Abstract
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8+ and CD4+ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Keywords: atherosclerosis, cardiovascular diseases, clinical relevance, endothelial cells, HIV, inflammation, risk factors
In recent decades, the landscape of HIV has undergone a transformative shift, attributed to remarkable advancements in antiretroviral therapy (ART). While these breakthroughs have significantly prolonged the lifespan of individuals living with HIV, a new set of challenges has emerged, particularly noncommunicable comorbidities such as cardiovascular disease (CVD).1 Traditionally perceived as primarily an immunodeficiency disorder, HIV is now recognized as a complex systemic condition that involves persistent immune activation, even in individuals effectively treated with ART.2,3 This sustained immune activation is associated with chronic inflammation, characterized by elevated levels of proinflammatory cytokines, immune cell activation, and heightened coagulation markers.3,4 The chronic immune activation and inflammation are major contributors to the pathogenesis of CVD in this population, which can persist even with effective ART.5,6 One mechanism for this persistent inflammation is viral persistence, where HIV can integrate into the genome of immune cells, creating a reservoir of virus that can persist for the lifetime of the infected individual.3,7 These infected cells can continue to produce viral proteins, leading to chronic immune activation and inflammation. Moreover, HIV proteins can directly activate endothelial cells with inflammation of the blood vessels.8 The combination of these factors can contribute to the development of atherosclerosis and other cardiovascular complications, implicating the chronic inflammatory state in the pathogenesis of various HIV-associated complications, including the acceleration of atherosclerotic cardiovascular disease (ASCVD).2,9
While we currently have no cure for HIV, the Berlin patient diagnosed with acute myeloid leukemia10 and the London patient diagnosed with Hodgkin lymphoma11 both achieved functional cure after allogeneic hematopoietic stem cell transplants with a homozygous CCR5 delta32 mutation. While the cancer treatments differed based on their diagnosis and responses, both patients had confirmed CCR5-tropic virus before transplant and did not experience HIV rebound after transplant.11 The London patient was noted to have cytomegalovirus (CMV) and Epstein-Barr virus reactivation 85 days after transplant requiring treatment.11 Notably, there were differences in the immune cell composition after transplant, the most significant being the lack of CCR5+ cluster of differentiation (CD)4 and CD8+ T cells. Inflated anti-CMVpp65 CD8+ pretransplant (8.5% of CD8 T cells) remained low after transplant (0.14%) at day 819, while anti-CMV CD4+ T cells, which at baseline are less clonal, went from 2% to 1.9% also at day 819 after transplant. There are more studies needed to understand the immune changes that happen after a functional cure for HIV, as we work toward a practical cure approach for the larger population who are not eligible for transplants.
Perturbations caused by HIV and its treatment can impact the drivers of CVD for both better or worse and are, therefore, of clinical relevance to people living with HIV (PLWH) and, more generally, to understand the pathogenesis of CVD in this population. These impacts have changed over the course of the HIV/AIDS epidemic (Table 1).
Table 1.
Changing Features of HIV Treatment and Outcomes in Different Eras of the Epidemic
Early ART (1985–1995) only transiently suppressed HIV replication in most patients until ART resistance, immunodeficiency, AIDS, and death almost inevitably occurred. Profound HIV-induced immunodeficiency was associated with the normalization of endothelial function, even in those with risk factors such as smoking and diabetes.12 During this era, most PLWH experienced anorexia and weight loss, and metabolic syndrome and CVD were rare.13 This pattern reversed in 1995 with the advent of highly effective ART, which sustainably inhibits HIV replication, preventing the emergence of resistance or immunodeficiency. While HAART was an advance in terms of prevention of AIDS and death, manifestations of metabolic syndrome more typical of the general population began to be seen.14,15
Despite earlier, more potent, and more on-target ART, PLWH still have up to twice the risk of CVD as those without HIV.1,16 The recently reported REPRIEVE study (Randomized Trial to Prevent Vascular Events in HIV) of 7769 PLWH with low-to-moderate risk of CVD reported a 35% reduction in major cardiovascular events in those randomized to pitavastatin compared with placebo.17 The findings are in keeping with an ongoing inflammatory response driving CVD in PLWH despite early and effective ART that may be mitigated by pitavastatin.
ATHEROSCLEROSIS IN PLWH
Atherosclerosis is a complex chronic inflammatory disease characterized by endothelial cell injury, accumulation of lipids, immune cells, and fibrous elements within arterial walls. The progression of this process leads to the formation of coronary plaque with gradual narrowing and hardening of arteries, eventually resulting in ischemic cardiovascular events.18 PLWH are at a higher risk of developing ASCVD compared with the general population. The increased risk cannot be solely attributed to traditional CVD risk factors,1,19–22 as even PLWH with low CVD risk have significant coronary plaque burden associated with immune activation.23 Furthermore, PLWH may be up to 4.5× more likely to die from sudden cardiac death compared with controls.24 The pathophysiological mechanisms underlying this link are multifaceted, involving interactions between the immune system, viral persistence, and the endothelium. Among 1612 participants in the MACS (Multicenter AIDS Cohort Study) cohort, men living with HIV who have sex with men recruited from 4 different cities in the United States were shown have been shown to have prolonged QT intervals on 12-led standing resting electrocardiograms, which was associated with systemic inflammatory markers, including ICAM-1 (intercellular adhesion molecule 1), IL (interleukin)-6, and BAFF (B-cell activating factor).25
Decades of research have implicated cells of the innate and adaptive immune systems in the pathogenesis of ASCVD.26 HIV itself, when untreated, is a significant risk factor.27,28 However, it has also been shown that PLWH with AIDS are protected from ASCVD.12 Despite treatment with ART, HIV exerts enduring alterations in the memory pool of immune cells.29 These changes occur through various means, such as direct effects of the virus, coinfections, or ART.7,30,31 As such, PLWH have persistent systemic inflammation, which contributes to multiple comorbidities, including CVD.2 Early initiation of ART with acute HIV is associated with the near normalization of the coagulation cascade and numerous systemic inflammatory biomarkers. However, several inflammatory markers, including the acute-phase reactants, monocyte activation, and fibrosis biomarkers, remain elevated (Table 2).5
Table 2.
Elevated Inflammatory Markers in PLWH Compared With People Without HIV
While monocytes are important in aging-related inflammation among PLWH and people without HIV, chronic infection with HIV is uniquely characterized by T-cell activation and exhaustion, as well as increased soluble inflammatory markers (such as sTNFR-1 [soluble tumor necrosis factor receptor 1], sTNFR-2, and IL-6) in ART recipients with undetectable viremia.3,43–46 Among PLWH, T-cell activation predicts short-term mortality and is associated with future cardiovascular events, emphasizing the potential impact on the broader health spectrum.43,47 In this review, we summarize the literature on the immune system’s role in CVD among PLWH. Specifically, we explore the role of cells of both the innate and adaptive immune systems and their impact on ASCVD progression. Furthermore, we discuss research gaps and summarize potential opportunities for future investigations to discover novel therapies (Figure 1).
Figure 1.
Cardiovascular disease (CVD) risk factors in people living with HIV (PLWH) and people without HIV. HIV-related risk factors (HIV infection, antiretroviral therapy, coinfections, and microbial translocation; A) and traditional risk factors (B) are major drivers of systemic inflammation that contribute to endothelial cell dysfunction and progression to atherosclerosis and CVD among PLWH (C). BMI indicates body mass index; CMV, cytomegalovirus; FH, family history; HCV, hepatitis C virus; and HTN, hypertension. Illustration credit: Sceyence Studios.
Endothelial Dysfunction
Endothelial dysfunction is an early precursor in the development of atherosclerosis.48 This manifests as impaired nitric oxide signaling with reduced ability for vascular relaxation. Injured endothelium allows the entry of cholesterol-rich lipoproteins, and ApoB (apolipoprotein B)–containing lipoproteins, into the subendothelial space. These lipoproteins undergo oxidative, enzymatic, and chemical modifications, activating endothelial cells and vascular smooth muscle cells.49 Activated endothelial cells increase the expression of adhesion molecules, including sVCAM-1 (soluble vascular cell adhesion molecule-1), sICAM-1 (soluble intercellular adhesion molecule 1), and MCP-1 (monocyte chemoattractant protein-1). The result is a chemokine gradient and increased expression of adhesion molecules that trigger a cascade of immune cell influx, dominated by innate and adaptive immune cells, particularly dendritic cells (DCs), monocytes/macrophages, and T cells.50
HIV induces endothelial dysfunction through direct and indirect mechanisms that are not fully defined. In vitro studies suggest a potential direct role for HIV replication, with viral proteins such as gp (glycoprotein) 120 and Tat (transactivator of transcription) exhibiting toxicity to cardiac and vascular cell lines.51,52 In vivo, HIV can infect some human endothelial cells (depending on the organ).53 In addition, HIV proteins, gp120, Tat, and Nef (negative regulatory factor), have been shown to stimulate endothelial cells to express inflammatory cytokines, such as IL-6, adhesion molecules (ICAM-1), and induce endothelial cell apoptosis. gp120 has also been associated with increased expression of MMP2/9 (matrix metalloproteinase 2/9) and IL-8.8 In addition to the direct effects of HIV, plasma cytokines and chemokines also stimulate the endothelium, leading to persistently elevated sVCAM-1 and sICAM-1 levels.32,41,54–56 Notably, sVCAM-1 levels decrease with ART.6,57 Despite the decrease in sVCAM-1 levels, PLWH on ART with effective viral suppression still have higher levels of endothelial inflammation compared with people without HIV. This is confounded by the data that implicate ART in endothelial dysfunction, adding complexity to the understanding of the underlying mechanisms in PLWH.57,58(p1) Endothelial dysfunction among PLWH on therapy has also been linked to monocyte activation.41 In nonhuman primates, simian immunodeficiency virus (SIV) and simian-HIV infections were shown to cause an increase in endothelial proliferation, increased recruitment of subendothelial CD8+T cells and CD68+ macrophages/monocytes, and decreased endothelial function with decreased eNOS (endothelial nitric oxide synthase) compared with negative controls.59
Beyond peripheral endothelial dysfunction, HIV has also been associated with coronary microvascular dysfunction as measured by myocardial blood flow reserve derived from positron emission tomography.60–62 The pathogenesis of coronary microvascular dysfunction among PLWH is probably mediated by chronic inflammation; however, this is not fully understood.61 Like CVD, accumulating immune cells around pulmonary arteries is a risk factor for pulmonary arterial hypertension.63 HIV-related and non-HIV–related factors cause endothelial injury, pulmonary vasculopathy, and eventually HIV-associated pulmonary arterial hypertension.
Fatty Streak Formation
With the accumulation of cholesterol-rich lipoproteins (Table 3), atherosclerotic plaques progress from intimal media thickening to early lesions, known as fatty streaks, characterized by cholesteryl ester-laden foam cells. This is followed by more advanced lesions containing a core region of activated immune cells, cholesterol crystals, and extracellular lipids.73 Elevated plasma cholesterol levels trigger the infiltration of cholesterol-containing lipoprotein particles into the artery wall. This infiltration is facilitated by interactions with proteoglycans of the extracellular matrix, prompting local cellular responses. Leukocyte adhesion molecules and chemokines expressed by vascular cells contribute to the recruitment of circulating monocytes into the tunica intima—the innermost layer of the artery.50
Table 3.
Plasma Lipoproteins in HIV and Atherosclerosis
Apart from its impact on the endothelium, HIV can also directly alter lipid metabolism.65 Immune activation in untreated PLWH may contribute to dyslipidemia due to direct effects of antiviral cytokines, such as IFN (interferon)-α, which are associated with increased serum triglycerides presumed to be from dysregulated clearance.66,74 In a more recent study, the MACS, in both untreated and treated PLWH, elevated IL-6, ICAM-1, and D-dimer levels were associated with decreased small HDL (high-density lipoprotein) particles.75 While treatment with the newer, highly active ART generally improves low HDL cholesterol, it does not fully normalize the lipid profile in treated PLWH. Notably, several classes of ART have also been implicated in altering lipid metabolism and increasing oxidative stress in PLWH, potentially independently contributing to an elevated cardiovascular risk.76
Plaque Progression
Plaque progression is characterized by increased recruitment of inflammatory immune cells and alterations in the function of vascular smooth muscle cells. Vascular smooth muscle cells are traditionally known for their role in extracellular matrix production and exhibit phenotypic plasticity, including the acquisition of plaque-promoting macrophage–like features and plaque-stabilizing fibroblast–like features.49 Classical monocytes, recruited via CCR2 and CCL2 (C-C motif chemokine ligand 2), dominate the monocyte influx in atherosclerosis and differentiate into macrophages on stimulation by mediators such as M-CSF (monocyte-colony stimulating factor). As they transition, the macrophages initiate the uptake and clearance of lipoproteins, forming lipid-rich foam cells.77,78 Macrophages take up cholesterol through various mechanisms, leading to the accumulation of cholesteryl ester droplets in the cytosol and the formation of foam cells. Over time, these foam cells become overloaded, leading to cell death and the formation of a necrotic core within the atherosclerotic lesion.79 Consequently, this process triggers the release of proinflammatory cytokines and adhesion molecules crucial to plaque formation and growth. HIV may hinder the cholesterol efflux of monocytes, leading to enhanced foam cell formation due to altered expression or increased degradation of genes/proteins that regulate cholesterol efflux, such as cholesterol transporter ABCA1 (ATP-binding cassette transporter A1).80,81 The impaired cholesterol transport appears Nef dependent, which has been shown to directly bind to ABCA1, leading to downregulation and translocation of ABCA1 and accumulation of cholesterol in macrophages.82 A study of rhesus macaques infected with SIV also supports that Nef suppresses cholesterol efflux by ABCA1.83 SIV-infected macaques had lower expression of ABCA1 in the livers and coexpression of Nef, without evidence of replicating SIV.83 The accumulation of cholesterol in HIV-infected cells is thought to be important for HIV trans infection from monocytes, B cells, and DCs to CD4+ T cells. There are PLWH who are long-term nonprogressors where an aberrant lipid metabolism appears to be an important advantage that restricts trans infection of CD4+T cells from antigen-presenting cells.84,85
Macrophages within atherosclerotic plaques can display proinflammatory or anti-inflammatory features, with subsets such as inflammatory macrophages and type I IFN-inducible cells leading to plaque growth.86 Macrophage differentiation is associated with increased scavenger receptors, including SR-A (scavenger receptor class a), CD36, MARCO (macrophage receptor with collagenous structure), and LOX-1 (lectin-like oxidized LDL receptor 1). These pattern recognition receptors mediate the internalization of various molecules, including oxidatively modified lipoprotein particles.86 Several factors including netrin 1, produced in plaque macrophages, prevent emigration and lead to macrophage accumulation within the atherosclerotic lesion.87
Noncalcified Versus Calcified Plaque
Several studies have shown that PLWH have a higher prevalence of noncalcified coronary plaque than calcified plaque.23,36,38,88,89 Previous studies in the general population suggest that unstable/vulnerable plaques have spotty calcification and a higher degree of inflammation.90–92 In PLWH, positive arterial remodeling in coronary arteries was seen with noncalcified plaque.93 There is limited literature that supports the role of monocyte activation in the development of noncalcified coronary plaque.38 In some studies, PLWH with noncalcified plaque were older and had higher sCD163.36 sCD163 is both a soluble and cell surface marker associated with atherosclerosis in people with and without HIV.34 While few studies point to the role of monocytes, the underlying mechanisms that promote noncalcified plaque in PLWH are not clear and need to be investigated further.
THE ROLE OF THE INNATE AND ADAPTIVE IMMUNE SYSTEM IN ATHEROSCLEROSIS AMONG PLWH
Here, we summarize the changes in cells of the innate and adaptive immune response and their association with subclinical atherosclerosis (Figure 2).
Figure 2.
Role of cells of the innate and adaptive immune system in atherosclerosis progression. A, People living with HIV (PLWH) with viral suppression on antiretroviral therapy (ART) have increased CD4+ and CD8+ T-cell activation. B, The endothelial cell lining, due to multiple factors including viral replication and exposure to inflammatory cytokines, has increased expression of adhesion molecules, ICAM-1 (intercellular adhesion molecule 1), and VCAM-1 (vascular cell adhesion molecule 1). C and D, Cells of the innate and adaptive immune system adhere to inflamed endothelium and are recruited into the subendothelial space. E, Monocytes present in atheroma and infected with HIV are characterized by dysfunctional cholesterol efflux. F and F-1, HIV-1 primes the inflammasome and leads to activation of NF-κβ (nuclear factor kappa B), activation of caspase-1, and the expression of IL (interleukin)-1β. While the picture depicts plasma cells, innate lymphoid cells (ILCs), dendritic cells (DCs), and natural killer (NK) cells, they have not been studied among PLWH. CVD indicates cardiovascular disease; HLA-DR, human leukocyte antigen-DR; MHC, major histocompatibility complex; TLR, toll-like receptor; and TRIF, TIR-domain-containing adapter-inducing interferon-β. Illustration credit: Sceyence Studios.
INNATE IMMUNE SYSTEM
Monocytes/Macrophages
The central role of monocytes/macrophages in atherosclerosis is well-established in the general population. Of all the different monocytes, nonclassical monocytes are more permissive to HIV infection.94 Like other inflammatory disorders, PLWH not on treatment have significantly higher proportions of intermediate and nonclassical monocytes.95(p14),96(p14) While ART reduces the level of inflammation observed in patients, longitudinal studies have shown that nonclassical monocytes remain elevated.95 In PLWH, several factors contribute to changes in the constitution of different monocyte subsets and their activation. In 2012, Funderburg et al showed that PLWH with and without treatment had higher proportions of both intermediate and nonclassical monocytes compared with people without HIV. Importantly, they showed that people without HIV with acute coronary syndrome and PLWH with viremia >400 had similar proportions of tissue factor (TF)+ monocytes. Furthermore, the increased expression of prothrombic markers (TF and CD62P) in PLWH appeared to be partly driven by both HIV and lipopolysaccharide.97–99 However, in a separate study with predominantly male participants on ART and undetectable, TF+ monocytes were not associated with carotid intima-media thickness (CIMT) as their CVD end point.100
Due to limitations in mechanistic studies with human study participants, biomarkers such as sCD163 and sCD14 have been heavily relied on as indicators of monocyte/macrophage activation and have been shown to be associated with multiple outcomes, including arterial inflammation, CIMT, and coronary plaque burden, among others (Table 4). Due to differences in study participants and known and unknown confounders, which include traditional CVD risk factors, the relationships between plasma biomarkers and cardiovascular outcomes are not always consistent.
Table 4.
Role of Monocytes/Macrophages in CVD Among PLWH
Mouse models have provided different insights into the possible role of monocytes/macrophages in HIV-1–related atherosclerosis. HIV-1 Tg26 (transgene insertion 26) transgenic mice generated on an inbred laboratory mouse strain (FVB/N [friend virus B/NIH]) background were crossed with ApoE−/− mice on the C57BL/6 background. The HIV-1 DNA introduced into these mice lacks the gag and pol genes with no productive HIV replication. However, these Tg26+/−/ApoE−/− mice express HIV-1 transcripts in the bone marrow and thymus. Tg26+/−/ApoE−/− mice developed accelerated atherosclerosis with a larger plaque area on a normal chow diet for 8 weeks compared with ApoE−/− mice. HIV increased the caspase-1 activity observed in the Tg26+/−/ApoE−/− mice with increased expression of the NLRP3 (NLR family pyrin domain containing 3) inflammasome and serum IL-1β (Table 5).114
Table 5.
Innate Immune Receptors and HIV
Macaque models have also played a pivotal role in advancing our understanding of human diseases, including HIV and cardiometabolic diseases, offering a closer approximation to human physiology than mouse models.115,116 The ability of macaques to be infected with simian immunodeficiency virus, a close relative of HIV, provides a unique opportunity to study immune responses, viral replication, and disease progression in a system that more closely mimics infection in humans.117 In a study of macaques infected with SIV and allowed to develop acquired immune deficiency rapidly, increased monocyte and macrophage activation appeared to be linked to the codevelopment of encephalitis and cardiac inflammation in macaques. In brief, the infected macaques that developed both SIV-related encephalitis and CVD had higher levels of plasma sCD163 and IL-18 and had a higher proportion of cardiac macrophages (CD68+, CD163+, CD206+, and MAC387+). There were no differences in total CD3+ T cells within the tissue.118 Notable, PLWH with HIV encephalitis were similarly found to have increased cardiac inflammation and fibrosis with more CD68+, CD163+, and MAC387+.
Monocytes and CHIP
CHIP is characterized by mutations in genes such as DNMT3A, TET2, JAK2, and ASXL1. These mutations confer a selective advantage to hematopoietic cells, leading to clonal expansion, and have been associated with an increased risk of atherosclerotic CVD due to its proinflammatory effects.119–123 Murine models have previously shown that accelerated development of atherosclerosis is an essential factor contributing to elevated cardiovascular mortality in the presence of clonal hematopoiesis. This acceleration is attributed to enhanced expression of IL-6/IL-1β and stimulation of the NLRP3 inflammasome–triggered endothelial damage. The enhanced inflammatory interactions between the endothelium and macrophages originate from circulating clonal monocytes.119,120,123 In hyperlipidemic mice, CHIP increases IL-6/IL-1β expression and the progression of atherosclerosis.124
While CHIP is commonly associated with aging, its presence in individuals with HIV introduces a unique dimension, potentially intensifying the proinflammatory environment inherent to HIV.125,126 Bick et al125 demonstrated a 2-fold increase in the prevalence of CHIP among PLWH with increased mutations of the ASXL1 gene. In this study, HIV was the second strongest factor, after age, linked to elevated CHIP prevalence among PLWH.125 These findings were consistent with findings from an Australian cohort where age was related to increased prevalence of CHIP among PLWH. Although mutations were observed in DNMT3A and TET2, the greatest mutations were reported in ASXL1 in this population. This cohort also reported an increase in the inflammatory biomarker IL-6.126 Notably, among PLWH from the REPRIEVE trial, participants from sub-Saharan Africa, South Asia, and Latin America/Caribbean had lower odds of CHIP. Smoking and age were associated with increased odds of CHIP.127 The ongoing inflammatory conditions in HIV may foster clonal dominance of specific mutated hematopoietic stem cells, potentially explaining the higher occurrence of clonal hematopoiesis in individuals with HIV. Furthermore, the weakened immune response during acute HIV or the persistent inflammation and immune activation in chronic HIV could impede the body’s ability to survey and eliminate these clonal hematopoietic stem cell populations, further contributing to the elevated prevalence of CHIP in PLWH.126 The chronic immune activation and inflammation associated with HIV, combined with the potential inflammatory effects of CHIP, may potentiate CVD risk in this population.
