkarni M, Gabriel J, Kettelhut A, et al. In Vitro Exposure of Leukocytes to HIV Preexposure Prophylaxis Decreases Mitochondrial Function and Alters Gene Expression Profiles. Antimicrob Agents Chemother. 2020;65(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Wallace J, Gonzalez H, Rajan R, Narasipura SD, Virdi AK, Olali AZ, et al. Anti-HIV Drugs Cause Mitochondrial Dysfunction in Monocyte-Derived Macrophages. Antimicrob Agents Chemother. 2022;66(4):e0194121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lightfoot M, Milburn N, Loeb Stanga L. Addressing Health Disparities in HIV: Introduction to the Special Issue. Journal of acquired immune deficiency syndromes (1999). 2021;88(S1):S1–S5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Freiberg MS, Chang CC, Kuller LH, Skanderson M, Lowy E, Kraemer KL, et al. HIV infection and the risk of acute myocardial infarction. JAMA internal medicine. 2013;173(8):614–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Womack JA, Chang CC, So-Armah KA, Alcorn C, Baker JV, Brown ST, et al. HIV infection and cardiovascular disease in women. Journal of the American Heart Association. 2014;3(5):e001035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Chow FC, Regan S, Feske S, Meigs JB, Grinspoon SK, Triant VA. Comparison of ischemic stroke incidence in HIV-infected and non-HIV-infected patients in a US health care system. Journal of acquired immune deficiency syndromes (1999). 2012;60(4):351–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Fitch KV, Srinivasa S, Abbara S, Burdo TH, Williams KC, Eneh P, et al. Noncalcified coronary atherosclerotic plaque and immune activation in HIV-infected women. J Infect Dis. 2013;208(11):1737–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ticona E, Bull ME, Soria J, Tapia K, Legard J, Styrchak SM, et al. Biomarkers of inflammation in HIV-infected Peruvian men and women before and during suppressive antiretroviral therapy. AIDS. 2015;29(13):1617–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mathad JS, Gupte N, Balagopal A, Asmuth D, Hakim J, Santos B, et al. Sex-Related Differences in Inflammatory and Immune Activation Markers Before and After Combined Antiretroviral Therapy Initiation. Journal of acquired immune deficiency syndromes (1999). 2016;73(2):123–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Griesbeck M, Scully E, Altfeld M. Sex and gender differences in HIV-1 infection. Clin Sci (Lond). 2016;130(16):1435–51. [DOI] [PubMed] [Google Scholar]
  • 74.Dirajlal-Fargo S, Mussi-Pinhata MM, Weinberg A, Yu Q, Cohen R, Harris DR, et al. HIV-exposed uninfected infants have increased inflammation and monocyte activation. AIDS. 2019. [DOI] [PMC free article] [PubMed]
  • 75.Siedner MJ, Zanni M, Tracy RP, Kwon DS, Tsai AC, Kakuhire B, et al. Increased Systemic Inflammation and Gut Permeability Among Women With Treated HIV Infection in Rural Uganda. J Infect Dis. 2018;218(6):922–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Markella V. Zanni BF, Kenneth Williams, Tricia H. Burdo, Sara McCallum, Sara E. Looby, Patrick Autissier, Kathleen Fitch, Judith S. Currier, Michael T. Lu, Pamela S. Douglas, Heather J. Ribaudo, Steven Grinspoon. SUBCLINICAL ATHEROSCLEROSIS AND IMMUNE ACTIVATION AMONG US FEMALES VS MALES WITH HIV. Conference on Retroviruses and Opportunistic Infectious; Virtual2022. [Google Scholar]
  • 77.Lakoski SG, Herrington DM. Effects of hormone therapy on C-reactive protein and IL-6 in postmenopausal women: a review article. Climacteric : the journal of the International Menopause Society. 2005;8(4):317–26. [DOI] [PubMed] [Google Scholar]
  • 78.Meier A, Chang JJ, Chan ES, Pollard RB, Sidhu HK, Kulkarni S, et al. Sex differences in the Toll-like receptor-mediated response of plasmacytoid dendritic cells to HIV-1. Nat Med. 2009;15(8):955–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Rizzetto L, Fava F, Tuohy KM, Selmi C. Connecting the immune system, systemic chronic inflammation and the gut microbiome: The role of sex. Journal of autoimmunity. 2018;92:12–34. [DOI] [PubMed] [Google Scholar]
  • 80.Dirajlal-Fargo S, El-Kamari V, Weiner L, Shan L, Sattar A, Kulkarni M, et al. Altered Intestinal Permeability and Fungal Translocation in Ugandan Children With Human Immunodeficiency Virus. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2020;70(11):2413–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Gosiker BJ, Lesko CR, Rich AJ, Crane HM, Kitahata MM, Reisner SL, et al. Cardiovascular disease risk among transgender women living with HIV in the United States. PloS one. 2020;15(7):e0236177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Alzahrani T, Nguyen T, Ryan A, Dwairy A, McCaffrey J, Yunus R, et al. Cardiovascular Disease Risk Factors and Myocardial Infarction in the Transgender Population. Circulation Cardiovascular quality and outcomes. 2019;12(4):e005597. [DOI] [PubMed] [Google Scholar]
  • 83.Streed CG Jr., Beach LB, Caceres BA, Dowshen NL, Moreau KL, Mukherjee M, et al. Assessing and Addressing Cardiovascular Health in People Who Are Transgender and Gender Diverse: A Scientific Statement From the American Heart Association. Circulation. 2021;144(6):e136–e48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Diamond LM, Dehlin AJ, Alley J. Systemic inflammation as a driver of health disparities among sexually-diverse and gender-diverse individuals. Psychoneuroendocrinology. 2021;129:105215. [DOI] [PubMed] [Google Scholar]