Dendritic Cells
DCs play a crucial role in bridging the innate and adaptive immune responses through their function of antigen capturing and presentation.128 DCs can be classified as conventional DCs/myeloid DCs and plasmacytoid DCs (pDCs) in blood and Langerhans cells in tissues.129 Each of these subsets has different phenotypes and functions.130 pDCs have proinflammatory roles as they produce ≈95% of type 1 IFNs in peripheral blood mononuclear cells in response to infection. Under noninflammatory conditions, DCs from healthy arteries likely present self-antigens, leading to T-cell tolerance. However, in atherosclerotic lesions, the maturation of plaque DCs in the presence of TLR (toll-like receptor) agonists, danger-associated ligands, and proinflammatory cytokines favors a proatherogenic activation status.131,132
In vitro studies show that conventional DCs and pDCs are both susceptible to HIV. HIV infects DCs, facilitating viral spread to memory CD4+ T cells.129,133 Conventional DCs from PLWH exhibit reduced levels and compromised function, which ART does not restore. HIV-1 may directly and indirectly activate pDCs, resulting in the production of IFN-α. The recognition of HIV ssRNA by TLR7 in pDCs activates the MYD88 pathway, stimulating IRF7 and IFN-α secretion (Table 5).112 Disease progression is, however, associated with a decrease in pDCs and a loss of their ability to produce IFN-α.112 While IFN-α controls virus spread, its continuous release can trigger immunosuppressive pathways, immunopathogenesis, and immunodeficient syndromes. The alternative TLR7-NF-κβ (nuclear factor kappa B) pathway also induces type I IFN production, although in lower amounts than lipopolysaccharide-stimulated DCs.134
It is important to note that, to our knowledge, no direct link has been established in the role of DCs in the progression of atherosclerosis in PLWH. However, HIV infection of DCs and subsequent activation may be important in atherosclerosis progression (Table 6). Further studies are needed to explore this potential connection.
Table 6.
C-Type Lectin Receptor and Their Significance in HIV and Coinfections
Hypercoagulability
HIV induces a chronic inflammatory response, leading to endothelial dysfunction and activation of the coagulation cascade.149 The virus may also directly infect endothelial cells, further contributing to a prothrombotic state. Activated endothelial cells may promote platelet activation, inflammatory reactions, and progression of atherosclerosis.150,151 As discussed in previous sections, persistent inflammation due to chronic HIV can lead to increased levels of proinflammatory cytokines, such as IL-6 and hs-CRP (high-sensitivity C-reactive protein), and other mediators, which, in turn, activate the coagulation system and increase the risk of CVD.2,5,9,150 D-dimer, a byproduct of the coagulation cascade and a marker for inflammation, is elevated in HIV. Research has indicated that it shares comparable associations with CVD and all-cause mortality, such as IL-6 and hs-CRP.4,150,152
Increased levels of circulating D-dimer, minor rises in blood coagulation, thrombin formation, and the turnover of cross-linked intravascular fibrin in PLWH suggest potential relevance to coronary heart disease and peripheral atherosclerosis.153 Additionally, D-dimer values could serve as indicators of inflammatory states.154 In a more recent study, the MACS study, in both untreated and treated PLW, elevated IL-6, ICAM-1, and D-dimer levels were associated with decreased small HDL particle number.75 Another subanalysis of the Strategic Management of ART (Antiretroviral Therapy) (SMART) cohort reported an increased risk of fatal CVD events with an increase in D-dimer score.152 The significant association of baseline D-dimer with the severity of CVD outcomes may reflect the activation of coagulation systems in response to low-grade inflammation associated with HIV infection.
Additionally, HIV has been associated with various changes in coagulation factors, such as an increase in procoagulant factors such as anti-phospholipid antibodies, a decrease in anticoagulant factors such as deficiencies in protein C or S, and heightened platelet activation.155–159 Some of these changes are similar to those seen in acute coronary syndrome.160 HIV and immune activation may also enhance the expression of TF on endothelial cells or leukocytes, thereby activating the extrinsic coagulation pathway.161,162 Previous studies have revealed higher TF expression on circulating blood mononuclear cells in PLWH than in uninfected controls. This study observed correlations between TF expression, HIV RNA levels, the proportion of CD8+ T cells expressing immune activation markers, and D-dimer levels.162 Based on their findings, direct activation of monocytes by microbial products (eg, via bacterial translocation from the gut) may be a key player in promoting thrombosis in HIV.
Innate Lymphoid Cells and Unconventional T Cells
Innate lymphoid cells (ILCs) include the noncytotoxic group 1 (ILC1), group 2 (ILC2), and group 3 (ILC3) and cytotoxic-like conventional natural killer cells.163,164 These cells are characterized by their lymphoid morphology, inability to undergo clonal expansion when stimulated and lack of antigen-specific receptors such as the BCR (B-cell receptor) and TCR (T-cell receptor).165,166 Recent studies have implicated ILCs, especially ILC1s and ILC3s, in modulating the inflammatory milieu within atherosclerotic plaques. ILCs can express proinflammatory cytokines, such as IFN-γ and IL-17A, thereby influencing the recruitment and activation of other immune cells in the atherosclerotic microenvironment.167 Some studies suggest a protective function, and others propose a pathogenic role through the production of proinflammatory cytokines such as IFN-γ and IL-17A.168 Vu et al168 demonstrated that in ApoE−/− mice, IL-17A may contribute to the proatherogenic role of γδ T cells, as many aortic γδ T cells in this study expressed IL-17A, but not IFN-γ.113 γδ T cells are abundant between the intima and atherosclerotic plaque and constitute about 20% of the T-cell population.169 These cells can recognize stress-induced antigens within atherosclerotic plaques in the early stages.
ILC1s contribute to the inflammatory cascade by activating macrophages and DCs. This activation fosters the formation of foam cells and exacerbates endothelial dysfunction, ultimately promoting plaque destabilization.170 On the other hand, ILC3s, with their production of IL-17A and IL-22, further intensify the inflammatory environment within arterial walls. IL-17 promotes the recruitment of neutrophils and macrophages, while IL-22 exhibits both protective and pathogenic effects.171(p3)
In atherosclerotic-prone murine populations, ILC2 expansion has been suggested to have an atheroprotective role. Results from one study highlighted that ILC2-derived IL-5 and IL-13 are important contributors to this regulatory mechanism.172 The atheroprotective effects of IL-5 were confined to the thoracic aorta, and the production of IL-5 and IL-13 by other cell types failed to compensate for the absence of these ILC2-derived cytokines. Furthermore, it was observed that IL-5–dependent atheroprotection could not be attributed to alterations in macrophage phenotype or B cells (B1-dependent) natural IgM production. IL-13–dependent atheroprotection was linked to significant changes in collagen deposition and macrophage phenotype, suggesting a potential involvement of alternative activation pathways. However, the direct connections between changes in macrophage phenotype and atheroprotection were not explicitly addressed in the study.172
Among PLWH, a study by Kløverpris et al173 showed that all ILCs (ILC1-3) were depleted from the peripheral blood at the time of acute infection, with the transcriptional analysis pointing to death due to a strong response to IFNγ and gut inflammation. If ART was initiated at the time of the acute infection, then there was preservation of ILC. Otherwise, the ILCs were irreversibly depleted. Notably, they found that the remaining ILCs were activated and did not migrate into tissues.
Trained Innate Immunity and HIV
PLWH have changes in cells of the innate immune system consistent with reprogramming, likely due to constant exposure to HIV and chronic inflammation from HIV. Specifically, one study found that monocytes from PLWH expressed higher levels of IL-1β when exposed to certain stimuli ex vivo (eg, E coli lipopolysaccharide and imiquimod). However, this increased sensitivity was not seen with lymphocyte activation.174 Trained immunity involves both transcriptional and metabolic reprogramming of innate immune cells. In addition to myeloid cells, other cells can develop trained immunity, such as natural killer cells (by CMV), ILCs, and stromal and epithelial stem cells.175 Many reviews synthesizing the literature on infections and CVD risk propose the role of trained immunity in atherosclerosis. Further research is needed in this area as new therapeutic targets are studied.176
ADAPTIVE IMMUNE SYSTEM
T cells and B cells, present in both the adventitia of healthy arteries and atherosclerotic lesions, are recruited to atheroma by cells of the innate immune system and play a crucial role in the pathogenesis of this chronic inflammatory disease.177,178 Chemokine receptors, such as CCR5 and CXCR6, mediate the infiltration of T cells into plaques, with CCL5 and CXCL16 serving as key chemotactic signals. Notably, blocking the CCL5-CCR5 interaction leads to reduced infiltration of effector CD4+ T cells into aorta explants, underscoring the importance of these interactions in atherosclerosis progression.179,180
T Cells
T cells are present in all stages of atheroma progression, and clonal T cells with oligoclonal TCRs are enriched in unstable plaques.181,182 The presence of oligoclonal TCRs suggests that T-cell expansion is antigen-driven and not stochastic. Antigens that have been implicated in CVD include autoantigens, such as oxidized LDL (low-density lipoprotein),183 ApoB, the core protein of LDL,184 heat-shock protein 60,185 and microbial-derived antigens from pathogens such as CMV.186 Blocking of T-cell activation using antibodies against CD40 ligand reduced the lipid content and size of atherosclerotic lesions and the numbers of T cells and macrophages present in the atheroma.187 Mechanistic studies in mice suggest a role for both CD4136,137 and CD8 T cells in accelerating atherosclerosis.188,189
CD4+ T-cell subpopulations (T helper [TH] 1,190 TH2,191 TH17,192 CD4+ T regulatory [Treg] cells, and cytotoxic T cells) are present in atheroma and influence the different stages of atherosclerosis. TH1 cells, characterized by the expression of the transcription factor T-bet, contribute to plaque development and instability through the secretion of IFN-γ.193 TH2 cells, expressing cytokines such as IL-4, IL-5, and IL-13, exhibit varying effects on atherosclerosis. IL-5 induces the production of oxLDL (oxidized low-density lipoprotein)-targeting IgM antibodies, contributing to antibody-dependent clearance of apoptotic cells and dampening inflammation. On the other hand, IL-13 deficiency accelerates atherosclerosis progression, while low-dose IL-13 stabilizes existing plaques through various mechanisms.179 The role of TH17 cells remains controversial, with evidence supporting both proatherogenic and plaque-stabilizing effects.184,194,195 T follicular helper cells, known for their role in humoral immunity, are implicated in atherosclerosis, particularly in regulating antibody isotype switching and affinity maturation.196 Conversely, Tregs, including FOXP3+ (forkhead box P3) Treg cells and IL-10+ type 1 regulatory T cells, are thought to exert an antiatherogenic.197 Treg cell depletion is associated with increased lesion size and proatherogenic shifts in the plasma cholesterol profile, emphasizing their regulatory role in vascular inflammation.18,180 Studies on atherosclerosis regression models indicate that the restoration of plaque Treg cell numbers is associated with an anti-inflammatory microenvironment, supporting tissue repair and dampening inflammation through enhanced efferocytosis and proresolving lipid mediators.131
Among PWLH, several studies suggest a role for T cells in the pathogenesis of CVD. HIV primarily infects CD4+ T cells, weakening the immune defenses of the host.198 PLWH with untreated and treated HIVs have higher levels of activation of both CD4+ and CD8+T cells, characterized by CD38 and HLA-DR coexpression.43,44,199,200 A study on PLWH on ART (viral load <400 copies/mL) and on rosuvastatin with fasting LDL cholesterol <130 mg/dL showed an association between activated CD8+ T cells (CD38+ HLA-DR+) and common carotid artery-intima media thickness (CCA-IMT).104 Other studies have also shown an association between CD8+ T-cell activation in PLWH on ART with virological suppression,47,201,202 CD4+ T-cell activation,199,200 and T-cell senescence (CD4 and CD8) with subclinical atherosclerosis.47,199,201 In a large study from the Veterans Aging Cohort Study, atherosclerotic CVD was associated with CD4+ T cells (T effector memory expressing RA) and TH17 cells. Incident CVD, on the other hand, has been associated with CD45RA+ CD4+T cells (T effector memory expressing RA), senescent cells, and TH17 cells.203 Notably, this study found an increased association between CD4+ Treg and the ASCVD risk, which could have been a compensatory increase with systemic activation.203
However, another notable difference in T cells by HIV is the inflation of CMV-specific CD4+ T cells, which is not evident in transplant patients and appears specific to PLWH. The inflation of anti-CMV–specific CD4+ T cells persists in PLWH on ART and may be driven by the patient’s HLA type.29 CMV-specific T cells in PLWH are associated with subclinical atherosclerosis.204 Furthermore, they express high levels of inflammatory markers (IFN-γ and TNF-α [tumor necrosis factor-α]) and can induce the expression of CX3CL1 (C-X3-C motif chemokine receptor 1) on the endothelium.205 In a study done in rhesus macaques and older PLWH, they found increased CX3CL1 and IL-15 expression, which was necessary for the recruitment of CX3CR1+ CD8+ T cells into atherosclerotic plaques.206 Several studies have shown an association between senescent T cells (CD28- CD8+ T cells) and subclinical atherosclerosis.207 A subset of CX3CR1+ CD4+ T cells coexpressing markers related to late activation/senescence (CD57) and cell adhesion (GPR56) is also associated with subclinical atherosclerosis, and immunohistochemistry of coronary arteries placed these cells within the adventitia and perivascular adipose tissue.208 Notably, late fibroatheroma from PLWH donors has been shown to have a lower percentage of CD4+ T cells in the coronary artery compared with HIV-negative.208 A relatively large cross-sectional study that combined male and female participants from the WIHS and MACS study found that among 1931 PLWH compared with 859 people without HIV, having a low CD4+ T-cell count was associated with a higher risk of developing subclinical atherosclerosis (1.74× higher in male participants and twice the risk in female participants).209
B Cells
B cells exhibit various functions that might modulate the immune response in atherosclerosis, including antigen presentation, cytokine production, and lymphoid-tissue organization.210 Unlike T cells, B cells exhibit a distinct distribution within atherosclerotic lesions. While only few B cells are detectable locally within the atheroma, a significant number is found in the adventitial layer of atherosclerotic vessels, forming structures resembling tertiary lymphoid organs.211 These B cells within atherosclerotic lesions display oligoclonality and undergo antigen-driven proliferation, indicating a Th-cell–dependent, delayed immune response resulting in high-affinity antibodies, including IgM, IgG, IgE, and IgA.212
The role of B cells in atherosclerosis remains controversial, with some subsets being atheroprotective while others being atherogenic.213,214 For example, follicular B cells may exacerbate atherogenesis by initiating germinal center responses and producing IgG antibodies aimed at oxLDL, a process associated with CVD.215 Furthermore, these follicular B cells participate in the creation of tertiary lymphoid organs in the tunica adventitia of blood vessels, which are also impacted by atherosclerosis, leading to the formation of germinal center and in situ production of anti-oxLDL IgG.216,217
Marginal zone (MZ) B cells, on the other hand, are thought to be atheroprotective, as they inhibit the function of follicular helper T cells in the germinal center through PD-1 (programmed death-1)/PD-L1 (programmed death-ligand 1) interactions.218,219 Interestingly, this protective function depends on the expression of NR4A1 (nuclear receptor subfamily 4 group A), with its deletion in mice exacerbating the development of atherosclerosis due to a reduction in PD-L1.220 Furthermore, unlike IgG, anti-oxLDL IgM is seen as protective against atherosclerosis.214 Given that MZ B cells are significant producers of IgM,215,221 this could be linked to their protective role against atherosclerosis.
B-cell dysfunction and dysregulation have been well reported in HIV,222–224 and this has been associated with the progression of HIV. However, few studies have demonstrated the role of B cells in the development and progression of atherosclerosis in PLWH. Targeting BAFF has previously been shown to reduce the progression of atherosclerosis.225 BAFF is a cytokine involved in the selection and activation of the MZ population and is also important in B-cell survival and differentiation of innate cells.226 BAFF is elevated in PLWH and is correlated with viral factors, Nef, and markers associated with inflammation.227 A Montreal cohort involving PLWH on ART for 15 years reported elevated levels of BAFF in this population. At the baseline, low expression of BAFF is associated with higher expression of Breg markers on MZ B cells. However, PLWH on ART 15 years had reduced expression of Breg markers on a subset of MZ B cells associated with subclinical atherosclerosis.228
OXIDATIVE STRESS AND LIPID OXIDATION
Oxidative stress occurs when there is an imbalance between the production of reactive oxygen species and the body’s ability to neutralize them with antioxidants.229 This imbalance can lead to the oxidation of lipids, including LDL cholesterol, a major contributor to the development of ASCVD. In HIV, oxidative stress is increased due to chronic inflammation and immune activation, which leads to the production of reactive oxygen species by activated immune cells.230 These reactive oxygen species can damage lipids, proteins, and DNA, leading to a wide range of cellular dysfunction.230 Additionally, ART has been shown to increase oxidative stress and lipid peroxidation. This is thought to be due to the interaction between ART and mitochondrial function, which can lead to the production of reactive oxygen species and cellular damage.
Lipid oxidation, specifically the oxidation of LDL cholesterol, has been implicated in the development of ASCVD in HIV. Oxidized LDL cholesterol is taken up by macrophages in the arterial wall, leading to the formation of foam cells and the initiation of the atherosclerotic process.231 In HIV, oxidized LDL cholesterol levels are increased, and this is thought to be due to the combination of chronic inflammation, immune activation, and ART-induced oxidative stress.232
GUT TRANSLOCATION
The gastrointestinal tract experiences significant CD4+ T-cell depletion throughout all phases of HIV disease and is a significant HIV reservoir.233 This reduction predominantly affects the CCR5+ CD4+ T-cell subset, constituting the majority of CD4+ T cells in the gastrointestinal tract.234 A loss of CD4+ T cells in the gastrointestinal tract disrupts the delicate balance of the intestinal mucosal barrier with increased gut permeability, facilitating the translocation of microbial products, such as bacterial lipopolysaccharide, into the systemic circulation.35,235 Microbial translocation has been linked to systemic inflammation because the presence of microbial products in the bloodstream activates immune cells, initiating a chronic inflammatory state.236 In PLWH, a low CD4+ T-cell count is associated with higher plasma lipopolysaccharide.32
POSSIBLE ROLE OF COINFECTIONS
Chronic HIV stands out among infectious diseases due to its association with coinfections. The HIV virus weakens the immune system, increasing vulnerability to exogenous pathogens.237 These coinfections, such as CMV, Epstein-Barr virus, tuberculosis, hepatitis C and B, and human papillomavirus, may promote the immune dysregulation caused by HIV, even after initiation of ART, creating an environment conducive to several inflammation-associated complications including CVD (Figure 3).239
Figure 3.
Proposed model of how HIV and other chronic persistent viruses maintain the HIV reservoir and drive chronic inflammation contributing to cardiovascular disease (CVD). HIV-1 reservoir cells consist of activated cluster of differentiation (CD)4+ T cells (predominantly cytotoxic, such as CGC+)218,219 that are resistant to killing by CD8+ T cells and natural killer (NK) cells.238 CD4+ T cells specific for Epstein-Barr virus (EBV) are stimulated by memory B cells that constitutively express latent EBV proteins and present them on MHC class II. Cytomegalovirus (CMV) expression by activated macrophages and endothelial cells can also activate T cells that express TCRs (T-cell receptors) that bind CMV epitopes, leading to the secretion of inflammatory cytokines that perpetuate the state of chronic immune activation. CCR5 indicates chemokine receptor type 5; IL, interleukin; IFN, interferon; MHC, major histocompatibility complex; and TNF-α, tumor necrosis factor-alpha. Illustration credit: Sceyence Studios.
Cytomegalovirus
CMV is a large herpes DNA virus that infects vascular endothelial cells.240,241 It induces memory CMV-specific T cells that increase with age irrespective of HIV status.242–244 Among octogenarians, CMV IgG seropositivity in the general population has been associated with increased mortality from CVD.245 In PLWH, CMV seropositivity has been associated with an increased risk of non-AIDS–defining events and CVD/cerebrovascular diseases.246 Furthermore, high CMV antibody titers have been associated with increased carotid intimal thickening in people without HIV.247–250 Similarly, in PLWH, CMV IgG has been associated with reduced carotid artery distensibility and carotid artery lesions.251
In addition to CMV antibody titers, T-cell immune responses against CMV have also been associated with CVD outcomes in PLWH. PLWH mount inflated T-cell responses against CMV with up to 20% of CD4+ T cells against a single CMV epitope in the absence of CMV viremia.29 CD8+ T-cell expansion has also been shown with HIV/CMV coinfection.252 CMV-specific cells express the chemokine receptor, CX3CR1, which traffics immune cells to inflamed endothelium. CX3CR1 is more highly expressed on CD4+ and CD8+ T cells in PLWH compared with people without HIV.77 CX3CR1+ T cells are important in CVD because they can induce fractalkine expression on endothelial cells,253 establishing a positive feedback loop that mediates adhesion and migration of CX3CR1 expressing lymphocytes into the vascular intima.254,255 CX3CR1+ GPR56+ CD57+ CD4+ T cells (CGC+ CD4+ T cells) have been shown to be frequently CMV-specific and have been associated with subclinical atherosclerosis in PLWH.256,257 GPR56 is a surface marker of cytotoxic T cells.258 It is a G-coupled protein receptor expressed by cytotoxic CD4+ T cells, whereas CD57 is a marker of late activation or senescence.258,259 Higher CX3CR1+ CD4+ T cells and CMV-specific CD8+ T cells have been associated with greater carotid intima thickness204,205 possibly due to increased vascular homing.260
Initial studies suggested that treating CMV with antiviral therapies could serve as an important CVD risk reduction intervention if CMV is a risk factor for atherosclerosis. A small randomized clinical trial with 30 PLWH treated with valganciclovir (n=14) or placebo (n=16) for 8 weeks showed a significant reduction in CMV DNA in treated participants compared with placebo. This intervention reduced inflammation, which is an important contributor to CVD.261 There is a more recent clinical trial by the AIDS Clinical Trials Group (A5383 trial). A study in patients with antineutrophil cytoplasmic antibody–associated vasculitis showed that treatment with valacyclovir reduced CMV reactivation and the proportion of CD28- CD4+ T cells.262 The reduction in CD28- CD4+ T cells was accompanied by improved responses to a pneumococcal vaccine. In a prior study, the same group showed that these cells were also linked to increased arterial stiffness in the same patient population.263 Taken together, these studies suggest that future therapies targeting CMV to reduce the anti-CMV T-cell immune response could have a role in the prevention of atherosclerosis progression in CMV-positive HIV(+) and HIV(−) people.
Hepatitis C Virus
Due to shared transmission routes, HIV/hepatitis C virus (HCV) coinfection is common, with about 20% of PLWH having HCV.264 Like HIV, chronic HCV infection is associated with coronary artery disease even after adjusting for traditional risk factors.265–267 The risk for CVD is 2.9 higher in HIV/HCV coinfected individuals compared with HIV monoinfected individuals.268 Studies suggest a potential mechanism involving higher TF activity and associated CD4+ T-cell activation in HCV patients, highlighting the complex relationship between immune activation, microbial translocation, coagulopathy, and CVD.269 In a study assessing immune activation in HIV/HCV coinfection, PLWH, chronic hepatitis, and cirrhosis had elevated levels of serum sCD14 and IL-6 alongside incomplete CD4 restoration. While the percentage of activated CD4+ and CD8+ T cells and Treg was higher in HIV-infected patients than in healthy controls, no significant differences were noted among different HIV patient groups.270 In a separate study, inflammation biomarkers, including IFN-α, were found to be higher in individuals coinfected with HCV and HIV compared with those with HCV alone. Some of these biomarkers decreased following successful sustained virological suppression of HCV.271 Successful sustained virological suppression of HCV may decrease inflammation and immune activation, potentially mitigating the risk of cardiovascular events.272
Tuberculosis
PLWH with active and latent tuberculosis have increased levels of immune activation, elevated plasma soluble biomarkers of inflammation, such as sCD14, and increased T-cell activation.273–277 These elevated biomarkers are indicative of the progression of HIV disease and are associated with a higher risk of mortality.278,279
PLWH with latent tuberculosis have a higher likelihood of converting to active tuberculosis.275 In a cohort of African adults living with HIV on ART, latent tuberculosis was associated with immune dysregulation and an increase in markers of inflammation (TNF-α, IL-6, IL-12p70, IL-15, and IL-17A).276 Some of these markers, such as IL-17A and TNF-α, may promote vascular and endothelial dysfunction, leading to the development and progression of CVD.167,194,280–282
Herpes Simplex Virus 2
Herpes simplex virus 2 (HSV-2) coinfection may act as a cofactor for increased HIV replication, and the use of acyclovir/valacyclovir to target HSV-2 may have localized benefits on HSV-2 with mucosal level HIV suppression and decreased monocyte activation.283,284 In a study of 291 men living with HIV in the MACS, infection with multiple herpes viruses was found to be associated with elevated coronary arterial calcium. However, HSV-2 seropositivity in this cohort had the strongest association with subclinical atherosclerosis, with a 4-fold increased risk.285 Note that in a different population-based cross-sectional study not specific to HIV, they found that HSV-2 was positively associated with CVD.286
Kaposi Sarcoma and HHV8
Human herpesvirus 8 (HHV8) is implicated in the development and progression of atherosclerosis. Support for this hypothesis is derived from proatherogenic characteristics observed in in vitro studies on the vessel wall.287 Additionally, a postmortem report of AIDS patients revealed a higher incidence of macroscopic atheromatous lesions in individuals with Kaposi sarcoma, providing further evidence for the association between the virus and atherogenesis.288 Vascular endothelial cells infected with HHV8 demonstrate the ability to stimulate the generation of growth factors, leading to angiogenesis, increased proliferation of endothelial cells, heightened vascular permeability, and cytokine production.288
The prevalence of HHV8 seropositivity among PLWH varies geographically, with the highest percentages reported among men who have sex with men.289–291 HHV8 seropositivity is associated with higher levels of CRP and T-cell activation, indicating its potential role in chronic immune activation in chronically treated patients with HIV.292 In a study involving 141 virologically suppressed PLWH on ART, coinfection with HSV-2 and HHV8 was associated with an increase in hs-CRP and progression of CIMT. Notably, in this study, HHV8 exhibited the strongest correlation with the progression of CIMT.293 A separate retrospective study of postmortem samples from PLWH had ≈3.4× more likely to have macroscopic atheroma than the Kaposi sarcoma negative group. Murine studies of APOE−/− mice on a high-fat diet infected with viruses such as HHV8 (MHV-68) had accelerated atherosclerosis, which further supports a possible role for HHV8 in accelerating atherosclerosis in the presence of other CVD risk factors.287
IMPACT OF ART ON CVD
ART has significantly improved the prognosis and quality of life for PLWH. Early ART initiation helps suppress viral replication and restore immune function.294 One of the primary objectives of ART is to suppress viral replication, allowing the immune system to recover. ART initiation at an early stage leads to a substantial reduction in viral load, enabling the restoration of CD4+ T-cell counts, which are crucial for immune function.3,295 Studies have shown an increased risk of CVD in PLWH independent of traditional risk factors,21,296 and some ART have been implicated in this increased risk.297–299 ART can improve endothelial function and reduce biomarkers of cardiovascular risk, but certain therapies, such as abacavir, were proposed to increase inflammatory markers and risk of myocardial infarction. However, controversy persists as the comparator drugs of that era, which are no longer used, are known to have cardioprotective effects.60,294 Combined ART has been reported to have approximately a 26% relative rise in the rate of myocardial infarction in PLWH for each year of exposure within the initial 4 to 6 years of usage.297 ART has also been associated with an increase in metabolic syndrome, a known risk of CVD among PLWH.288
Older ART
Most of the drugs used to treat HIV in this early ART era had either unfavorable or favorable off-target (ie, non-HIV related) effects, confounding the interpretation of these newly emerging clinical manifestations. Broadly, some first-generation protease inhibitors (PIs) were associated with visceral obesity (eg, Crixivan Crix belly) and adverse lipid profiles14 The thymidine analogs zidovudine and stavudine induced adipocyte-specific mitochondrial toxicity, impairing the ability of adipose tissue to trap free fatty acids.15,300 Tenofovir is associated with bone and renal toxicity but favorable lipids and relative CVD protection.301 Efavirenz is associated with anorexia and weight loss, which may mitigate the emergence of metabolic syndrome.302 These drugs have now been replaced by drugs better designed to only bind their target HIV protein without off-target binding to any host proteins. For example, tenofovir alafenamide (TAF) has replaced tenofovir, which does not have bone or renal toxicity or CVD protective effects.301
First generations of antiretroviral drugs, particularly PIs and certain nonnucleoside reverse transcriptase inhibitors, were associated with metabolic complications, including dyslipidemia.298,303 PIs, such as indinavir and lopinavir, are notorious for elevating total cholesterol, LDL cholesterol, and triglyceride levels and are implicated in increased progression of subclinical atherosclerosis.304,305 To promote optimal virological response, PIs require boosting with ritonavir. Ritonavir is, however, associated with increased plasma lipids, insulin resistance,68 endothelial dysfunction,306 and macrophage accumulation of cholesterol.304 Switching from boosted PIs to raltegravir or dolutegravir results in a better plasma lipid profile and changes in biomarkers associated with cardiovascular events than continuing boosted PI.294,307 Older NRTIs, such as stavudine and zidovudine, were also linked to lipid abnormalities. Patients on stavudine were reported to develop a syndrome of short-term fat wasting associated with increased plasma-free fatty acid and triglyceride levels and increased daily fat intake without insulin resistance.308,309
PLWH previously exposed to thymidine analogs and didanosine may experience persisting changes in the distribution of visceral adipose tissue and subcutaneous adipose tissue for many years after discontinuation of therapy.310 In the visceral compartment, thymidine analogs have been associated with increased mitochondrial toxicity in rats, thus affecting adipocyte distribution and leading to pathological adipose accumulation.311 This increases the risk of developing conditions such as hypertension, hypercholesterolemia, and low levels of HDL even years after stopping the treatment.310
Newer Therapies
Newer antiretroviral drugs with improved side effect profiles are available with integrase strand transfer inhibitors gaining prominence. Second-generation integrase strand transfer inhibitors, such as dolutegravir, elvitegravir, and bictegravir, have been well-tolerated and effective in suppressing viral replication. However, an emerging concern has been the association between newer therapies, particularly dolutegravir, and weight gain. Mechanisms of weight gain are poorly understood. However, it has been hypothesized that dolutegravir can directly affect adipocytes, leading to adipogenesis, lipogenesis, oxidative stress, fibrosis, and insulin resistance.312 In recent trials, more significant weight gain has been observed in dolutegravir groups compared with efavirenz.302,313,314 Some evidence suggests that the greater weight gain observed in people taking dolutegravir-based ART compared with those on efavirenz-based ART might be linked to impaired weight gain in individuals with certain genetic traits, that is, loss of polymorphism in cytochrome P450 2B6 enzyme (CYP2B6; CYP2B6 intermediate or slow metabolizer genotypes) as the CYP2B6 primarily metabolizes efavirenz. These individuals likely experienced slow metabolism of efavirenz, leading to toxicity, rather than dolutegravir causing changes in appetite or metabolism.315 Weight gain appears to be most closely related to the antiviral potency of the regimen, and there is no good mechanistic evidence to date that weight gain relates to any off-target effect of any drug or drug class.302
Switching from tenofovir disoproxil fumarate (TDF) to TAF has also been associated with significant weight gain in PLWH.316,317 One of the proposed mechanisms of weight gain is due to the reduction of plasma HIV RNA levels and mitigating the catabolic effects associated with viremia; therefore, a restorative process leading to improved health occurs. As a result, some individuals experience weight gain that exceeds normal, healthy levels.318,319 It is unclear whether TAF independently contributes to weight gain or does not suppress dolutegravir-induced weight gain as TDF does. Although the long-term implications of ART-associated weight gain are not fully understood, limited evidence suggests an increased incidence of hyperglycemia, hypertension, and metabolic syndrome, coupled with an increased risk of cardiovascular complications.320
Another shift in HIV practice has been the immediate rather than deferred initiation of ART after diagnosis based on studies such as Strategic Timing of Antiretroviral Treatment (START) (nadir CD4>600 cells/μL), which showed reduced serious AIDS and non-AIDS events in those treated early.321 In contrast, the SMART study (nadir CD4 <250 cells/μL) found a 70% decrease in the risk of cardiovascular events in those randomized to early rather than deferred or interrupted ART.322 This increased risk correlated with increased proinflammatory markers in the deferred or interrupted group.4
TARGETING THE IMMUNE SYSTEM: BEFORE AND AFTER THE REPRIEVE TRIAL
Low-Dose Methotrexate
Methotrexate, a competitive dihydrofolate reductase inhibitor known for modifying rheumatoid arthritis progression, has shown promise in reducing mortality among PLWH.323 The CIRT (Cardiovascular Inflammation Reduction Trial) was designed to assess whether low-dose methotrexate could prevent CVD events in the general population. There was no reduction in inflammation or CVD effects compared with placebo in this study.324 Another pilot study (clinical trial number: NCT01949116) that evaluated the safety and impact of low-dose methotrexate on endothelial function in PLWH reported no significant effect on endothelial function or inflammatory biomarkers with low-dose methotrexate. There was, however, a significant decrease in CD8+ T cells. This study was associated with more safety events in the methotrexate group (Table 7).325
Table 7.
Targeting Immune Activation and Inflammation to Decrease CVD Risk Among PLWH
IL-1β Antagonists
Canakinumab is a monoclonal antibody against IL-1β that was first shown to reduce inflammation and improve CVD events independent of lipids.324 Notably, participants in that study (HIV-negative) received 3 doses of the antibody, and they observed a higher rate of fatal infections than in the placebo group. Among PLWH, a single dose of canakinumab resulted in reduced arterial inflammation measured by fluorodeoxyglucose/positron emission tomography/CT, as well as reduced levels in circulating inflammatory markers, including IL-6 and sCD163. There was no difference in the proportion of activated T cells and monocytes.335 IL-1 antagonism has also been shown in smaller studies to improve heart failure through the reduction of systemic inflammatory response.345,346
TNF-α Antagonists
There have been mixed outcomes in the general population with TNF-α antagonists in heart failure trials.347,348 No TNF-α has been done among PLWH.
IL-6 Antagonist
Tocilizumab is a monoclonal antibody against IL-6 and, in a crossover clinical trial among PLWH, has been shown to decrease markers of circulating inflammatory markers, including sCD14, sCD40L, d-dimer, p-selectin, and sTNFR1/2. Notably, there was an increase in sCD163, but this was not significant. Like a previous study among people without HIV, there was an increase in LDL cholesterol, HDL cholesterol, and Lp-PLA2 (lipoprotein-associated phospholipase A2).334 The increase in lipid classes was not altered by statin treatment. Clinically, there was no improvement in endothelial function as measured by brachial artery flow–mediated dilation.334
Antiplatelet Agents
Aspirin, a traditional cardiovascular prevention treatment, has shown promise in reducing immune activation in PLWH on ART. Aspirin inhibits cyclooxygenase, which is in the pathway that leads to the formation of prostaglandins, downstream of which is the induction of inflammatory cytokines, including IL-6 and IL-1β.349 One study observed that low-dose aspirin attenuated sCD14 and activated T cells in ART-treated PLWH.350 A longer 12-week randomized controlled trial showed no effects of high or low-dose aspirin on monocyte activation (sCD14 and sCD163), proatherogenic monocyte subsets, T-cell activation, and endothelial dysfunction.326 Ongoing trials are exploring aspirin’s immune and vascular effects in PLWH. However, the potential benefits of primary prevention must be weighed against bleeding risks.
RAAS Regulation
Drugs targeting the RAAS (renin-angiotensin-aldosterone system) may also reduce CVD events by mitigating inflammation and tissue fibrosis pathways. Limited trials have explored the effects of lisinopril and telmisartan in PLWH subjects, showing potential benefits in reducing visceral adiposity, hs-CRP, and TNF-α.238,330 No adipocyte fibrosis reduction is seen in telmisartan therapy in addition to ART.329 A large retrospective study by Klein et al351 indicated a declining risk of myocardial infarction over time in HIV-infected individuals, correlating with increased prescriptions for lipid-lowering and antihypertensive treatments. One study on the impact of losartan on inflammation and tissue fibrosis found that losartan had no effect on carotid intima thickness, T-cell activation, markers of inflammation, or monocyte differentiation in PLWH on ART.328
Omega-3 Fatty Acids
Omega-3 fatty acids, while modestly improving systemic inflammation in PLWH by decreasing levels of IL-6 and TNF-α, do not seem to significantly affect endothelial function based on one small study.352
Statins
Statins, known for their lipid-lowering and potential anti-inflammatory properties, have shown promise in preventing CVD in PLWH.353 Studies on atorvastatin and rosuvastatin demonstrated reductions in noncalcified plaque volume, high-risk coronary plaque features, and slowed progression of CIMT.339 The Study to Assess the Tolerability and Anti-Retroviral Activity of MK-0518 (SATURN-HIV) trial further highlighted the role of statins in reducing T-cell activation and innate immune activation markers.339 The large REPRIEVE focused on the use of a statin, pitavastatin, to reduce cardiovascular events in PLWH. While the primary end point of the trial was CVD prevention, substudies within REPRIEVE explored the impact of pitavastatin on immune activation and reported a reduced risk of a major adverse cardiovascular event compared with those who received a placebo.17 A small randomized, double-blind study on the effects of pitavastatin on atherosclerotic-associated inflammatory biomarkers when administered with PI ritonavir-boosted atazanavir in PLWH with dyslipidemia demonstrated that pitavastatin lowered the proportion of activated T cells (HLA-DR+ CD38−CD4+) and programmed cell death-protein PD1+ CD4+ T cells, thus reducing immune activation.354 There was also an increase in basic fibroblast growth factor, which has atherosclerotic-protective effects.
PCSK9 Inhibitors
PCSK9 (proprotein convertase subtilisin/kexin type 9) plays a vital role in cholesterol metabolism through modulation of the expression of LDL receptors, which bind and remove LDL cholesterol355 and predict future cardiovascular risk.356 PLWH with or without dyslipidemia have been reported to have elevated levels of PCSK971 associated with foam cell formation and endothelial dysfunction in these individuals.71 Leucker et al357 showed that rapid improvement in coronary endothelial function in PLWH treated with PCSK9 inhibitor was associated with reduced Lp(a) (lipoprotein[a]) and not LDL cholesterol. PCSK9 antibodies (PCSK9i) have also shown effectiveness in reducing cardiovascular events in individuals with coronary artery disease who are intolerant to statin therapy.358 In a small study involving 11 PLWH with dyslipidemia not diagnosed with coronary artery disease and 19 PLWH with nearly optimal or above goal low-density lipoprotein cholesterol levels, the inhibition of PCSK9 with evolocumab demonstrated an improvement in abnormal coronary endothelial function.344 In this study, no improvement was reported in serum inflammation biomarkers. Another study is underway and will shed more light on the effects of PCSK9 inhibition with alirocumab on vascular inflammation, endothelial function, and noncalcified plaque in ART-treated participants.72
CONCLUSION AND FUTURE DIRECTION
The persistent dysregulation of both adaptive and innate immunity in chronic HIV infection, in the presence of clinically suppressed viral load through effective ART, underscores the complexity of immune activation dynamics. ART reduces but does not normalize immune activation. While the specific factors that drive heightened immune activity may vary among individuals, the resultant systemic inflammation significantly contributes to the amplification of well-established proatherogenic mechanisms. This intricate interplay between chronic HIV infection, immune activation, and ASCVD emphasizes the need for targeted therapeutic approaches. Epidemiological and mechanistic research has enhanced our understanding of the extent and drivers of CVD in the general population and among PLWH. Nevertheless, several gaps in the literature need to be investigated further to help improve patient outcomes (Table 8).
Table 8.
Gaps in Literature and Future Directions
ARTICLE INFORMATION
Acknowledgments
Images were created using BioRender.
Sources of Funding
L.M. Obare was supported by the Gilead HIV Scholars Research Program. T. Temu was supported by grant K01HL147723. S.A. Mallal was supported by the Tennessee Center for AIDS Research grant P30 AI110527. C.N. Wanjalla was supported by the Doris Duke CSDA 2021193 (grant K23 HL156759), the Burroughs Wellcome Fund grant 1021480, and the Gilead HIV Scholars Research Program.
Disclosures
None.
Nonstandard Abbreviations and Acronyms
- ABCA1
- ATP-binding cassette transporter A1
- ApoB
- apolipoprotein B
- ART
- antiretroviral therapy
- ASCVD
- atherosclerotic cardiovascular disease
- BCR
- B-cell receptor
- CCL2
- C-C motif chemokine ligand 2
- CD
- cluster of differentiation
- CHIP
- clonal hematopoiesis of indeterminate potential
- CIMT
- carotid intima-media thickness
- CIRT
- Cardiovascular Inflammation Reduction Trial
- CLR
- C-type lectin receptor
- CMV
- cytomegalovirus
- CVD
- cardiovascular disease
- DC
- dendritic cell
- DC-SIGN
- dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin
- eNOS
- endothelial nitric oxide synthase
- gp
- glycoprotein
- HCV
- hepatitis C virus
- HDL
- high-density lipoprotein
- HHV8
- human herpesvirus 8
- HSV-2
- herpes simplex virus 2
- hs-CRP
- high-sensitivity C-reactive protein
- ICAM-1
- intercellular adhesion molecule 1
- IDL
- intermediate-density lipoprotein
- IL
- interleukin
- ILC
- innate lymphoid cell
- IFN
- interferon
- LDL
- low-density lipoprotein
- Lp(a)
- lipoprotein(a)
- Lp-PLA2
- lipoprotein-associated phospholipase A2
- MACS
- Multicenter AIDS Cohort Study
- MBL
- mannose-binding lectin
- MCP-1
- monocyte chemoattractant protein 1
- M-CSF
- monocyte-colony stimulating factor
- MMP2/9
- matrix metalloproteinase 2/9
- MZ
- marginal zone
- Nef
- negative regulatory factor
- NLRP3
- NLR family pyrin domain containing 3
- PCSK9
- proprotein convertase subtilisin/kexin type 9
- pDC
- plasmacytoid dendritic cell
- PI
- protease inhibitor
- PLWH
- people living with HIV
- RAAS
- renin-angiotensin-aldosterone system
- REPRIEVE
- Randomized Trial to Prevent Vascular Events in HIV
- sICAM-1
- soluble intercellular adhesion molecule 1
- SIV
- simian immunodeficiency virus
- sVCAM-1
- soluble vascular cell adhesion molecule-1
- Tat
- transactivator of transcription
- TF
- tissue factor
- TH
- T helper
- TLR
- toll-like receptor
- TNF-α
- tumor necrosis factor-α
- Treg
- T regulatory
- VLDL
- very low-density lipoprotein
For Sources of Funding and Disclosures, see page 1535.
REFERENCES
- 1.Shah ASV, Stelzle D, Lee KK, Beck EJ, Alam S, Clifford S, Longenecker CT, Strachan F, Bagchi S, Whiteley W, et al. Global burden of atherosclerotic cardiovascular disease in people living with HIV: systematic review and meta-analysis. Circulation. 2018;138:1100–1112. doi: 10.1161/CIRCULATIONAHA.117.033369 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Nou E, Lo J, Grinspoon SK. Inflammation, immune activation, and cardiovascular disease in HIV. AIDS. 2016;30:1495–1509. doi: 10.1097/QAD.0000000000001109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.French MA, King MS, Tschampa JM, da Silva BA, Landay AL. Serum immune activation markers are persistently increased in patients with HIV infection after 6 years of antiretroviral therapy despite suppression of viral replication and reconstitution of CD4+ T cells. J Infect Dis. 2009;200:1212–1215. doi: 10.1086/605890 [DOI] [PubMed] [Google Scholar]
- 4.Kuller LH, Tracy R, Belloso W, De Wit S, Drummond F, Lane HC, Ledergerber B, Lundgren J, Neuhaus J, Nixon D, et al. ; INSIGHT SMART Study Group. Inflammatory and coagulation biomarkers and mortality in patients with HIV infection. PLoS Med. 2008;5:e203. doi: 10.1371/journal.pmed.0050203 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sereti I, Krebs SJ, Phanuphak N, Fletcher JL, Slike B, Pinyakorn S, O'Connell RJ, Rupert A, Chomont N, Valcour V, et al. ; RV254/SEARCH 010, RV304/SEARCH 013 and SEARCH 011 Protocol Teams. Persistent, albeit reduced, chronic inflammation in persons starting antiretroviral therapy in acute HIV infection. Clin Infect Dis. 2017;64:124–131. doi: 10.1093/cid/ciw683 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Temu TM, Zifodya JS, Polyak SJ, Wagoner J, Wanjalla CN, Masyuko S, Nyabiage J, Kinuthia J, Bloomfield GS, Page ST, et al. Antiretroviral therapy reduces but does not normalize immune and vascular inflammatory markers in adults with chronic HIV infection in Kenya. AIDS. 2021;35:45–51. doi: 10.1097/QAD.0000000000002729 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ruggiero A, Cozzi-Lepri A, Beloukas A, Richman D, Khoo S, Phillips A, Geretti AM; ERAS Study Group. Factors associated with persistence of plasma HIV-1 RNA during long-term continuously suppressive firstline antiretroviral therapy. Open Forum Infect Dis. 2018;5:ofy032. doi: 10.1093/ofid/ofy032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Anand AR, Rachel G, Parthasarathy D. HIV proteins and endothelial dysfunction: implications in cardiovascular disease. Front Cardiovasc Med. 2018;5:185. doi: 10.3389/fcvm.2018.00185 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Worm SW, Hsue P. Role of biomarkers in predicting CVD risk in the setting of HIV infection? Curr Opin HIV AIDS. 2010;5:467–472. doi: 10.1097/COH.0b013e32833f2ea6 [DOI] [PubMed] [Google Scholar]
- 10.Brown TR. I am the Berlin patient: a personal reflection. AIDS Res Hum Retroviruses. 2015;31:2–3. doi: 10.1089/AID.2014.0224 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gupta RK, Abdul-Jawad S, McCoy LE, Mok HP, Peppa D, Salgado M, Martinez-Picado J, Nijhuis M, Wensing AMJ, Lee H, et al. HIV-1 remission following CCR5Δ32/Δ32 haematopoietic stem-cell transplantation. Nature. 2019;568:244–248. doi: 10.1038/s41586-019-1027-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Nolan D, Watts GF, Herrmann SE, French MA, John M, Mallal S. Endothelial function in HIV-infected patients receiving protease inhibitor therapy: does immune competence affect cardiovascular risk? QJM. 2003;96:825–832. doi: 10.1093/qjmed/hcg145 [DOI] [PubMed] [Google Scholar]
- 13.Grunfeld C, Feingold KR. Metabolic disturbances and wasting in the acquired immunodeficiency syndrome. N Engl J Med. 1992;327:329–337. doi: 10.1056/NEJM199207303270506 [DOI] [PubMed] [Google Scholar]
- 14.Nolan D, John M, Mallal S. Antiretoviral therapy and the lipodystrophy syndrome, part 2: concepts in aetiopathogenesis. Antivir Ther. 2001;6:145–160. doi: 10.1177/135965350100600301 [PubMed] [Google Scholar]
- 15.John M, Nolan D, Mallal S. Antiretroviral therapy and the lipodystrophy syndrome. Antivir Ther. 2001;6:9–20. doi: 10.1177/135965350100600102 [DOI] [PubMed] [Google Scholar]
- 16.Freiberg MS. HIV and cardiovascular disease - an ounce of prevention. N Engl J Med. 2023;389:760–761. doi: 10.1056/NEJMe2306782 [DOI] [PubMed] [Google Scholar]
- 17.Grinspoon SK, Fitch KV, Zanni MV, Fichtenbaum CJ, Umbleja T, Aberg JA, Overton ET, Malvestutto CD, Bloomfield GS, Currier JS, et al. ; REPRIEVE Investigators. Pitavastatin to prevent cardiovascular disease in HIV infection. N Engl J Med. 2023;389:687–699. doi: 10.1056/NEJMoa2304146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Roy P, Orecchioni M, Ley K. How the immune system shapes atherosclerosis: roles of innate and adaptive immunity. Nat Rev Immunol. 2022;22:251–265. doi: 10.1038/s41577-021-00584-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Currier JS, Taylor A, Boyd F, Dezii CM, Kawabata H, Burtcel B, Maa JF, Hodder S. Coronary heart disease in HIV-infected individuals. J Acquir Immune Defic Syndr. 2003;33:506–512. doi: 10.1097/00126334-200308010-00012 [DOI] [PubMed] [Google Scholar]
- 20.Currier JS, Lundgren JD, Carr A, Klein D, Sabin CA, Sax PE, Schouten JT, Smieja M; Working Group 2. Epidemiological evidence for cardiovascular disease in HIV-infected patients and relationship to highly active antiretroviral therapy. Circulation. 2008;118:e29–e35. doi: 10.1161/CIRCULATIONAHA.107.189624 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Freiberg MS, Chang CCH, Kuller LH, Skanderson M, Lowy E, Kraemer KL, Butt AA, Bidwell Goetz M, Leaf D, Oursler KA, et al. HIV infection and the risk of acute myocardial infarction. JAMA Intern Med. 2013;173:614–622. doi: 10.1001/jamainternmed.2013.3728 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Triant VA, Lee H, Hadigan C, Grinspoon SK. Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease. J Clin Endocrinol Metab. 2007;92:2506–2512. doi: 10.1210/jc.2006-2190 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hoffmann U, Lu MT, Foldyna B, Zanni MV, Karady J, Taron J, Zhai BK, Burdo T, Fitch KV, Kileel EM, et al. ; REPRIEVE Trial. Assessment of coronary artery disease with computed tomography angiography and inflammatory and immune activation biomarkers among adults with HIV eligible for primary cardiovascular prevention. JAMA Netw Open. 2021;4:e2114923. doi: 10.1001/jamanetworkopen.2021.14923 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tseng ZH, Secemsky EA, Dowdy D, Vittinghoff E, Moyers B, Wong JK, Havlir DV, Hsue PY. Sudden cardiac death in patients with human immunodeficiency virus infection. J Am Coll Cardiol. 2012;59:1891–1896. doi: 10.1016/j.jacc.2012.02.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wu KC, Zhang L, Haberlen SA, Ashikaga H, Brown TT, Budoff MJ, D'Souza G, Kingsley LA, Palella FJ, Margolick JB, et al. Predictors of electrocardiographic QT interval prolongation in men with HIV. Heart. 2019;105:559–565. doi: 10.1136/heartjnl-2018-313667 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Nus M, Mallat Z. Immune-mediated mechanisms of atherosclerosis and implications for the clinic. Expert Rev Clin Immunol. 2016;12:1217–1237. doi: 10.1080/1744666X.2016.1195686 [DOI] [PubMed] [Google Scholar]
- 27.Baker JV, Duprez D, Rapkin J, Hullsiek KH, Quick H, Grimm R, Neaton JD, Henry K. Untreated HIV infection and large and small artery elasticity. J Acquir Immune Defic Syndr. 2009;52:25–31. doi: 10.1097/qai.0b013e3181b02e6a [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Torriani FJ, Komarow L, Parker RA, Cotter BR, Currier JS, Dubé MP, Fichtenbaum CJ, Gerschenson M, Mitchell CKC, Murphy RL, et al. ; ACTG 5152s Study Team. Endothelial function in human immunodeficiency virus-infected antiretroviral-naive subjects before and after starting potent antiretroviral therapy: the ACTG (AIDS Clinical Trials Group) Study 5152s. J Am Coll Cardiol. 2008;52:569–576. doi: 10.1016/j.jacc.2008.04.049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Abana CO, Pilkinton MA, Gaudieri S, Chopra A, McDonnell WJ, Wanjalla C, Barnett L, Gangula R, Hager C, Jung DK, et al. Cytomegalovirus (CMV) epitope-specific CD4+ T cells are inflated in HIV+ CMV+ subjects. J Immunol. 2017;199:3187–3201. doi: 10.4049/jimmunol.1700851 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Amu S, Lantto Graham R, Bekele Y, Nasi A, Bengtsson C, Rethi B, Sorial S, Meini G, Zazzi M, Hejdeman B, et al. Dysfunctional phenotypes of CD4+ and CD8+ T cells are comparable in patients initiating ART during early or chronic HIV-1 infection. Medicine (Baltim). 2016;95:e3738. doi: 10.1097/MD.0000000000003738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Korencak M, Byrne M, Richter E, Schultz BT, Juszczak P, Ake JA, Ganesan A, Okulicz JF, Robb ML, de Los Reyes B, et al. Effect of HIV infection and antiretroviral therapy on immune cellular functions. JCI Insight. 2019;4:e126675. doi: 10.1172/jci.insight.126675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Alcaide ML, Parmigiani A, Pallikkuth S, Roach M, Freguja R, Della Negra M, Bolivar H, Fischl MA, Pahwa S. Immune activation in HIV-infected aging women on antiretrovirals--implications for age-associated comorbidities: a cross-sectional pilot study. PLoS One. 2013;8:e63804. doi: 10.1371/journal.pone.0063804 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hearps AC, Maisa A, Cheng WJ, Angelovich TA, Lichtfuss GF, Palmer CS, Landay AL, Jaworowski A, Crowe SM. HIV infection induces age-related changes to monocytes and innate immune activation in young men that persist despite combination antiretroviral therapy. AIDS. 2012;26:843–853. doi: 10.1097/QAD.0b013e328351f756 [DOI] [PubMed] [Google Scholar]
- 34.Burdo TH, Lentz MR, Autissier P, Krishnan A, Halpern E, Letendre S, Rosenberg ES, Ellis RJ, Williams KC. Soluble CD163 made by monocyte/macrophages is a novel marker of HIV activity in early and chronic infection prior to and after anti-retroviral therapy. J Infect Dis. 2011;204:154–163. doi: 10.1093/infdis/jir214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, Kazzaz Z, Bornstein E, Lambotte O, Altmann D, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12:1365–1371. doi: 10.1038/nm1511 [DOI] [PubMed] [Google Scholar]
- 36.Fitch KV, Srinivasa S, Abbara S, Burdo TH, Williams KC, Eneh P, Lo J, Grinspoon SK. Noncalcified coronary atherosclerotic plaque and immune activation in HIV-infected women. J Infect Dis. 2013;208:1737–1746. doi: 10.1093/infdis/jit508 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kelesidis T, Kendall MA, Yang OO, Hodis HN, Currier JS. Biomarkers of microbial translocation and macrophage activation: association with progression of subclinical atherosclerosis in HIV-1 infection. J Infect Dis. 2012;206:1558–1567. doi: 10.1093/infdis/jis545 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.McKibben RA, Margolick JB, Grinspoon S, Li X, Palella FJ, Kingsley LA, Witt MD, George RT, Jacobson LP, Budoff M, et al. Elevated levels of monocyte activation markers are associated with subclinical atherosclerosis in men with and those without HIV infection. J Infect Dis. 2015;211:1219–1228. doi: 10.1093/infdis/jiu594 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tawakol A, Ishai A, Li D, Takx RAP, Hur S, Kaiser Y, Pampaloni M, Rupert A, Hsu D, Sereti I, et al. Association of arterial and lymph node inflammation with distinct inflammatory pathways in human immunodeficiency virus infection. JAMA Cardiol. 2017;2:163–171. doi: 10.1001/jamacardio.2016.4728 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Díaz-Pollán B, Estrada V, Fuentes-Ferrer M, Gómez-Garré D, San Román-Montero J. Lp-PLA2 levels in HIV-infected patients. J Int AIDS Soc. 2014;17(4 Suppl 3):19721. doi: 10.7448/IAS.17.4.19721 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Temu TM, Polyak SJ, Zifodya JS, Wanjalla CN, Koethe JR, Masyuko S, Nyabiage J, Kinuthia J, Gervassi AL, Oyugi J, et al. Endothelial dysfunction is related to monocyte activation in antiretroviral-treated people with HIV and HIV-negative adults in Kenya. Open Forum Infect Dis. 2020;7:ofaa425. doi: 10.1093/ofid/ofaa425 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Taylor BS, So-Armah K, Tate JP, Marconi VC, Koethe JR, Bedimo RJ, Butt AA, Gibert CL, Goetz MB, Rodriguez-Barradas MC, et al. HIV and obesity comorbidity increase interleukin 6 but not soluble CD14 or D-dimer. J Acquir Immune Defic Syndr. 2017;75:500–508. doi: 10.1097/QAI.0000000000001444 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hunt PW, Martin JN, Sinclair E, Bredt B, Hagos E, Lampiris H, Deeks SG. T cell activation is associated with lower CD4 + T cell gains in human immunodeficiency virus–infected patients with sustained viral suppression during antiretroviral therapy. J Infect Dis. 2003;187:1534–1543. doi: 10.1086/374786 [DOI] [PubMed] [Google Scholar]
- 44.Hunt PW, Brenchley J, Sinclair E, McCune JM, Roland M, Page-Shafer K, Hsue P, Emu B, Krone M, Lampiris H, et al. Relationship between T cell activation and CD4+ T cell count in HIV-seropositive individuals with undetectable plasma HIV RNA levels in the absence of therapy. J Infect Dis. 2008;197:126–133. doi: 10.1086/524143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Neuhaus J, Jacobs DR, Baker JV, Calmy A, Duprez D, La Rosa A, Kuller LH, Pett SL, Ristola M, Ross MJ, et al. Markers of inflammation, coagulation, and renal function are elevated in adults with HIV infection. J Infect Dis. 2010;201:1788–1795. doi: 10.1086/652749 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sereti I, Rodger AJ, French MA. Biomarkers in immune reconstitution inflammatory syndrome: signals from pathogenesis. Curr Opin HIV AIDS. 2010;5:504–510. doi: 10.1097/COH.0b013e32833ed774 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kaplan RC, Sinclair E, Landay AL, Lurain N, Sharrett AR, Gange SJ, Xue X, Hunt P, Karim R, Kern DM, et al. T cell activation and senescence predict subclinical carotid artery disease in HIV-infected women. J Infect Dis. 2011;203:452–463. doi: 10.1093/infdis/jiq071 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Mudau M, Genis A, Lochner A, Strijdom H. Endothelial dysfunction: the early predictor of atherosclerosis. Cardiovasc J Afr. 2012;23:222–231. doi: 10.5830/CVJA-2011-068 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Basatemur GL, Jørgensen HF, Clarke MCH, Bennett MR, Mallat Z. Vascular smooth muscle cells in atherosclerosis. Nat Rev Cardiol. 2019;16:727–744. doi: 10.1038/s41569-019-0227-9 [DOI] [PubMed] [Google Scholar]
- 50.Libby P, Lichtman AH, Hansson GK. Immune effector mechanisms implicated in atherosclerosis: from mice to humans. Immunity. 2013;38:1092–1104. doi: 10.1016/j.immuni.2013.06.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Paladugu R, Fu W, Conklin BS, Lin PH, Lumsden AB, Yao Q, Chen C. HIV Tat protein causes endothelial dysfunction in porcine coronary arteries. J Vasc Surg. 2003;38:549–55; discussion 555. doi: 10.1016/s0741-5214(03)00770-5 [DOI] [PubMed] [Google Scholar]
- 52.Schecter AD, Berman AB, Yi L, Mosoian A, McManus CM, Berman JW, Klotman ME, Taubman MB. HIV envelope gp120 activates human arterial smooth muscle cells. Proc Natl Acad Sci USA. 2001;98:10142–10147. doi: 10.1073/pnas.181328798 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kanmogne GD, Kennedy RC, Grammas P. Analysis of human lung endothelial cells for susceptibility to HIV type 1 infection, coreceptor expression, and cytotoxicity of gp120 protein. AIDS Res Hum Retroviruses. 2001;17:45–53. doi: 10.1089/088922201750056771 [DOI] [PubMed] [Google Scholar]
- 54.Galea P, Vermot-Desroches C, Le Contel C, Wijdenes J, Chermann JC. Circulating cell adhesion molecules in HIV1-infected patients as indicator markers for AIDS progression. Res Immunol. 1997;148:109–117. doi: 10.1016/s0923-2494(97)82482-0 [DOI] [PubMed] [Google Scholar]
- 55.Hwang SJ, Ballantyne CM, Sharrett AR, Smith LC, Davis CE, Gotto AM, Boerwinkle E. Circulating adhesion molecules VCAM-1, ICAM-1, and E-selectin in carotid atherosclerosis and incident coronary heart disease cases: the Atherosclerosis Risk In Communities (ARIC) study. Circulation. 1997;96:4219–4225. doi: 10.1161/01.cir.96.12.4219 [DOI] [PubMed] [Google Scholar]
- 56.Semaan HB, Gurbel PA, Anderson JL, Muhlestein JB, Carlquist JF, Horne BD, Serebruany VL. Soluble VCAM-1 and E-selectin, but not ICAM-1 discriminate endothelial injury in patients with documented coronary artery disease. Cardiology. 2000;93:7–10. doi: 10.1159/000006995 [DOI] [PubMed] [Google Scholar]
- 57.Ross AC, Rizk N, O’Riordan MA, Dogra V, El-Bejjani D, Storer N, Harrill D, Tungsiripat M, Adell J, McComsey GA. Relationship between inflammatory markers, endothelial activation markers, and carotid intima-media thickness in HIV-infected patients receiving antiretroviral therapy. Clin Infect Dis. 2009;49:1119–1127. doi: 10.1086/605578 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Francisci D, Giannini S, Baldelli F, Leone M, Belfiori B, Guglielmini G, Malincarne L, Gresele P. HIV type 1 infection, and not short-term HAART, induces endothelial dysfunction. AIDS. 2009;23:589–596. doi: 10.1097/QAD.0b013e328325a87c [DOI] [PubMed] [Google Scholar]
- 59.Panigrahi S, Freeman ML, Funderburg NT, Mudd JC, Younes SA, Sieg SF, Zidar DA, Paiardini M, Villinger F, Calabrese LH, et al. SIV/SHIV infection triggers vascular inflammation, diminished expression of Krüppel-like factor 2 and endothelial dysfunction. J Infect Dis. 2016;213:1419–1427. doi: 10.1093/infdis/jiv749 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Huck DM, Weber B, Parks S, Divakaran S, Brown JM, Bibbo CF, Barrett L, Hainer J, Bay C, Martell L, et al. Coronary microcirculatory dysfunction in people with HIV and its association with antiretroviral therapy. J Am Heart Assoc. 2023;12:e029541. doi: 10.1161/JAHA.123.029541 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Rethy L, Feinstein MJ, Sinha A, Achenbach C, Shah SJ. Coronary microvascular dysfunction in HIV: a review. J Am Heart Assoc. 2020;9:e014018. doi: 10.1161/JAHA.119.014018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Thomas TS, Walpert AR, Srinivasa S. Large lessons learned from small vessels: coronary microvascular dysfunction in HIV. Curr Opin Infect Dis. 2024;37:26–34. doi: 10.1097/QCO.0000000000000987 [DOI] [PubMed] [Google Scholar]
- 63.Price LC, Wort SJ, Perros F, Dorfmüller P, Huertas A, Montani D, Cohen-Kaminsky S, Humbert M. Inflammation in pulmonary arterial hypertension. Chest. 2012;141:210–221. doi: 10.1378/chest.11-0793 [DOI] [PubMed] [Google Scholar]
- 64.Feingold KR. Introduction to lipids and lipoproteins. In: Feingold KR, Anawalt B, Blackman MR, et al. , eds. Endotext. MDText.com, Inc.; 2000. Accessed February 19, 2024. http://www.ncbi.nlm.nih.gov/books/NBK305896/ [PubMed] [Google Scholar]
- 65.Rose H, Hoy J, Woolley I, Tchoua U, Bukrinsky M, Dart A, Sviridov D. HIV infection and high density lipoprotein metabolism. Atherosclerosis. 2008;199:79–86. doi: 10.1016/j.atherosclerosis.2007.10.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Grunfeld C, Pang M, Doerrler W, Shigenaga JK, Jensen P, Feingold KR. Lipids, lipoproteins, triglyceride clearance, and cytokines in human immunodeficiency virus infection and the acquired immunodeficiency syndrome. J Clin Endocrinol Metab. 1992;74:1045–1052. doi: 10.1210/jcem.74.5.1373735 [DOI] [PubMed] [Google Scholar]
- 67.Spósito AC, Caramelli B, Sartori AM, F Ramires JA. The lipoprotein profile in HIV infected patients. Braz J Infect Dis. 1997;1:275–283. [PubMed] [Google Scholar]
- 68.Hui DY. Effects of HIV protease inhibitor therapy on lipid metabolism. Prog Lipid Res. 2003;42:81–92. doi: 10.1016/s0163-7827(02)00046-2 [DOI] [PubMed] [Google Scholar]
- 69.Likozar AR, Zavrtanik M, Šebeštjen M. Lipoprotein(a) in atherosclerosis: from pathophysiology to clinical relevance and treatment options. Ann Med. 2020;52:162. doi: 10.1080/07853890.2020.1775287 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Ugovšek S, Šebeštjen M. Lipoprotein(a)—the crossroads of atherosclerosis, atherothrombosis and inflammation. Biomolecules. 2021;12:26. doi: 10.3390/biom12010026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Leucker TM, Weiss RG, Schär M, Bonanno G, Mathews L, Jones SR, Brown TT, Moore R, Afework Y, Gerstenblith G, et al. Coronary endothelial dysfunction is associated with elevated serum PCSK9 levels in people with HIV independent of low-density lipoprotein cholesterol. J Am Heart Assoc. 2018;7:e009996. doi: 10.1161/JAHA.118.009996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Hsue P. Effect of PCSK9 Inhibition on Cardiovascular Risk in Treated HIV Infection (EPIC-HIV Study). clinicaltrials.gov; 2023. Accessed December 31, 2023. https://clinicaltrials.gov/study/NCT03207945 [Google Scholar]
- 73.Melaku L, Dabi A. The cellular biology of atherosclerosis with atherosclerotic lesion classification and biomarkers. Bull Natl Res Cent. 2021;45:225. doi: 10.1186/s42269-021-00685-w [Google Scholar]
- 74.Grunfeld C, Kotler DP, Shigenaga JK, Doerrler W, Tierney A, Wang J, Pierson RN, Feingold KR. Circulating interferon-alpha levels and hypertriglyceridemia in the acquired immunodeficiency syndrome. Am J Med. 1991;90:154–162. doi: 10.1016/0002-9343(91)80154-E [PubMed] [Google Scholar]
- 75.Sarkar S, Haberlen S, Whelton S, E Schneider E, Kingsley L, Palella F, Witt MD, Kelesidis T, Rodriguez A, Post WS, et al. Greater IL-6, D-dimer, and ICAM-1 levels are associated with lower small HDL particle concentration in the multicenter AIDS cohort study. Open Forum Infect Dis. 2019;6:ofz474. doi: 10.1093/ofid/ofz474 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Jiménez-Osorio AS, Jaen-Vega S, Fernández-Martínez E, Ortíz-Rodríguez MA, Martínez-Salazar MF, Jiménez-Sánchez RC, Flores-Chávez OR, Ramírez-Moreno E, Arias-Rico J, Arteaga-García F, et al. Antiretroviral therapy-induced dysregulation of gene expression and lipid metabolism in HIV+ patients: beneficial role of antioxidant phytochemicals. Int J Mol Sci. 2022;23:5592. doi: 10.3390/ijms23105592 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Combadière C, Potteaux S, Rodero M, Simon T, Pezard A, Esposito B, Merval R, Proudfoot A, Tedgui A, Mallat Z. Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6C(hi) and Ly6C(lo) monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation. 2008;117:1649–1657. doi: 10.1161/CIRCULATIONAHA.107.745091 [DOI] [PubMed] [Google Scholar]
- 78.Tacke F, Alvarez D, Kaplan TJ, Jakubzick C, Spanbroek R, Llodra J, Garin A, Liu J, Mack M, van Rooijen N, et al. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest. 2007;117:185–194. doi: 10.1172/JCI28549 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Tabas I, Lichtman AH. Monocyte-macrophages and T cells in atherosclerosis. Immunity. 2017;47:621–634. doi: 10.1016/j.immuni.2017.09.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Feeney ER, McAuley N, O’Halloran JA, Rock C, Low J, Satchell CS, Lambert JS, Sheehan GJ, Mallon PWG. The expression of cholesterol metabolism genes in monocytes from HIV-infected subjects suggests intracellular cholesterol accumulation. J Infect Dis. 2013;207:628–637. doi: 10.1093/infdis/jis723 [DOI] [PubMed] [Google Scholar]
- 81.Maisa A, Hearps AC, Angelovich TA, Pereira CF, Zhou J, Shi MDY, Palmer CS, Muller WA, Crowe SM, Jaworowski A. Monocytes from HIV-infected individuals show impaired cholesterol efflux and increased foam cell formation after transendothelial migration. AIDS. 2015;29:1445–1457. doi: 10.1097/QAD.0000000000000739 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Mujawar Z, Rose H, Morrow MP, Pushkarsky T, Dubrovsky L, Mukhamedova N, Fu Y, Dart A, Orenstein JM, Bobryshev YV, et al. Human immunodeficiency virus impairs reverse cholesterol transport from macrophages. PLoS Biol. 2006;4:e365. doi: 10.1371/journal.pbio.0040365 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Asztalos BF, Mujawar Z, Morrow MP, Grant A, Pushkarsky T, Wanke C, Shannon R, Geyer M, Kirchhoff F, Sviridov D, et al. Circulating Nef induces dyslipidemia in simian immunodeficiency virus-infected macaques by suppressing cholesterol efflux. J Infect Dis. 2010;202:614–623. doi: 10.1086/654817 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Okpaise D, Sluis-Cremer N, Rappocciolo G, Rinaldo CR. Cholesterol metabolism in antigen-presenting cells and HIV-1 trans-infection of CD4+ T cells. Viruses. 2023;15:2347. doi: 10.3390/v15122347 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Rappocciolo G, Jais M, Piazza P, Reinhart TA, Berendam SJ, Garcia-Exposito L, Gupta P, Rinaldo CR. Alterations in cholesterol metabolism restrict HIV-1 trans infection in nonprogressors. mBio. 2014;5:e01031–e01013. doi: 10.1128/mBio.01031-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kruth HS. Sequestration of aggregated low-density lipoproteins by macrophages. Curr Opin Lipidol. 2002;13:483–488. doi: 10.1097/00041433-200210000-00003 [DOI] [PubMed] [Google Scholar]
- 87.Clinton SK, Underwood R, Hayes L, Sherman ML, Kufe DW, Libby P. Macrophage colony-stimulating factor gene expression in vascular cells and in experimental and human atherosclerosis. Am J Pathol. 1992;140:301–316. [PMC free article] [PubMed] [Google Scholar]
- 88.Post WS, Budoff M, Kingsley L, Palella FJ, Witt MD, Li X, George RT, Brown TT, Jacobson LP. Associations between HIV infection and subclinical coronary atherosclerosis. Ann Intern Med. 2014;160:458–467. doi: 10.7326/M13-1754 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.D’Ascenzo F, Cerrato E, Calcagno A, Grossomarra W, Ballocca F, Omedè P, Montefusco A, Veglia S, Barbero U, Gili S, et al. High prevalence at computed coronary tomography of non-calcified plaques in asymptomatic HIV patients treated with HAART: a meta-analysis. Atherosclerosis. 2015;240:197–204. doi: 10.1016/j.atherosclerosis.2015.03.019 [DOI] [PubMed] [Google Scholar]
- 90.Mauriello A, Servadei F, Zoccai GB, Giacobbi E, Anemona L, Bonanno E, Casella S. Coronary calcification identifies the vulnerable patient rather than the vulnerable Plaque. Atherosclerosis. 2013;229:124–129. doi: 10.1016/j.atherosclerosis.2013.03.010 [DOI] [PubMed] [Google Scholar]
- 91.Motoyama S, Kondo T, Sarai M, Sugiura A, Harigaya H, Sato T, Inoue K, Okumura M, Ishii J, Anno H, et al. Multislice computed tomographic characteristics of coronary lesions in acute coronary syndromes. J Am Coll Cardiol. 2007;50:319–326. doi: 10.1016/j.jacc.2007.03.044 [DOI] [PubMed] [Google Scholar]
- 92.Virmani R, Burke AP, Farb A, Kolodgie FD. Pathology of the vulnerable plaque. J Am Coll Cardiol. 2006;47(8 Suppl):C13–C18. doi: 10.1016/j.jacc.2005.10.065 [DOI] [PubMed] [Google Scholar]
- 93.Miller PE, Haberlen SA, Metkus T, Rezaeian P, Palella F, Kingsley LA, Witt MD, George RT, Jacobson LP, Brown TT, et al. HIV and coronary arterial remodeling from the Multicenter AIDS Cohort Study (MACS). Atherosclerosis. 2015;241:716–722. doi: 10.1016/j.atherosclerosis.2015.06.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ellery PJ, Tippett E, Chiu YL, Paukovics G, Cameron PU, Solomon A, Lewin SR, Gorry PR, Jaworowski A, Greene WC, et al. The CD16+ monocyte subset is more permissive to infection and preferentially harbors HIV-1 in vivo. J Immunol. 2007;178:6581–6589. doi: 10.4049/jimmunol.178.10.6581 [DOI] [PubMed] [Google Scholar]
- 95.Han J, Wang B, Han N, Zhao Y, Song C, Feng X, Mao Y, Zhang F, Zhao H, Zeng H. CD14(high)CD16(+) rather than CD14(low)CD16(+) monocytes correlate with disease progression in chronic HIV-infected patients. J Acquir Immune Defic Syndr. 2009;52:553–559. doi: 10.1097/qai.0b013e3181c1d4fe [DOI] [PubMed] [Google Scholar]
- 96.Kawanaka N, Yamamura M, Aita T, Morita Y, Okamoto A, Kawashima M, Iwahashi M, Ueno A, Ohmoto Y, Makino H. CD14+,CD16+ blood monocytes and joint inflammation in rheumatoid arthritis. Arthritis Rheum. 2002;46:2578–2586. doi: 10.1002/art.10545 [DOI] [PubMed] [Google Scholar]
- 97.Chow DC, Kagihara JM, Zhang G, Souza SA, Hodis HN, Li Y, Mitchell BI, Nakamoto BK, Kallianpur KJ, Keating SM, et al. Non-classical monocytes predict progression of carotid artery bifurcation intima-media thickness in HIV-infected individuals on stable antiretroviral therapy. HIV Clin Trials. 2016;17:114–122. doi: 10.1080/15284336.2016.1162386 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Funderburg NT, Zidar DA, Shive C, Lioi A, Mudd J, Musselwhite LW, Simon DI, Costa MA, Rodriguez B, Sieg SF, et al. Shared monocyte subset phenotypes in HIV-1 infection and in uninfected subjects with acute coronary syndrome. Blood. 2012;120:4599–4608. doi: 10.1182/blood-2012-05-433946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Pereyra F, Lo J, Triant VA, Wei J, Buzon MJ, Fitch KV, Hwang J, Campbell JH, Burdo TH, Williams KC, et al. Increased coronary atherosclerosis and immune activation in HIV-1 elite controllers. AIDS. 2012;26:2409–2412. doi: 10.1097/QAD.0b013e32835a9950 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hsu DC, Ma YF, Hur S, Li D, Rupert A, Scherzer R, Kalapus SC, Deeks S, Sereti I, Hsue PY. Plasma IL-6 levels are independently associated with atherosclerosis and mortality in HIV-infected individuals on suppressive antiretroviral therapy. AIDS. 2016;30:2065–2074. doi: 10.1097/QAD.0000000000001149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Barbour JD, Jalbert EC, Chow DC, Gangcuangco LMA, Norris PJ, Keating SM, Heitman J, Nagamine L, Seto T, Ndhlovu LC, et al. Reduced CD14 expression on classical monocytes and vascular endothelial adhesion markers independently associate with carotid artery intima media thickness in chronically HIV-1 infected adults on virologically suppressive anti-retroviral therapy. Atherosclerosis. 2014;232:52–58. doi: 10.1016/j.atherosclerosis.2013.10.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Shaked I, Hanna DB, Gleißner C, Marsh B, Plants J, Tracy D, Anastos K, Cohen M, Golub ET, Karim R, et al. Macrophage inflammatory markers are associated with subclinical carotid artery disease in women with human immunodeficiency virus or hepatitis C virus infection. Arterioscler Thromb Vasc Biol. 2014;34:1085–1092. doi: 10.1161/ATVBAHA.113.303153 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Longenecker CT, Jiang Y, Yun CH, Debanne S, Funderburg NT, Lederman MM, Storer N, Labbato DE, Bezerra HG, McComsey GA. Perivascular fat, inflammation, and cardiovascular risk in HIV-infected patients on antiretroviral therapy. Int J Cardiol. 2013;168:4039–4045. doi: 10.1016/j.ijcard.2013.06.059 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Longenecker CT, Funderburg NT, Jiang Y, Debanne S, Storer N, Labbato DE, Lederman MM, McComsey GA. Markers of inflammation and CD8 T-cell activation, but not monocyte activation, are associated with subclinical carotid artery disease in HIV-infected individuals. HIV Med. 2013;14:385–390. doi: 10.1111/hiv.12013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Rogacev KS, Cremers B, Zawada AM, Seiler S, Binder N, Ege P, Große-Dunker G, Heisel I, Hornof F, Jeken J, et al. CD14++CD16+ monocytes independently predict cardiovascular events: a cohort study of 951 patients referred for elective coronary angiography. J Am Coll Cardiol. 2012;60:1512–1520. doi: 10.1016/j.jacc.2012.07.019 [DOI] [PubMed] [Google Scholar]
- 106.Subramanian S, Tawakol A, Burdo TH, Abbara S, Wei J, Vijayakumar J, Corsini E, Abdelbaky A, Zanni MV, Hoffmann U, et al. Arterial inflammation in patients with HIV. JAMA. 2012;308:379–386. doi: 10.1001/jama.2012.6698 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Barton GM. Viral recognition by toll-like receptors. Semin Immunol. 2007;19:33–40. doi: 10.1016/j.smim.2007.01.003 [DOI] [PubMed] [Google Scholar]
- 108.Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science. 2004;303:1529–1531. doi: 10.1126/science.1093616 [DOI] [PubMed] [Google Scholar]
- 109.Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, Lipford G, Wagner H, Bauer S. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science. 2004;303:1526–1529. doi: 10.1126/science.1093620 [DOI] [PubMed] [Google Scholar]
- 110.Siegal FP, Kadowaki N, Shodell M, Fitzgerald-Bocarsly PA, Shah K, Ho S, Antonenko S, Liu YJ. The nature of the principal type 1 interferon-producing cells in human blood. Science. 1999;284:1835–1837. doi: 10.1126/science.284.5421.1835 [DOI] [PubMed] [Google Scholar]
- 111.Buitendijk M, Eszterhas SK, Howell AL. Toll-like receptor agonists are potent inhibitors of human immunodeficiency virus-type 1 replication in peripheral blood mononuclear cells. AIDS Res Hum Retroviruses. 2014;30:457–467. doi: 10.1089/AID.2013.0199 [DOI] [PubMed] [Google Scholar]
- 112.Lehmann C, Harper JM, Taubert D, Hartmann P, Fätkenheuer G, Jung N, van Lunzen J, Stellbrink HJ, Gallo RC, Romerio F. Increased interferon alpha expression in circulating plasmacytoid dendritic cells of HIV-1-infected patients. J Acquir Immune Defic Syndr. 2008;48:522–530. doi: 10.1097/QAI.0b013e31817f97cf [DOI] [PubMed] [Google Scholar]
- 113.Hernandez JC, Latz E, Urcuqui-Inchima S. HIV-1 induces the first signal to activate the NLRP3 inflammasome in monocyte-derived macrophages. Intervirology. 2014;57:36–42. doi: 10.1159/000353902 [DOI] [PubMed] [Google Scholar]
- 114.Kearns AC, Liu F, Dai S, Robinson JA, Kiernan E, Tesfaye Cheru L, Peng X, Gordon J, Morgello S, Abuova A, et al. Caspase-1 activation is related with HIV-associated atherosclerosis in an HIV transgenic mouse model and HIV patient cohort. Arterioscler Thromb Vasc Biol. 2019;39:1762–1775. doi: 10.1161/ATVBAHA.119.312603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Gardner MB, Luciw PA. Macaque models of human infectious disease. ILAR J. 2008;49:220–255. doi: 10.1093/ilar.49.2.220 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Petkov DI, Liu DX, Allers C, Didier PJ, Didier ES, Kuroda MJ. Characterization of heart macrophages in rhesus macaques as a model to study cardiovascular disease in humans. J Leukoc Biol. 2019;106:1241–1255. doi: 10.1002/JLB.1A0119-017R [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Lackner AA, Veazey RS. Current concepts in AIDS pathogenesis: insights from the SIV/macaque model. Annu Rev Med. 2007;58:461–476. doi: 10.1146/annurev.med.58.082405.094316 [DOI] [PubMed] [Google Scholar]
- 118.White KS, Walker JA, Wang J, Autissier P, Miller AD, Abuelezan NN, Burrack R, Li Q, Kim W-K, Williams KC. Simian immunodeficiency virus-infected rhesus macaques with AIDS co-develop cardiovascular pathology and encephalitis. Front Immunol. 2023;14:1240946. doi: 10.3389/fimmu.2023.1240946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Bick AG, Pirruccello JP, Griffin GK, Gupta N, Gabriel S, Saleheen D, Libby P, Kathiresan S, Natarajan P. Genetic IL-6 signaling deficiency attenuates cardiovascular risk in clonal hematopoiesis. Circulation. 2020;141:124–131. doi: 10.1161/CIRCULATIONAHA.119.044362 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, Lindsley RC, Mermel CH, Burtt N, Chavez A, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371:2488–2498. doi: 10.1056/NEJMoa1408617 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Jaiswal S, Libby P. Clonal haematopoiesis: connecting ageing and inflammation in cardiovascular disease. Nat Rev Cardiol. 2020;17:137–144. doi: 10.1038/s41569-019-0247-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Marnell CS, Bick A, Natarajan P. Clonal hematopoiesis of indeterminate potential (CHIP): linking somatic mutations, hematopoiesis, chronic inflammation and cardiovascular disease. J Mol Cell Cardiol. 2021;161:98–105. doi: 10.1016/j.yjmcc.2021.07.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Steensma DP, Bejar R, Jaiswal S, Lindsley RC, Sekeres MA, Hasserjian RP, Ebert BL. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood. 2015;126:9–16. doi: 10.1182/blood-2015-03-631747 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Fuster JJ, MacLauchlan S, Zuriaga MA, Polackal MN, Ostriker AC, Chakraborty R, Wu CL, Sano S, Muralidharan S, Rius C, et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science. 2017;355:842–847. doi: 10.1126/science.aag1381 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Bick AG, Popadin K, Thorball CW, Uddin MM, Zanni MV, Yu B, Cavassini M, Rauch A, Tarr P, Schmid P, et al. ; Swiss HIV Cohort Study. Increased prevalence of clonal hematopoiesis of indeterminate potential amongst people living with HIV. Sci Rep. 2022;12:577. doi: 10.1038/s41598-021-04308-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Dharan NJ, Yeh P, Bloch M, Yeung MM, Baker D, Guinto J, Roth N, Ftouni S, Ognenovska K, Smith D, et al. ; ARCHIVE Study Group. HIV is associated with an increased risk of age-related clonal hematopoiesis among older adults. Nat Med. 2021;27:1006–1011. doi: 10.1038/s41591-021-01357-y [DOI] [PubMed] [Google Scholar]
- 127.Bhattacharya R, Uddin MM, Patel AP, Niroula A, Finneran P, Bernardo R, Fitch KV, Lu MT, Bloomfield GS, Malvestutto C, et al. Risk factors for clonal hematopoiesis of indeterminate potential in people with HIV: a report from the REPRIEVE trial. Blood Adv. 2024;8:959–967. doi: 10.1182/bloodadvances.2023011324 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Liu K. Dendritic cells. Encyclopedia of Cell Biology. 2016;741–749. doi: 10.1016/B978-0-12-394447-4.30111-0 [Google Scholar]
- 129.Wu L, KewalRamani VN. Dendritic-cell interactions with HIV: infection and viral dissemination. Nat Rev Immunol. 2006;6:859–868. doi: 10.1038/nri1960 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Naik SH, Sathe P, Park HY, Metcalf D, Proietto AI, Dakic A, Carotta S, O'Keeffe M, Bahlo M, Papenfuss A, et al. Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo. Nat Immunol. 2007;8:1217–1226. doi: 10.1038/ni1522 [DOI] [PubMed] [Google Scholar]
- 131.Subramanian M, Thorp E, Hansson GK, Tabas I. Treg-mediated suppression of atherosclerosis requires MYD88 signaling in DCs. J Clin Invest. 2013;123:179–188. doi: 10.1172/JCI64617 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zernecke A. Dendritic cells in atherosclerosis: evidence in mice and humans. Arterioscler Thromb Vasc Biol. 2015;35:763–770. doi: 10.1161/ATVBAHA.114.303566 [DOI] [PubMed] [Google Scholar]
- 133.Pope M, Gezelter S, Gallo N, Hoffman L, Steinman RM. Low levels of HIV-1 infection in cutaneous dendritic cells promote extensive viral replication upon binding to memory CD4+ T cells. J Exp Med. 1995;182:2045–2056. doi: 10.1084/jem.182.6.2045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Bender AT, Tzvetkov E, Pereira A, Wu Y, Kasar S, Przetak MM, Vlach J, Niewold TB, Jensen MA, Okitsu SL. TLR7 and TLR8 differentially activate the IRF and NF-κB pathways in specific cell types to promote inflammation. Immunohorizons. 2020;4:93–107. doi: 10.4049/immunohorizons.2000002 [DOI] [PubMed] [Google Scholar]
- 135.Zelensky AN, Gready JE. The C-type lectin-like domain superfamily. FEBS J. 2005;272:6179–6217. doi: 10.1111/j.1742-4658.2005.05031.x [DOI] [PubMed] [Google Scholar]
- 136.Bates EEM, Fournier N, Garcia E, Valladeau J, Durand I, Pin JJ, Zurawski SM, Patel S, Abrams JS, Lebecque S, et al. APCs express DCIR, a novel C-type lectin surface receptor containing an immunoreceptor tyrosine-based inhibitory motif. J Immunol. 1999;163:1973–1983. doi: 10.4049/jimmunol.163.4.1973 [PubMed] [Google Scholar]
- 137.Staeheli P, Haller O. Human MX2/MxB: a potent interferon-induced postentry inhibitor of herpesviruses and HIV-1. J Virol. 2018;92:e00709–e00718. doi: 10.1128/JVI.00709-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Yan N, Chen ZJ. Intrinsic antiviral immunity. Nat Immunol. 2012;13:214–222. doi: 10.1038/ni.2229 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Sandler NG, Bosinger SE, Estes JD, Zhu RTR, Tharp GK, Boritz E, Levin D, Wijeyesinghe S, Makamdop KN, del Prete GQ, et al. Type i interferon responses in rhesus macaques prevent SIV infection and slow disease progression. Nature. 2014;511:601–605. doi: 10.1038/nature13554 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Moris A, Nobile C, Buseyne F, Porrot F, Abastado JP, Schwartz O. DC-SIGN promotes exogenous MHC-I-restricted HIV-1 antigen presentation. Blood. 2004;103:2648–2654. doi: 10.1182/blood-2003-07-2532 [DOI] [PubMed] [Google Scholar]
- 141.Chatterjee B, Smed-Sörensen A, Cohn L, Chalouni C, Vandlen R, Lee BC, Widger J, Keler T, Delamarre L, Mellman I. Internalization and endosomal degradation of receptor-bound antigens regulate the efficiency of cross presentation by human dendritic cells. Blood. 2012;120:2011–2020. doi: 10.1182/blood-2012-01-402370 [DOI] [PubMed] [Google Scholar]
- 142.Yang K, Liu X, Liu Y, Wang X, Cao L, Zhang X, Xu C, Shen W, Zhou T. DC-SIGN and toll-like receptor 4 mediate oxidized low-density lipoprotein-induced inflammatory responses in macrophages. Sci Rep. 2017;7:3296. doi: 10.1038/s41598-017-03740-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Xue Y, Li Q, Park CG, Klena JD, Anisimov AP, Sun Z, Wei X, Chen T. Proteus mirabilis targets atherosclerosis plaques in human coronary arteries via DC-SIGN (CD209). Front Immunol. 2020;11:579010. doi: 10.3389/fimmu.2020.579010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Wang Z, Peters BA, Usyk M, Xing J, Hanna DB, Wang T, Post WS, Landay AL, Hodis HN, Weber K, et al. Gut microbiota, plasma metabolomic profiles, and carotid artery atherosclerosis in HIV infection. Arterioscler Thromb Vasc Biol. 2022;42:1081–1093. doi: 10.1161/ATVBAHA.121.317276 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Merle NS, Noe R, Halbwachs-Mecarelli L, Fremeaux-Bacchi V, Roumenina LT. Complement system part II: role in immunity. Front Immunol. 2015;6:257. doi: 10.3389/fimmu.2015.00257 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Ji X, Gewurz H, Spear GT. Mannose binding lectin (MBL) and HIV. Mol Immunol. 2005;42:145–152. doi: 10.1016/j.molimm.2004.06.015 [DOI] [PubMed] [Google Scholar]
- 147.Mellman I, Steinman RM. Dendritic cells: specialized and regulated antigen processing machines. Cell. 2001;106:255–258. doi: 10.1016/s0092-8674(01)00449-4 [DOI] [PubMed] [Google Scholar]
- 148.Geijtenbeek TBH, Gringhuis SI. C-type lectin receptors in the control of T helper cell differentiation. Nat Rev Immunol. 2016;16:433–448. doi: 10.1038/nri.2016.55 [DOI] [PubMed] [Google Scholar]
- 149.Lv T, Cao W, Li T. HIV-related immune activation and inflammation: current understanding and strategies. J Immunol Res. 2021;2021:7316456. doi: 10.1155/2021/7316456 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Duprez DA, Neuhaus J, Kuller LH, Tracy R, Belloso W, De Wit S, Drummond F, Lane HC, Ledergerber B, Lundgren J, et al. ; INSIGHT SMART Study Group. Inflammation, coagulation and cardiovascular disease in HIV-infected individuals. PLoS One. 2012;7:e44454. doi: 10.1371/journal.pone.0044454 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Wolf K, Tsakiris DA, Weber R, Erb P, Battegay M; Swiss HIV Cohort Study. Antiretroviral therapy reduces markers of endothelial and coagulation activation in patients infected with human immunodeficiency virus type 1. J Infect Dis. 2002;185:456–462. doi: 10.1086/338572 [DOI] [PubMed] [Google Scholar]
- 152.Nordell AD, McKenna M, Borges AH, Duprez D, Neuhaus J, Neaton JD; INSIGHT SMART, ESPRIT Study Groups. Severity of cardiovascular disease outcomes among patients with HIV is related to markers of inflammation and coagulation. J Am Heart Assoc. 2014;3:e000844. doi: 10.1161/JAHA.114.000844 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Lassila R, Peltonen S, Lepäntalo M, Saarinen O, Kauhanen P, Manninen V. Severity of peripheral atherosclerosis is associated with fibrinogen and degradation of cross-linked fibrin. Arterioscler Thromb. 1993;13:1738–1742. doi: 10.1161/01.atv.13.12.1738 [DOI] [PubMed] [Google Scholar]
- 154.Danesh J, Whincup P, Walker M, Lennon L, Thomson A, Appleby P, Rumley A, Lowe GD. Fibrin D-dimer and coronary heart disease: prospective study and meta-analysis. Circulation. 2001;103:2323–2327. doi: 10.1161/01.cir.103.19.2323 [DOI] [PubMed] [Google Scholar]
- 155.Abuaf N, Laperche S, Rajoely B, Carsique R, Deschamps A, Rouquette AM, Barthet C, Khaled Z, Marbot C, Saab N, et al. Autoantibodies to phospholipids and to the coagulation proteins in AIDS. Thromb Haemost. 1997;77:856–861. [PubMed] [Google Scholar]
- 156.Bissuel F, Berruyer M, Causse X, Dechavanne M, Trepo C. Acquired protein S deficiency: correlation with advanced disease in HIV-1-infected patients. J Acquir Immune Defic Syndr (1988). 1992;5:484–489. [PubMed] [Google Scholar]
- 157.Blann A, Constans J, Dignat-George F, Seigneur M. The platelet and endothelium in HIV infection. Br J Haematol. 1998;100:613–614. doi: 10.1046/j.1365-2141.1998.0636j.x [DOI] [PubMed] [Google Scholar]
- 158.Blann AD, Seigneur M, Constans J, Pellegrin JL, Conri C. Soluble P-selectin, thrombocytopenia and von Willebrand factor in HIV infected patients. Thromb Haemost. 1997;77:1221–1222. [PubMed] [Google Scholar]
- 159.Shen YMP, Frenkel EP. Thrombosis and a hypercoagulable state in HIV-infected patients. Clin Appl Thromb Hemost. 2004;10:277–280. doi: 10.1177/107602960401000311 [DOI] [PubMed] [Google Scholar]
- 160.Brummel-Ziedins K, Undas A, Orfeo T, Gissel M, Butenas S, Zmudka K, Mann KG. Thrombin generation in acute coronary syndrome and stable coronary artery disease: dependence on plasma factor composition. J Thromb Haemost. 2008;6:104–110. doi: 10.1111/j.1538-7836.2007.02799.x [DOI] [PubMed] [Google Scholar]
- 161.Baker JV, Brummel-Ziedins K, Neuhaus J, Duprez D, Cummins N, Dalmau D, DeHovitz J, Lehmann C, Sullivan A, Woolley I, et al. ; INSIGHT SMART Study Team. HIV replication alters the composition of extrinsic pathway coagulation factors and increases thrombin generation. J Am Heart Assoc. 2013;2:e000264. doi: 10.1161/JAHA.113.000264 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Funderburg NT, Mayne E, Sieg SF, Asaad R, Jiang W, Kalinowska M, Luciano AA, Stevens W, Rodriguez B, Brenchley JM, et al. Increased tissue factor expression on circulating monocytes in chronic HIV infection: relationship to in vivo coagulation and immune activation. Blood. 2010;115:161–167. doi: 10.1182/blood-2009-03-210179 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Vivier E, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley RM, McKenzie ANJ, Mebius RE, et al. Innate lymphoid cells: 10 years on. Cell. 2018;174:1054–1066. doi: 10.1016/j.cell.2018.07.017 [DOI] [PubMed] [Google Scholar]
- 164.Seillet C, Belz GT, Huntington ND. Development, homeostasis, and heterogeneity of NK cells and ILC1. Curr Top Microbiol Immunol. 2016;395:37–61. doi: 10.1007/82_2015_474 [DOI] [PubMed] [Google Scholar]
- 165.Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley RM, McKenzie ANJ, Mebius RE, et al. Innate lymphoid cells--a proposal for uniform nomenclature. Nat Rev Immunol. 2013;13:145–149. doi: 10.1038/nri3365 [DOI] [PubMed] [Google Scholar]
- 166.Eberl G, Colonna M, Di Santo JP, McKenzie ANJ. Innate lymphoid cells. Innate lymphoid cells: a new paradigm in immunology. Science. 2015;348:aaa6566. doi: 10.1126/science.aaa6566 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Taleb S, Tedgui A, Mallat Z. IL-17 and Th17 cells in atherosclerosis: subtle and contextual roles. Arterioscler Thromb Vasc Biol. 2015;35:258–264. doi: 10.1161/ATVBAHA.114.303567 [DOI] [PubMed] [Google Scholar]
- 168.Vu DM, Tai A, Tatro JB, Karas RH, Huber BT, Beasley D. γδT cells are prevalent in the proximal aorta and drive nascent atherosclerotic lesion progression and neutrophilia in hypercholesterolemic mice. PLoS One. 2014;9:e109416. doi: 10.1371/journal.pone.0109416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Kleindienst R, Xu Q, Willeit J, Waldenberger FR, Weimann S, Wick G. Immunology of atherosclerosis. Demonstration of heat shock protein 60 expression and T lymphocytes bearing alpha/beta or gamma/delta receptor in human atherosclerotic lesions. Am J Pathol. 1993;142:1927–1937. [PMC free article] [PubMed] [Google Scholar]
- 170.Taggenbrock RLRE, van Gisbergen KPJM. ILC1: development, maturation, and transcriptional regulation. Eur J Immunol. 2023;53:e2149435. doi: 10.1002/eji.202149435 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Montaldo E, Juelke K, Romagnani C. Group 3 innate lymphoid cells (ILC3s): origin, differentiation, and plasticity in humans and mice. Eur J Immunol. 2015;45:2171–2182. doi: 10.1002/eji.201545598 [DOI] [PubMed] [Google Scholar]
- 172.Newland SA, Mohanta S, Clément M, Taleb S, Walker JA, Nus M, Sage AP, Yin C, Hu D, Kitt LL, et al. Type-2 innate lymphoid cells control the development of atherosclerosis in mice. Nat Commun. 2017;8:15781. doi: 10.1038/ncomms15781 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Kløverpris HN, Kazer SW, Mjösberg J, Mabuka JM, Wellmann A, Ndhlovu Z, Yadon MC, Nhamoyebonde S, Muenchhoff M, Simoni Y, et al. Innate lymphoid cells are depleted irreversibly during acute HIV-1 infection in the absence of viral suppression. Immunity. 2016;44:391–405. doi: 10.1016/j.immuni.2016.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.van der Heijden WA, Van de Wijer L, Keramati F, Trypsteen W, Rutsaert S, Horst RT, Jaeger M, Koenen HJ, Stunnenberg HG, Joosten I, et al. Chronic HIV infection induces transcriptional and functional reprogramming of innate immune cells. JCI Insight. 2021;6:e145928. doi: 10.1172/jci.insight.145928 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Netea MG, Domínguez-Andrés J, Barreiro LB, Chavakis T, Divangahi M, Fuchs E, Joosten LAB, van der Meer JWM, Mhlanga MM, Mulder WJM, et al. Defining trained immunity and its role in health and disease. Nat Rev Immunol. 2020;20:375–388. doi: 10.1038/s41577-020-0285-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Leentjens J, Bekkering S, Joosten LAB, Netea MG, Burgner DP, Riksen NP. Trained innate immunity as a novel mechanism linking infection and the development of atherosclerosis. Circ Res. 2018;122:664–669. doi: 10.1161/CIRCRESAHA.117.312465 [DOI] [PubMed] [Google Scholar]
- 177.Ammirati E, Moroni F, Magnoni M, Camici PG. The role of T and B cells in human atherosclerosis and atherothrombosis. Clin Exp Immunol. 2015;179:173–187. doi: 10.1111/cei.12477 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Jonasson L, Holm J, Skalli O, Bondjers G, Hansson GK. Regional accumulations of T cells, macrophages, and smooth muscle cells in the human atherosclerotic plaque. Arteriosclerosis. 1986;6:131–138. doi: 10.1161/01.atv.6.2.131 [DOI] [PubMed] [Google Scholar]
- 179.Fatkhullina AR, Peshkova IO, Koltsova EK. The role of cytokines in the development of atherosclerosis. Biochemistry (Mosc). 2016;81:1358–1370. doi: 10.1134/S0006297916110134 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Lahoute C, Herbin O, Mallat Z, Tedgui A. Adaptive immunity in atherosclerosis: mechanisms and future therapeutic targets. Nat Rev Cardiol. 2011;8:348–358. doi: 10.1038/nrcardio.2011.62 [DOI] [PubMed] [Google Scholar]
- 181.Liuzzo G, Goronzy JJ, Yang H, Kopecky SL, Holmes DR, Frye RL, Weyand CM. Monoclonal T-cell proliferation and plaque instability in acute coronary syndromes. Circulation. 2000;101:2883–2888. doi: 10.1161/01.cir.101.25.2883 [DOI] [PubMed] [Google Scholar]
- 182.Paulsson G, Zhou X, Törnquist E, Hansson GK. Oligoclonal T cell expansions in atherosclerotic lesions of apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2000;20:10–17. doi: 10.1161/01.atv.20.1.10 [DOI] [PubMed] [Google Scholar]
- 183.Stemme S, Faber B, Holm J, Wiklund O, Witztum JL, Hansson GK. T lymphocytes from human atherosclerotic plaques recognize oxidized low density lipoprotein. Proc Natl Acad Sci USA. 1995;92:3893–3897. doi: 10.1073/pnas.92.9.3893 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Ley K. Role of the adaptive immune system in atherosclerosis. Biochem Soc Trans. 2020;48:2273–2281. doi: 10.1042/BST20200602 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Kol A, Lichtman AH, Finberg RW, Libby P, Kurt-Jones EA. Cutting edge: heat shock protein (HSP) 60 activates the innate immune response: CD14 is an essential receptor for HSP60 activation of mononuclear cells. J Immunol. 2000;164:13–17. doi: 10.4049/jimmunol.164.1.13 [DOI] [PubMed] [Google Scholar]
- 186.Vliegen I, Herngreen SB, Grauls GELM, Bruggeman CA, Stassen FRM. Mouse cytomegalovirus antigenic immune stimulation is sufficient to aggravate atherosclerosis in hypercholesterolemic mice. Atherosclerosis. 2005;181:39–44. doi: 10.1016/j.atherosclerosis.2004.12.035 [DOI] [PubMed] [Google Scholar]
- 187.Mach F, Schönbeck U, Sukhova GK, Atkinson E, Libby P. Reduction of atherosclerosis in mice by inhibition of CD40 signalling. Nature. 1998;394:200–203. doi: 10.1038/28204 [DOI] [PubMed] [Google Scholar]
- 188.Ludewig B, Freigang S, Jäggi M, Kurrer MO, Pei YC, Vlk L, Odermatt B, Zinkernagel RM, Hengartner H. Linking immune-mediated arterial inflammation and cholesterol-induced atherosclerosis in a transgenic mouse model. Proc Natl Acad Sci USA. 2000;97:12752–12757. doi: 10.1073/pnas.220427097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Olofsson PS, Söderström LA, Wågsäter D, Sheikine Y, Ocaya P, Lang F, Rabu C, Chen L, Rudling M, Aukrust P, et al. CD137 is expressed in human atherosclerosis and promotes development of plaque inflammation in hypercholesterolemic mice. Circulation. 2008;117:1292–1301. doi: 10.1161/CIRCULATIONAHA.107.699173 [DOI] [PubMed] [Google Scholar]
- 190.Frostegård J, Ulfgren AK, Nyberg P, Hedin U, Swedenborg J, Andersson U, Hansson GK. Cytokine expression in advanced human atherosclerotic plaques: dominance of pro-inflammatory (Th1) and macrophage-stimulating cytokines. Atherosclerosis. 1999;145:33–43. doi: 10.1016/s0021-9150(99)00011-8 [DOI] [PubMed] [Google Scholar]
- 191.King VL, Szilvassy SJ, Daugherty A. Interleukin-4 deficiency decreases atherosclerotic lesion formation in a site-specific manner in female LDL receptor-/- mice. Arterioscler Thromb Vasc Biol. 2002;22:456–461. doi: 10.1161/hq0302.104905 [DOI] [PubMed] [Google Scholar]
- 192.Eid RE, Rao DA, Zhou J, Lo SL, Ranjbaran H, Gallo A, Sokol SI, Pfau S, Pober JS, Tellides G. Interleukin-17 and interferon-gamma are produced concomitantly by human coronary artery-infiltrating T cells and act synergistically on vascular smooth muscle cells. Circulation. 2009;119:1424–1432. doi: 10.1161/CIRCULATIONAHA.108.827618 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Choi JH, Do Y, Cheong C, Koh H, Boscardin SB, Oh Y-S, Bozzacco L, Trumpfheller C, Park CG, Steinman RM. Identification of antigen-presenting dendritic cells in mouse aorta and cardiac valves. J Exp Med. 2009;206:497–505. doi: 10.1084/jem.20082129 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Madhur MS, Funt SA, Li L, Vinh A, Chen W, Lob HE, Iwakura Y, Blinder Y, Rahman A, Quyyumi AA, et al. Role of interleukin 17 in inflammation, atherosclerosis, and vascular function in apolipoprotein e-deficient mice. Arterioscler Thromb Vasc Biol. 2011;31:1565–1572. doi: 10.1161/ATVBAHA.111.227629 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Xie JJ, Wang J, Tang TT, Chen J, Gao XL, Yuan J, Zhou ZH, Liao MY, Yao R, Yu X, et al. The Th17/Treg functional imbalance during atherogenesis in ApoE(-/-) mice. Cytokine. 2010;49:185–193. doi: 10.1016/j.cyto.2009.09.007 [DOI] [PubMed] [Google Scholar]
- 196.Eivazi S, Bagheri S, Hashemzadeh MS, Ghalavand M, Qamsari ES, Dorostkar R, Yasemi M. Development of T follicular helper cells and their role in disease and immune system. Biomed Pharmacother. 2016;84:1668–1678. doi: 10.1016/j.biopha.2016.10.083 [DOI] [PubMed] [Google Scholar]
- 197.Van Es T, Van Puijvelde GHM, Foks AC, Habets KLL, Bot I, Gilboa E, Van Berkel TJC, Kuiper J. Vaccination against Foxp3+ regulatory T cells aggravates atherosclerosis. Atherosclerosis. 2010;209:74–80. doi: 10.1016/j.atherosclerosis.2009.08.041 [DOI] [PubMed] [Google Scholar]
- 198.Bekker LG, Beyrer C, Mgodi N, Lewin SR, Delany-Moretlwe S, Taiwo B, Masters MC, Lazarus JV. HIV infection. Nat Rev Dis Primers. 2023;9:1–21. doi: 10.1038/s41572-023-00452-3 [DOI] [PubMed] [Google Scholar]
- 199.Kaplan RC, Sinclair E, Landay AL, Lurain N, Sharrett AR, Gange SJ, Xue X, Parrinello CM, Hunt P, Deeks SG, et al. T cell activation predicts carotid artery stiffness among HIV-infected women. Atherosclerosis. 2011;217:207–213. doi: 10.1016/j.atherosclerosis.2011.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Karim R, Mack WJ, Kono N, Tien PC, Anastos K, Lazar J, Young M, Desai S, Golub ET, Kaplan RC, et al. T-cell activation, both pre- and post-HAART levels, correlates with carotid artery stiffness over 6.5 years among HIV-infected women in the WIHS. J Acquir Immune Defic Syndr. 2014;67:349–356. doi: 10.1097/QAI.0000000000000311 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.D’Abramo A, Zingaropoli MA, Oliva A, D'Agostino C, Al Moghazi S, De Luca G, Iannetta M, Mastroianni CM, Vullo V. Immune activation, immunosenescence, and osteoprotegerin as markers of endothelial dysfunction in subclinical HIV-associated atherosclerosis. Mediators Inflamm. 2014;2014:192594. doi: 10.1155/2014/192594 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Merlini E, Luzi K, Suardi E, Barassi A, Cerrone M, Martínez JS, Bai F, D'Eril GVM, Monforte AD, Marchetti G. T-cell phenotypes, apoptosis and inflammation in HIV+ patients on virologically effective cART with early atherosclerosis. PLoS One. 2012;7:e46073. doi: 10.1371/journal.pone.0046073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Kundu S, Freiberg MS, Tracy RP, So-Armah KA, Koethe JR, Duncan MS, Tindle HA, Beckman JA, Feinstein MJ, McDonnell WJ, et al. ; Veterans Aging Cohort Study. Circulating T cells and cardiovascular risk in people with and without HIV infection. J Am Coll Cardiol. 2022;80:1633–1644. doi: 10.1016/j.jacc.2022.08.756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Hsue PY, Hunt PW, Sinclair E, Bredt B, Franklin A, Killian M, Hoh R, Martin JN, McCune JM, Waters DD, et al. Increased carotid intima-media thickness in HIV patients is associated with increased cytomegalovirus-specific T-cell responses. AIDS. 2006;20:2275–2283. doi: 10.1097/QAD.0b013e3280108704 [DOI] [PubMed] [Google Scholar]
- 205.Sacre K, Hunt PW, Hsue PY, Maidji E, Martin JN, Deeks SG, Autran B, McCune JM. A role for CMV-specific CD4+CX3CR1+ T cells and CMV-induced T cell immunopathology in HIV-associated atherosclerosis. AIDS. 2012;26:805–814. doi: 10.1097/QAD.0b013e328351f780 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Panigrahi S, Chen B, Fang M, Potashnikova D, Komissarov AA, Lebedeva A, Michaelson GM, Wyrick JM, Morris SR, Sieg SF, et al. CX3CL1 and IL-15 promote CD8 T cell chemoattraction in HIV and in atherosclerosis. PLoS Pathog. 2020;16:e1008885. doi: 10.1371/journal.ppat.1008885 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Bernal E, Martinez M, Torres A, Guillamón CF, Alcaraz A, Alcaraz MJ, Muñoz A, Valero S, Botella C, Campillo JA, et al. T cell senescence predicts subclinical atherosclerosis in HIV-infected patients similarly to traditional cardiovascular risk factors. Antiviral Res. 2019;162:163–170. doi: 10.1016/j.antiviral.2018.12.014 [DOI] [PubMed] [Google Scholar]
- 208.Wanjalla CN, Mashayekhi M, Bailin S, Gabriel CL, Meenderink LM, Temu T, Fuller DT, Guo L, Kawai K, Virmani R, et al. Anticytomegalovirus CD4+ T cells are associated with subclinical atherosclerosis in persons with HIV. Arterioscler Thromb Vasc Biol. 2021;41:1459–1473. doi: 10.1161/ATVBAHA.120.315786 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Kaplan RC, Kingsley LA, Gange SJ, Benning L, Jacobson LP, Lazar J, Anastos K, Tien PC, Sharrett AR, Hodis HN. Low CD4+ T-cell count as a major atherosclerosis risk factor in HIV-infected women and men. AIDS. 2008;22:1615–1624. doi: 10.1097/QAD.0b013e328300581d [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Mauri C, Bosma A. Immune regulatory function of B cells. Annu Rev Immunol. 2012;30:221–241. doi: 10.1146/annurev-immunol-020711-074934 [DOI] [PubMed] [Google Scholar]
- 211.Houtkamp MA, de Boer OJ, van der Loos CM, van der Wal AC, Becker AE. Adventitial infiltrates associated with advanced atherosclerotic plaques: structural organization suggests generation of local humoral immune responses. J Pathol. 2001;193:263–269. doi: 10.1002/1096-9896(2000)9999:9999<::AID-PATH774>3.0.CO;2-N [DOI] [PubMed] [Google Scholar]
- 212.Burioni R, Canducci F, Saita D, Perotti M, Mancini N, De Marco D, Clementi N, Chieffo A, Denaro M, Cianflone D, et al. Antigen-driven evolution of B lymphocytes in coronary atherosclerotic plaques. J Immunol. 2009;183:2537–2544. doi: 10.4049/jimmunol.0901076 [DOI] [PubMed] [Google Scholar]
- 213.Sage AP, Tsiantoulas D, Binder CJ, Mallat Z. The role of B cells in atherosclerosis. Nat Rev Cardiol. 2019;16:180–196. doi: 10.1038/s41569-018-0106-9 [DOI] [PubMed] [Google Scholar]
- 214.Tsiantoulas D, Diehl CJ, Witztum JL, Binder CJ. B cells and humoral immunity in atherosclerosis. Circ Res. 2014;114:1743–1756. doi: 10.1161/CIRCRESAHA.113.301145 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Prasad A, Clopton P, Ayers C, Khera A, de Lemos JA, Witztum JL, Tsimikas S. Relationship of autoantibodies to MDA-LDL and ApoB-immune complexes to sex, ethnicity, subclinical atherosclerosis and cardiovascular events. Arterioscler Thromb Vasc Biol. 2017;37:1213–1221. doi: 10.1161/ATVBAHA.117.309101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Srikakulapu P, Hu D, Yin C, Mohanta SK, Bontha SV, Peng L, Beer M, Weber C, McNamara CA, Grassia G, et al. Artery tertiary lymphoid organs control multilayered territorialized atherosclerosis B-cell responses in aged ApoE-/- mice. Arterioscler Thromb Vasc Biol. 2016;36:1174–1185. doi: 10.1161/ATVBAHA.115.306983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Tay C, Liu YH, Kanellakis P, Kallies A, Li Y, Cao A, Hosseini H, Tipping P, Toh BH, Bobik A, et al. Follicular B cells promote atherosclerosis via T cell-mediated differentiation into plasma cells and secreting pathogenic immunoglobulin G. Arterioscler Thromb Vasc Biol. 2018;38:e71–e84. doi: 10.1161/ATVBAHA.117.310678 [DOI] [PubMed] [Google Scholar]
- 218.Burger F, Miteva K, Baptista D, Roth A, Fraga-Silva RA, Martel C, Stergiopulos N, Mach F, Brandt KJ. Follicular regulatory helper T cells control the response of regulatory B cells to a high-cholesterol diet. Cardiovasc Res. 2020;117:743–755. doi: 10.1093/cvr/cvaa069 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Nus M, Sage AP, Lu Y, Masters L, Lam BYH, Newland S, Weller S, Tsiantoulas D, Raffort J, Marcus D, et al. Marginal zone B cells control the response of follicular helper T cells to a high-cholesterol diet. Nat Med. 2017;23:601–610. doi: 10.1038/nm.4315 [DOI] [PubMed] [Google Scholar]
- 220.Nus M, Basatemur G, Galan M, Cros-Brunsó L, Zhao TX, Masters L, Harrison J, Figg N, Tsiantoulas D, Geissmann F, et al. NR4A1 deletion in marginal zone B cells exacerbates atherosclerosis in mice—brief report. Arterioscler Thromb Vasc Biol. 2020;40:2598–2604. doi: 10.1161/ATVBAHA.120.314607 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Cerutti A, Cols M, Puga I. Marginal zone B cells: virtues of innate-like antibody-producing lymphocytes. Nat Rev Immunol. 2013;13:118–132. doi: 10.1038/nri3383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Moir S, Fauci AS. Pathogenic mechanisms of B-lymphocyte dysfunction in HIV disease. J Allergy Clin Immunol. 2008;122:12–9; quiz 20. doi: 10.1016/j.jaci.2008.04.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Moir S, Fauci AS. B cells in HIV infection and disease. Nat Rev Immunol. 2009;9:235–245. doi: 10.1038/nri2524 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Moir S, Fauci AS. B-cell responses to HIV infection. Immunol Rev. 2017;275:33–48. doi: 10.1111/imr.12502 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Sage AP, Tsiantoulas D, Baker L, Harrison J, Masters L, Murphy D, Loinard C, Binder CJ, Mallat Z. BAFF receptor deficiency reduces the development of atherosclerosis in mice—brief report. Arterioscler Thromb Vasc Biol. 2012;32:1573–1576. doi: 10.1161/ATVBAHA.111.244731 [DOI] [PubMed] [Google Scholar]
- 226.Mackay F, Browning JL. BAFF: a fundamental survival factor for B cells. Nat Rev Immunol. 2002;2:465–475. doi: 10.1038/nri844 [DOI] [PubMed] [Google Scholar]
- 227.Chagnon-Choquet J, Gauvin J, Roger J, Fontaine J, Poudrier J, Roger M; Montreal Primary HIV Infection and Slow Progressor Study Groups. HIV nef promotes expression of B-lymphocyte stimulator by blood dendritic cells during HIV infection in humans. J Infect Dis. 2015;211:1229–1240. doi: 10.1093/infdis/jiu611 [DOI] [PubMed] [Google Scholar]
- 228.Aranguren M, Doyon-Laliberté K, El-Far M, Chartrand-Lefebvre C, Routy JP, Barril JG, Trottier B, Tremblay C, Durand M, Poudrier J, et al. ; Canadian HIV and Aging Cohort Study. Subclinical atherosclerosis is associated with discrepancies in BAFF and APRIL levels and altered Breg potential of precursor-like marginal zone B-cells in long-term HIV treated individuals. Vaccines (Basel). 2022;11:81. doi: 10.3390/vaccines11010081 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Pizzino G, Irrera N, Cucinotta M, Pallio G, Mannino F, Arcoraci V, Squadrito F, Altavilla D, Bitto A. Oxidative stress: harms and benefits for human health. Oxid Med Cell Longev. 2017;2017:8416763. doi: 10.1155/2017/8416763 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Ivanov AV, Valuev-Elliston VT, Ivanova ON, Kochetkov SN, Starodubova ES, Bartosch B, Isaguliants MG. Oxidative stress during HIV infection: mechanisms and consequences. Oxid Med Cell Longev. 2016;2016:8910396. doi: 10.1155/2016/8910396 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Moore K, Sheedy F, Fisher E. Macrophages in atherosclerosis: a dynamic balance. Nat Rev Immunol. 2013;13:709–721. doi: 10.1038/nri3520 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Hileman CO, Turner R, Funderburg NT, Semba RD, McComsey GA. Changes in oxidized lipids drive the improvement in monocyte activation and vascular disease after statin therapy in HIV. AIDS. 2016;30:65–73. doi: 10.1097/QAD.0000000000000885 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Vellas C, Nayrac M, Collercandy N, Requena M, Jeanne N, Latour J, Dimeglio C, Cazabat M, Barange K, Alric L, et al. Intact proviruses are enriched in the colon and associated with PD-1+TIGIT- mucosal CD4+ T cells of people with HIV-1 on antiretroviral therapy. EBioMedicine. 2023;100:104954. doi: 10.1016/j.ebiom.2023.104954 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Brenchley JM, Schacker TW, Ruff LE, Price DA, Taylor JH, Beilman GJ, Nguyen PL, Khoruts A, Larson M, Haase AT, et al. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J Exp Med. 2004;200:749–759. doi: 10.1084/jem.20040874 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Yoo JY, Groer M, Dutra SVO, Sarkar A, McSkimming DI. Gut microbiota and immune system interactions. Microorganisms. 2020;8:1587. doi: 10.3390/microorganisms8101587 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Jiang W, Lederman MM, Hunt P, Sieg SF, Haley K, Rodriguez B, Landay A, Martin J, Sinclair E, Asher AI, et al. Plasma levels of bacterial DNA correlate with immune activation and the magnitude of immune restoration in persons with antiretroviral-treated HIV infection. J Infect Dis. 2009;199:1177–1185. doi: 10.1086/597476 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Zanoni BC, Gandhi RT. Update on opportunistic infections in the era of effective antiretroviral therapy. Infect Dis Clin North Am. 2014;28:501–518. doi: 10.1016/j.idc.2014.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Lake JE, Tseng CH, Currier JS. A pilot study of telmisartan for visceral adiposity in HIV infection: the metabolic abnormalities, telmisartan, and HIV infection (MATH) trial. PLoS One. 2013;8:e58135. doi: 10.1371/journal.pone.0058135 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Deeks SG, Lewin SR, Havlir DV. The end of AIDS: HIV infection as a chronic disease. Lancet. 2013;382:1525–1533. doi: 10.1016/S0140-6736(13)61809-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Ricotta D, Alessandri G, Pollara C, Fiorentini S, Favilli F, Tosetti M, Mantovani A, Grassi M, Garrafa E, Dei Cas L, et al. Adult human heart microvascular endothelial cells are permissive for non-lytic infection by human cytomegalovirus. Cardiovasc Res. 2001;49:440–448. doi: 10.1016/s0008-6363(00)00258-3 [DOI] [PubMed] [Google Scholar]
- 241.Smiley ML, Mar EC, Huang ES. Cytomegalovirus infection and viral-induced transformation of human endothelial cells. J Med Virol. 1988;25:213–226. doi: 10.1002/jmv.1890250212 [DOI] [PubMed] [Google Scholar]
- 242.Khan N, Hislop A, Gudgeon N, Cobbold M, Khanna R, Nayak L, Rickinson AB, Moss PAH. Herpesvirus-specific CD8 T cell immunity in old age: cytomegalovirus impairs the response to a coresident EBV infection. J Immunol. 2004;173:7481–7489. doi: 10.4049/jimmunol.173.12.7481 [DOI] [PubMed] [Google Scholar]
- 243.Morgan MD, Pachnio A, Begum J, Roberts D, Rasmussen N, Neil DAH, Bajema I, Savage COS, Moss PA, Harper L. CD4+CD28- T cell expansion in granulomatosis with polyangiitis (Wegener’s) is driven by latent cytomegalovirus infection and is associated with an increased risk of infection and mortality. Arthritis Rheum. 2011;63:2127–2137. doi: 10.1002/art.30366 [DOI] [PubMed] [Google Scholar]
- 244.Sester M, Sester U, Gärtner B, Kubuschok B, Girndt M, Meyerhans A, Köhler H. Sustained high frequencies of specific CD4 T cells restricted to a single persistent virus. J Virol. 2002;76:3748–3755. doi: 10.1128/jvi.76.8.3748-3755.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Spyridopoulos I, Martin-Ruiz C, Hilkens C, Yadegarfar ME, Isaacs J, Jagger C, Kirkwood T, von Zglinicki T. CMV seropositivity and T-cell senescence predict increased cardiovascular mortality in octogenarians: results from the Newcastle 85+ study. Aging Cell. 2016;15:389–392. doi: 10.1111/acel.12430 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Lichtner M, Cicconi P, Vita S, Cozzi-Lepri A, Galli M, Lo Caputo S, Saracino A, De Luca A, Moioli M, Maggiolo F, et al. ; ICONA Foundation Study. Cytomegalovirus coinfection is associated with an increased risk of severe non-AIDS-defining events in a large cohort of HIV-infected patients. J Infect Dis. 2015;211:178–186. doi: 10.1093/infdis/jiu417 [DOI] [PubMed] [Google Scholar]
- 247.Adam E, Melnick JL, Probtsfield JL, Petrie BL, Burek J, Bailey KR, McCollum CH, DeBakey ME. High levels of cytomegalovirus antibody in patients requiring vascular surgery for atherosclerosis. Lancet. 1987;2:291–293. doi: 10.1016/s0140-6736(87)90888-9 [DOI] [PubMed] [Google Scholar]
- 248.Nieto FJ, Adam E, Sorlie P, Farzadegan H, Melnick JL, Comstock GW, Szklo M. Cohort study of cytomegalovirus infection as a risk factor for carotid intimal-medial thickening, a measure of subclinical atherosclerosis. Circulation. 1996;94:922–927. doi: 10.1161/01.cir.94.5.922 [DOI] [PubMed] [Google Scholar]
- 249.Simanek AM, Dowd JB, Pawelec G, Melzer D, Dutta A, Aiello AE. Seropositivity to cytomegalovirus, inflammation, all-cause and cardiovascular disease-related mortality in the United States. PLoS One. 2011;6:e16103. doi: 10.1371/journal.pone.0016103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Sorlie PD, Nieto FJ, Adam E, Folsom AR, Shahar E, Massing M. A prospective study of cytomegalovirus, herpes simplex virus 1, and coronary heart disease: the atherosclerosis risk in communities (ARIC) study. Arch Intern Med. 2000;160:2027–2032. doi: 10.1001/archinte.160.13.2027 [DOI] [PubMed] [Google Scholar]
- 251.Parrinello CM, Sinclair E, Landay AL, Lurain N, Sharrett AR, Gange SJ, Xue X, Hunt PW, Deeks SG, Hodis HN, et al. Cytomegalovirus immunoglobulin G antibody is associated with subclinical carotid artery disease among HIV-infected women. J Infect Dis. 2012;205:1788–1796. doi: 10.1093/infdis/jis276 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Freeman ML, Mudd JC, Shive CL, Younes S-A, Panigrahi S, Sieg SF, Lee SA, Hunt PW, Calabrese LH, Gianella S, et al. CD8 T-cell expansion and inflammation linked to CMV coinfection in ART-treated HIV infection. Clin Infect Dis. 2016;62:392–396. doi: 10.1093/cid/civ840 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Bolovan-Fritts CA, Trout RN, Spector SA. Human cytomegalovirus-specific CD4+-T-cell cytokine response induces fractalkine in endothelial cells. J Virol. 2004;78:13173–13181. doi: 10.1128/JVI.78.23.13173-13181.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Imai T, Hieshima K, Haskell C, Baba M, Nagira M, Nishimura M, Kakizaki M, Takagi S, Nomiyama H, Schall TJ, et al. Identification and molecular characterization of fractalkine receptor CX3CR1, which mediates both leukocyte migration and adhesion. Cell. 1997;91:521–530. doi: 10.1016/s0092-8674(00)80438-9 [DOI] [PubMed] [Google Scholar]
- 255.Nishimura M, Umehara H, Nakayama T, Yoneda O, Hieshima K, Kakizaki M, Dohmae N, Yoshie O, Imai T. Dual functions of fractalkine/CX3C ligand 1 in trafficking of perforin+/granzyme B+ cytotoxic effector lymphocytes that are defined by CX3CR1 expression. J Immunol. 2002;168:6173–6180. doi: 10.4049/jimmunol.168.12.6173 [DOI] [PubMed] [Google Scholar]
- 256.Wanjalla CN, McDonnell WJ, Ram R, Chopra A, Gangula R, Leary S, Mashayekhi M, Simmons JD, Warren CM, Bailin S, et al. Single-cell analysis shows that adipose tissue of persons with both HIV and diabetes is enriched for clonal, cytotoxic, and CMV-specific CD4+ T cells. Cell Rep Med. 2021;2:100205. doi: 10.1016/j.xcrm.2021.100205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Wanjalla CN, Gabriel CL, Fuseini H, Bailin SS, Mashayekhi M, Simmons J, Warren CM, Glass DR, Oakes J, Gangula R, et al. CD4+ T cells expressing CX3CR1, GPR56, with variable CD57 are associated with cardiometabolic diseases in persons with HIV. Front Immunol. 2023;14:1099356. doi: 10.3389/fimmu.2023.1099356 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Peng YM, van de Garde MDB, Cheng KF, Baars PA, Remmerswaal EBM, van Lier RAW, Mackay CR, Lin HH, Hamann J. Specific expression of GPR56 by human cytotoxic lymphocytes. J Leukoc Biol. 2011;90:735–740. doi: 10.1189/jlb.0211092 [DOI] [PubMed] [Google Scholar]
- 259.Tian Y, Babor M, Lane J, Schulten V, Patil VS, Seumois G, Rosales SL, Fu Z, Picarda G, Burel J, et al. Unique phenotypes and clonal expansions of human CD4 effector memory T cells re-expressing CD45RA. Nat Commun. 2017;8:1473. doi: 10.1038/s41467-017-01728-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Chen B, Morris SR, Panigrahi S, Michaelson GM, Wyrick JM, Komissarov AA, Potashnikova D, Lebedeva A, Younes S-A, Harth K, et al. Cytomegalovirus coinfection is associated with increased vascular-homing CD57+ CD4 T cells in HIV infection. J Immunol. 2020;204:2722–2733. doi: 10.4049/jimmunol.1900734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Hunt PW, Martin JN, Sinclair E, Epling L, Teague J, Jacobson MA, Tracy RP, Corey L, Deeks SG. Valganciclovir reduces T cell activation in HIV-infected individuals with incomplete CD4+ T cell recovery on antiretroviral therapy. J Infect Dis. 2011;203:1474–1483. doi: 10.1093/infdis/jir060 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Chanouzas D, Sagmeister M, Faustini S, Nightingale P, Richter A, Ferro CJ, Morgan MD, Moss P, Harper L. Subclinical reactivation of cytomegalovirus drives CD4+CD28null T-cell expansion and impaired immune response to pneumococcal vaccination in antineutrophil cytoplasmic antibody-associated vasculitis. J Infect Dis. 2019;219:234–244. doi: 10.1093/infdis/jiy493 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Chanouzas D, Dyall L, Dale J, Moss P, Morgan M, Harper L. CD4+CD28- T-cell expansions in ANCA-associated vasculitis and association with arterial stiffness: baseline data from a randomised controlled trial. Lancet. 2015;385(Suppl 1):S30. doi: 10.1016/S0140-6736(15)60345-2 [DOI] [PubMed] [Google Scholar]
- 264.Kim AY, Onofrey S, Church DR. An epidemiologic update on hepatitis C infection in persons living with or at risk of HIV infection. J Infect Dis. 2013;207(Suppl 1):S1–S6. doi: 10.1093/infdis/jis927 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Butt AA, Xiaoqiang W, Budoff M, Leaf D, Kuller LH, Justice AC. Hepatitis C virus infection and the risk of coronary disease. Clin Infect Dis. 2009;49:225–232. doi: 10.1086/599371 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Freiberg MS, Cheng DM, Kraemer KL, Saitz R, Kuller LH, Samet JH. The association between hepatitis C infection and prevalent cardiovascular disease among HIV-infected individuals. AIDS. 2007;21:193–197. doi: 10.1097/QAD.0b013e3280118a0d [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Pothineni NVKC, Delongchamp R, Vallurupalli S, Ding Z, Dai Y, Hagedorn CH, Mehta JL. Impact of hepatitis C seropositivity on the risk of coronary heart disease events. Am J Cardiol. 2014;114:1841–1845. doi: 10.1016/j.amjcard.2014.09.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Fernández-Montero JV, Barreiro P, de Mendoza C, Labarga P, Soriano V. Hepatitis C virus coinfection independently increases the risk of cardiovascular disease in HIV-positive patients. J Viral Hepat. 2016;23:47–52. doi: 10.1111/jvh.12447 [DOI] [PubMed] [Google Scholar]
- 269.Peters L, Neuhaus J, Duprez D, Neaton JD, Tracy R, Klein MB, Mocroft A, Rockstroh J, Dore G, Lundgren JD; INSIGHT SMART Study Group. Biomarkers of inflammation, coagulation and microbial translocation in HIV/HCV co-infected patients in the SMART study. J Clin Virol. 2014;60:295–300. doi: 10.1016/j.jcv.2014.03.017 [DOI] [PubMed] [Google Scholar]
- 270.Márquez M, Romero-Cores P, Montes-Oca M, Martín-Aspas A, Soto-Cárdenas MJ, Guerrero F, Fernández-Gutiérrez C, Girón-González JA. Immune activation response in chronic HIV-infected patients: influence of Hepatitis C virus coinfection. PLoS One. 2015;10:e0119568. doi: 10.1371/journal.pone.0119568 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Kushner LE, Wendelboe AM, Lazzeroni LC, Chary A, Winters MA, Osinusi A, Kottilil S, Polis MA, Holodniy M. Immune biomarker differences and changes comparing HCV mono-infected, HIV/HCV co-infected, and HCV spontaneously cleared patients. PLoS One. 2013;8:e60387. doi: 10.1371/journal.pone.0060387 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Chew KW, Hua L, Bhattacharya D, Butt AA, Bornfleth L, Chung RT, Andersen JW, Currier JS. The effect of hepatitis C virologic clearance on cardiovascular disease biomarkers in human immunodeficiency virus/hepatitis C virus coinfection. Open Forum Infect Dis. 2014;1:ofu104. doi: 10.1093/ofid/ofu104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.LaVergne S, Umlauf A, McCutchan A, Heaton R, Benson C, Kumarasamy N, Bharti AR. Impact of latent tuberculosis infection on neurocognitive functioning and inflammation in HIV-infected and uninfected South Indians. J Acquir Immune Defic Syndr. 2020;84:430–436. doi: 10.1097/QAI.0000000000002368 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Mariotti S, Pardini M, Gagliardi MC, Teloni R, Giannoni F, Fraziano M, Lozupone F, Meschini S, Nisini R. Dormant Mycobacterium tuberculosis fails to block phagosome maturation and shows unexpected capacity to stimulate specific human T lymphocytes. J Immunol. 2013;191:274–282. doi: 10.4049/jimmunol.1202900 [DOI] [PubMed] [Google Scholar]
- 275.Sullivan ZA, Wong EB, Ndung’u T, Kasprowicz VO, Bishai WR. Latent and active tuberculosis infection increase immune activation in individuals co-infected with HIV. EBioMedicine. 2015;2:334–340. doi: 10.1016/j.ebiom.2015.03.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Temu TM, Polyak SJ, Wanjalla CN, Mandela NA, Dabee S, Mogaka JN, Masyuko S, Longernecker C, Shakil S, Chohan B, et al. Latent tuberculosis is associated with heightened levels of pro-and anti-inflammatory cytokines among Kenyan men and women living with HIV on long-term antiretroviral therapy. AIDS. 2023;37:1065–1075. doi: 10.1097/QAD.0000000000003523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Toossi Z, Funderburg NT, Sirdeshmuk S, Whalen CC, Nanteza MW, Johnson DF, Mayanja-Kizza H, Hirsch CS. Systemic immune activation and microbial translocation in dual HIV/tuberculosis-infected subjects. J Infect Dis. 2013;207:1841–1849. doi: 10.1093/infdis/jit092 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Boulware DR, Hullsiek KH, Puronen CE, Rupert A, Baker JV, French MA, Bohjanen PR, Novak RM, Neaton JD, Sereti I; INSIGHT Study Group. Higher levels of CRP, D-dimer, IL-6, and hyaluronic acid before initiation of antiretroviral therapy (ART) are associated with increased risk of AIDS or death. J Infect Dis. 2011;203:1637–1646. doi: 10.1093/infdis/jir134 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Sandler NG, Wand H, Roque A, Law M, Nason MC, Nixon DE, Pedersen C, Ruxrungtham K, Lewin SR, Emery S, et al. ; INSIGHT SMART Study Group. Plasma levels of soluble CD14 independently predict mortality in HIV infection. J Infect Dis. 2011;203:780–790. doi: 10.1093/infdis/jiq118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Karbach S, Croxford AL, Oelze M, Schüler R, Minwegen D, Wegner J, Koukes L, Yogev N, Nikolaev A, Reißig S, et al. Interleukin 17 drives vascular inflammation, endothelial dysfunction, and arterial hypertension in psoriasis-like skin disease. Arterioscler Thromb Vasc Biol. 2014;34:2658–2668. doi: 10.1161/ATVBAHA.114.304108 [DOI] [PubMed] [Google Scholar]
- 281.Wanjalla CN, Temu TM, Mashayekhi M, Warren CM, Shepherd BE, Gangula R, Fuseini H, Bailin S, Gabriel CL, Gangula P, et al. Interleukin-17A is associated with flow-mediated dilation and interleukin-4 with carotid plaque in persons with HIV. AIDS. 2022;36:963–973. doi: 10.1097/QAD.0000000000003196 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Zhang H, Park Y, Wu J, Chen X, Lee S, Yang J, Dellsperger KC, Zhang C. Role of TNF-alpha in vascular dysfunction. Clin Sci (Lond). 2009;116:219–230. doi: 10.1042/CS20080196 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Nagot N, Ouédraogo A, Foulongne V, Konaté I, Weiss HA, Vergne L, Defer MC, Djagbaré D, Sanon A, Andonaba JB, et al. ; ANRS 1285 Study Group. Reduction of HIV-1 RNA levels with therapy to suppress herpes simplex virus. N Engl J Med. 2007;356:790–799. doi: 10.1056/NEJMoa062607 [DOI] [PubMed] [Google Scholar]
- 284.Redd AD, Newell K, Patel EU, Nalugoda F, Ssebbowa P, Kalibbala S, Frank MA, Tobian AAR, Gray RH, Quinn TC, et al. Decreased monocyte activation with daily acyclovir use in HIV-1/HSV-2 coinfected women. Sex Transm Infect. 2015;91:485–488. doi: 10.1136/sextrans-2014-051867 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Hechter RC, Budoff M, Hodis HN, Rinaldo CR, Jenkins FJ, Jacobson LP, Kingsley LA, Taiwo B, Post WS, Margolick JB, et al. Herpes simplex virus type 2 (HSV-2) as a coronary atherosclerosis risk factor in HIV-infected men: multicenter AIDS cohort study. Atherosclerosis. 2012;223:433–436. doi: 10.1016/j.atherosclerosis.2012.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Mendy A, Vieira ER, Gasana J. Seropositivity to herpes simplex virus type 2, but not type 1 is associated with premature cardiovascular diseases: a population-based cross-sectional study. Atherosclerosis. 2013;231:18–21. doi: 10.1016/j.atherosclerosis.2013.08.020 [DOI] [PubMed] [Google Scholar]
- 287.Alber DG, Powell KL, Vallance P, Goodwin DA, Grahame-Clarke C. Herpesvirus infection accelerates atherosclerosis in the apolipoprotein E–deficient mouse. Circulation. 2000;102:779–785. doi: 10.1161/01.cir.102.7.779 [DOI] [PubMed] [Google Scholar]
- 288.Grahame-Clarke C, Alber DG, Lucas SB, Miller R, Vallance P. Association between Kaposi’s sarcoma and atherosclerosis: implications for gammaherpesviruses and vascular disease. AIDS. 2001;15:1902–1904. doi: 10.1097/00002030-200109280-00029 [DOI] [PubMed] [Google Scholar]
- 289.Casper C, Meier AS, Wald A, Morrow RA, Corey L, Moscicki AB. Human herpesvirus 8 infection among adolescents in the REACH cohort. Arch Pediatr Adolesc Med. 2006;160:937–942. doi: 10.1001/archpedi.160.9.937 [DOI] [PubMed] [Google Scholar]
- 290.Kedes DH, Operskalski E, Busch M, Kohn R, Flood J, Ganem D. The seroepidemiology of human herpesvirus 8 (Kaposi’s sarcoma-associated herpesvirus): distribution of infection in KS risk groups and evidence for sexual transmission. Nat Med. 1996;2:918–924. doi: 10.1038/nm0896-918 [DOI] [PubMed] [Google Scholar]
- 291.Renwick N, Halaby T, Weverling GJ, Dukers NH, Simpson GR, Coutinho RA, Lange JM, Schulz TF, Goudsmit J. Seroconversion for human herpesvirus 8 during HIV infection is highly predictive of Kaposi’s sarcoma. AIDS. 1998;12:2481–2488. doi: 10.1097/00002030-199818000-00018 [DOI] [PubMed] [Google Scholar]
- 292.Masiá M, Robledano C, Ortiz de la Tabla V, Antequera P, Lumbreras B, Hernández I, Gutiérrez F. Coinfection with human herpesvirus 8 is associated with persistent inflammation and immune activation in virologically suppressed HIV-infected patients. PLoS One. 2014;9:e105442. doi: 10.1371/journal.pone.0105442 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Lidón F, Padilla S, García JA, Fernández M, García J, Ortiz de la Tabla V, Gutiérrez F, Masiá M. Contribution of human herpesvirus 8 and herpes simplex type 2 to progression of carotid intima-media thickness in people living with HIV. Open Forum Infect Dis. 2019;6:ofz041. doi: 10.1093/ofid/ofz041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Martínez E, D’Albuquerque PM, Llibre JM, Gutierrez F, Podzamczer D, Antela A, Berenguer J, Domingo P, Moreno X, Perez I, et al. ; SPIRAL Trial Group. Changes in cardiovascular biomarkers in HIV-infected patients switching from ritonavir-boosted protease inhibitors to raltegravir. AIDS. 2012;26:2315–2326. doi: 10.1097/QAD.0b013e328359f29c [DOI] [PubMed] [Google Scholar]
- 295.Lundgren JD, Babiker AG, Sharma S, Grund B, Phillips AN, Matthews G, Kan VL, Aagaard B, Abo I, Alston B, et al. ; INSIGHT Strategic Timing of AntiRetroviral Treatment (START) Study Group. Long-term benefits from early antiretroviral therapy initiation in HIV infection. NEJM Evid. 2023;2:10.1056/evidoa2200302. doi: 10.1056/evidoa2200302 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Marcus JL, Leyden WA, Chao CR, Chow FC, Horberg MA, Hurley LB, Klein DB, Quesenberry CP, Towner WJ, Silverberg MJ. HIV infection and incidence of ischemic stroke. AIDS. 2014;28:1911–1919. doi: 10.1097/QAD.0000000000000352 [DOI] [PubMed] [Google Scholar]
- 297.Friis-Møller N, Sabin CA, Weber R, d'Arminio Monforte A, El-Sadr WM, Reiss P, Thiébaut R, Morfeldt L, De Wit S, Pradier C, et al. ; Data Collection on Adverse Events of Anti-HIV Drugs (DAD) Study Group. Combination antiretroviral therapy and the risk of myocardial infarction. N Engl J Med. 2003;349:1993–2003. doi: 10.1056/NEJMoa030218 [DOI] [PubMed] [Google Scholar]
- 298.Mary-Krause M, Cotte L, Simon A, Partisani M, Costagliola D; Clinical Epidemiology Group From the French Hospital Database. Increased risk of myocardial infarction with duration of protease inhibitor therapy in HIV-infected men. AIDS. 2003;17:2479–2486. doi: 10.1097/00002030-200311210-00010 [DOI] [PubMed] [Google Scholar]
- 299.Osoti A, Temu TM, Kirui N, Ngetich EK, Kamano JH, Page S, Farquhar C, Bloomfield GS. Metabolic syndrome among antiretroviral therapy-naive versus experienced HIV-infected patients without preexisting cardiometabolic disorders in Western Kenya. AIDS Patient Care STDS. 2018;32:215–222. doi: 10.1089/apc.2018.0052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Nolan D, Hammond E, James I, McKinnon E, Mallal S. Contribution of nucleoside-analogue reverse transcriptase inhibitor therapy to lipoatrophy from the population to the cellular level. Antivir Ther. 2003;8:617–626. doi: 10.1177/135965350300800614 [PubMed] [Google Scholar]
- 301.Arribas JR, Thompson M, Sax PE, Haas B, McDonald C, Wohl DA, DeJesus E, Clarke AE, Guo S, Wang H, et al. Brief report: randomized, double-blind comparison of tenofovir alafenamide (TAF) vs tenofovir disoproxil fumarate (TDF), each coformulated with elvitegravir, cobicistat, and emtricitabine (E/C/F) for initial HIV-1 treatment: week 144 results. J Acquir Immune Defic Syndr. 2017;75:211–218. doi: 10.1097/QAI.0000000000001350 [DOI] [PubMed] [Google Scholar]
- 302.Norwood J, Turner M, Bofill C, Rebeiro P, Shepherd B, Bebawy S, Hulgan T, Raffanti S, Haas DW, Sterling TR, et al. Brief report: weight gain in persons with HIV switched from efavirenz-based to integrase strand transfer inhibitor-based regimens. J Acquir Immune Defic Syndr. 2017;76:527–531. doi: 10.1097/QAI.0000000000001525 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Usach I, Melis V, Peris JE. Non-nucleoside reverse transcriptase inhibitors: a review on pharmacokinetics, pharmacodynamics, safety and tolerability. J Int AIDS Soc. 2013;16:18567. doi: 10.7448/IAS.16.1.18567 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Dressman J, Kincer J, Matveev SV, Guo L, Greenberg RN, Guerin T, Meade D, Li XA, Zhu W, Uittenbogaard A, et al. HIV protease inhibitors promote atherosclerotic lesion formation independent of dyslipidemia by increasing CD36-dependent cholesteryl ester accumulation in macrophages. J Clin Invest. 2003;111:389–397. doi: 10.1172/JCI16261 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Sribenjalux W, Nuntawit T, Meesing A, Chetchotisakd P. Virological outcomes and metabolic effects after switching from ritonavir-boosted protease inhibitors to a dolutegravir-based regimen in virologically suppressed patients living with HIV. Int J STD AIDS. 2023;34:98–107. doi: 10.1177/09564624221137972 [DOI] [PubMed] [Google Scholar]
- 306.Wang X, Chai H, Yao Q, Chen C. Molecular mechanisms of HIV protease inhibitor-induced endothelial dysfunction. J Acquir Immune Defic Syndr. 2007;44:493–499. doi: 10.1097/QAI.0b013e3180322542 [DOI] [PubMed] [Google Scholar]
- 307.González-Cordón A, Assoumou L, Moyle G, Waters L, Johnson M, Domingo P, Fox J, Stellbrink H-J, Guaraldi G, Masiá M, et al. ; NEAT 022 Study Group. Long-term effects on subclinical cardiovascular disease of switching from boosted protease inhibitors to dolutegravir. J Antimicrob Chemother. 2023;78:2361–2365. doi: 10.1093/jac/dkad247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308.Koethe JR, Lagathu C, Lake JE, Domingo P, Calmy A, Falutz J, Brown TT, Capeau J. HIV and antiretroviral therapy-related fat alterations. Nat Rev Dis Primers. 2020;6:1–20. doi: 10.1038/s41572-020-0181-1 [DOI] [PubMed] [Google Scholar]
- 309.Saint-Marc T, Partisani M, Poizot-Martin I, Bruno F, Rouviere O, Lang JM, Gastaut JA, Touraine JL. A syndrome of peripheral fat wasting (lipodystrophy) in patients receiving long-term nucleoside analogue therapy. AIDS. 1999;13:1659–1667. doi: 10.1097/00002030-199909100-00009 [DOI] [PubMed] [Google Scholar]
- 310.Gelpi M, Afzal S, Fuchs A, Lundgren J, Knudsen AD, Drivsholm N, Mocroft A, Lebech A-M, Lindegaard B, Kühl JT, et al. Prior exposure to thymidine analogs and didanosine is associated with long-lasting alterations in adipose tissue distribution and cardiovascular risk factors. AIDS. 2019;33:675–683. doi: 10.1097/QAD.0000000000002119 [DOI] [PubMed] [Google Scholar]
- 311.Deveaud C, Beauvoit B, Hagry S, Galinier A, Carrière A, Salin B, Schaeffer J, Caspar-Bauguil S, Fernandez Y, Gordien JB, et al. Site specific alterations of adipose tissue mitochondria in 3’-azido-3’-deoxythymidine (AZT)-treated rats: an early stage in lipodystrophy? Biochem Pharmacol. 2005;70:90–101. doi: 10.1016/j.bcp.2005.04.015 [DOI] [PubMed] [Google Scholar]
- 312.Gorwood J, Bourgeois C, Pourcher V, Pourcher G, Charlotte F, Mantecon M, Rose C, Morichon R, Atlan M, Le Grand R, et al. The integrase inhibitors dolutegravir and raltegravir exert proadipogenic and profibrotic effects and induce insulin resistance in human/simian adipose tissue and human adipocytes. Clin Infect Dis. 2020;71:e549–e560. doi: 10.1093/cid/ciaa259 [DOI] [PubMed] [Google Scholar]
- 313.Kouanfack C, Mpoudi-Etame M, Omgba Bassega P, Eymard-Duvernay S, Leroy S, Boyer S, Peeters M, Calmy A, Delaporte E; NAMSAL ANRS 12313 Study Group. Dolutegravir-based or low-dose efavirenz-based regimen for the treatment of HIV-1. N Engl J Med. 2019;381:816–826. doi: 10.1056/NEJMoa1904340 [DOI] [PubMed] [Google Scholar]
- 314.Venter WDF, Moorhouse M, Sokhela S, Fairlie L, Mashabane N, Masenya M, Serenata C, Akpomiemie G, Qavi A, Chandiwana N, et al. Dolutegravir plus two different prodrugs of tenofovir to treat HIV. N Engl J Med. 2019;381:803–815. doi: 10.1056/NEJMoa1902824 [DOI] [PubMed] [Google Scholar]
- 315.Griesel R, Maartens G, Chirehwa M, Sokhela S, Akpomiemie G, Moorhouse M, Venter F, Sinxadi P. CYP2B6 genotype and weight gain differences between dolutegravir and efavirenz. Clin Infect Dis. 2021;73:e3902–e3909. doi: 10.1093/cid/ciaa1073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316.Mallon PW, Brunet L, Hsu RK, Fusco JS, Mounzer KC, Prajapati G, Beyer AP, Wohlfeiler MB, Fusco GP. Weight gain before and after switch from TDF to TAF in a U.S. cohort study. J Int AIDS Soc. 2021;24:e25702. doi: 10.1002/jia2.25702 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Waters L, Assoumou L, González-Cordón A, Rusconi S, Domingo P, Gompels M, de Wit S, Raffi F, Stephan C, Masiá M, et al. ; NEAT 022 Study Group. Limited weight impact after switching from boosted protease inhibitors to dolutegravir in persons with human immunodeficiency virus with high cardiovascular risk: a post hoc analysis of the 96-week NEAT-022 randomized trial. Clin Infect Dis. 2023;76:861–870. doi: 10.1093/cid/ciac827 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 318.Eckard AR, McComsey GA. Weight gain and integrase inhibitors. Curr Opin Infect Dis. 2020;33:10–19. doi: 10.1097/QCO.0000000000000616 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319.Lake JE, Trevillyan J. Impact of Integrase inhibitors and tenofovir alafenamide on weight gain in people with HIV. Curr Opin HIV AIDS. 2021;16:148–151. doi: 10.1097/COH.0000000000000680 [DOI] [PubMed] [Google Scholar]
- 320.McCann K, Shah S, Hindley L, Hill A, Qavi A, Simmons B, Serenata C, Sokhela S, Venter WDF. Implications of weight gain with newer anti-retrovirals: 10-year predictions of cardiovascular disease and diabetes. AIDS. 2021;35:1657–1665. doi: 10.1097/QAD.0000000000002930 [DOI] [PubMed] [Google Scholar]
- 321.Lundgren JD, Babiker AG, Gordin F, Emery S, Grund B, Sharma S, Avihingsanon A, Cooper DA, Fätkenheuer G, Llibre JM, et al. ; INSIGHT START Study Group. Initiation of antiretroviral therapy in early asymptomatic HIV infection. N Engl J Med. 2015;373:795–807. doi: 10.1056/NEJMoa1506816 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322.El-Sadr WM, Lundgren JD, Neaton JD, Gordin F, Abrams D, Arduino RC, Babiker A, Burman W, Clumeck N, Cohen CJ, et al. ; Strategies for Management of Antiretroviral Therapy (SMART) Study Group. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med. 2006;355:2283–2296. doi: 10.1056/NEJMoa062360 [DOI] [PubMed] [Google Scholar]
- 323.National Institute of Allergy and Infectious Diseases (NIAID). Effect of Reducing Inflammation With Low Dose Methotrexate on Inflammatory Markers and Endothelial Function in Treated and Suppressed HIV Infection. clinicaltrials.gov; 2021. Accessed December 31, 2023. https://clinicaltrials.gov/study/NCT01949116 [Google Scholar]
- 324.Ridker PM, Everett BM, Pradhan A, MacFadyen JG, Solomon DH, Zaharris E, Mam V, Hasan A, Rosenberg Y, Iturriaga E, et al. ; CIRT Investigators. Low-dose methotrexate for the prevention of atherosclerotic events. N Engl J Med. 2019;380:752–762. doi: 10.1056/NEJMoa1809798 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325.Hsue PY, Ribaudo HJ, Deeks SG, Bell T, Ridker PM, Fichtenbaum C, Daar ES, Havlir D, Yeh E, Tawakol A, et al. Safety and impact of low-dose methotrexate on endothelial function and inflammation in individuals with treated human immunodeficiency virus: AIDS clinical trials group study A5314. Clin Infect Dis. 2019;68:1877–1886. doi: 10.1093/cid/ciy781 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326.O’Brien MP, Hunt PW, Kitch DW, Klingman K, Stein JH, Funderburg NT, Berger JS, Tebas P, Clagett B, Moisi D, et al. A randomized placebo controlled trial of aspirin effects on immune activation in chronically human immunodeficiency virus-infected adults on virologically suppressive antiretroviral therapy. Open Forum Infect Dis. 2017;4:ofw278. doi: 10.1093/ofid/ofw278 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 327.Macatangay BJC, Jackson EK, Abebe KZ, Comer D, Cyktor J, Klamar-Blain C, Borowski L, Gillespie DG, Mellors JW, Rinaldo CR, et al. A randomized, placebo-controlled, pilot clinical trial of dipyridamole to decrease human immunodeficiency virus-associated chronic inflammation. J Infect Dis. 2020;221:1598–1606. doi: 10.1093/infdis/jiz344 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 328.Torres B, Guardo AC, Squarcia M, Diaz A, Fabra A, Caballero M, Ugarte A, Leal L, Gatell JM, Plana M, et al. Impact of switching to raltegravir and/or adding losartan in lymphoid tissue fibrosis and inflammation in people living with HIV. A randomized clinical trial. HIV Med. 2021;22:674–681. doi: 10.1111/hiv.13114 [DOI] [PubMed] [Google Scholar]
- 329.Utay NS, Kitch DW, Yeh E, Fichtenbaum CJ, Lederman MM, Estes JD, Deleage C, Magyar C, Nelson SD, Klingman KL, et al. ; A5317 AIDS Clinical Trials Group Team. Telmisartan therapy does not improve lymph node or adipose tissue fibrosis more than continued antiretroviral therapy alone. J Infect Dis. 2018;217:1770–1781. doi: 10.1093/infdis/jiy064 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330.Baker JV, Huppler Hullsiek K, Prosser R, Duprez D, Grimm R, Tracy RP, Rhame F, Henry K, Neaton JD. Angiotensin converting enzyme inhibitor and HMG-