  • 85.Dirajlal-Fargo S, Albar Z, Sattar A, Kulkarni M, Bowman E, Funderburg N, et al. Relationship between economic insecurity, inflammation, monocyte activation and intestinal integrity in children living with HIV in Uganda. AIDS Care. 2020;32(11):1451–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.McComsey GA, O’Riordan M, Hazen SL, El-Bejjani D, Bhatt S, Brennan ML, et al. Increased carotid intima media thickness and cardiac biomarkers in HIV infected children. AIDS. 2007;21(8):921–7. [DOI] [PubMed] [Google Scholar]
  • 87.Ross AC, O’Riordan MA, Storer N, Dogra V, McComsey GA. Heightened inflammation is linked to carotid intima-media thickness and endothelial activation in HIV-infected children. Atherosclerosis. 2010;211(2):492–8. [DOI] [PubMed] [Google Scholar]
  • 88.Sainz T, Alvarez-Fuente M, Navarro ML, Diaz L, Rojo P, Blazquez D, et al. Subclinical Atherosclerosis and Markers of Immune Activation in HIV-Infected Children and Adolescents: The CaroVIH Study. Journal of acquired immune deficiency syndromes (1999). 2014;65(1):42–9. [DOI] [PubMed] [Google Scholar]
  • 89.Dirajlal-Fargo S, Albar Z, Bowman E, Labbato D, Sattar A, Karungi C, et al. Subclinical Vascular Disease in Children With Human Immunodeficiency Virus in Uganda Is Associated With Intestinal Barrier Dysfunction. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2020;71(12):3025–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Dirajlal-Fargo S, Albar Z, Bowman E, Labbato D, Sattar A, Karungi C, et al. Increased monocyte and T-cell activation in treated HIV+ Ugandan children: associations with gut alteration and HIV factors. AIDS. 2020;34(7):1009–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Carrasco I, Tarancon-Diez L, Vázquez-Alejo E, Jiménez de Ory S, Sainz T, Apilanez M, et al. Innate and adaptive abnormalities in youth with vertically acquired HIV through a multicentre cohort in Spain. Journal of the International AIDS Society. 2021;24(10):e25804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Eckard AR, Rosebush JC, Lee ST, O’Riordan MA, Habib JG, Daniels JE, et al. Increased Immune Activation and Exhaustion in HIV-infected Youth. Pediatr Infect Dis J. 2016;35(12):e370–e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Flygel TT, Sovershaeva E, Claassen-Weitz S, Hjerde E, Mwaikono KS, Odland J, et al. Composition of Gut Microbiota of Children and Adolescents With Perinatal Human Immunodeficiency Virus Infection Taking Antiretroviral Therapy in Zimbabwe. J Infect Dis. 2020;221(3):483–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kaur US, Shet A, Rajnala N, Gopalan BP, Moar P, D H, et al. High Abundance of genus Prevotella in the gut of perinatally HIV-infected children is associated with IP-10 levels despite therapy. Sci Rep. 2018;8(1):17679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Sessa L, Reddel S, Manno E, Quagliariello A, Cotugno N, Del Chierico F, et al. Distinct gut microbiota profile in antiretroviral therapy-treated perinatally HIV-infected patients associated with cardiac and inflammatory biomarkers. AIDS. 2019;33(6):1001–11. [DOI] [PubMed] [Google Scholar]
  • 96.Jao J, Jacobson DL, Russell JS, Wang J, Yu W, Gojanovich GS, et al. Perinatally acquired HIV infection is associated with abnormal blood mitochondrial function during childhood/adolescence. AIDS. 2021;35(9):1385–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Shen J, Liberty A, Shiau S, Strehlau R, Pierson S, Patel F, et al. Mitochondrial Impairment in Well-Suppressed Children with Perinatal HIV-Infection on Antiretroviral Therapy. AIDS research and human retroviruses. 2020;36(1):27–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Sereti I, Krebs SJ, Phanuphak N, Fletcher JL, Slike B, Pinyakorn S, et al. Persistent, Albeit Reduced, Chronic Inflammation in Persons Starting Antiretroviral Therapy in Acute HIV Infection. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2017;64(2):124–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Schnittman SR, Deitchman AN, Beck-Engeser G, Ahn H, York VA, Hartig H, et al. Abnormal Levels of Some Biomarkers of Immune Activation Despite Very Early Treatment of Human Immunodeficiency Virus. J Infect Dis. 2021;223(9):1621–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Younas M, Psomas C, Reynes C, Cezar R, Kundura L, Portales P, et al. Residual Viremia Is Linked to a Specific Immune Activation Profile in HIV-1-Infected Adults Under Efficient Antiretroviral Therapy. Front Immunol. 2021;12:663843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Younas M, Psomas C, Reynes C, Cezar R, Kundura L, Portales P, et al. Microbial Translocation Is Linked to a Specific Immune Activation Profile in HIV-1-Infected Adults With Suppressed Viremia. Front Immunol. 2019;10:2185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Psomas C, Younas M, Reynes C, Cezar R, Portales P, Tuaillon E, et al. One of the immune activation profiles observed in HIV-1-infected adults with suppressed viremia is linked to metabolic syndrome: The ACTIVIH study. EBioMedicine. 2016;8:265–76. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES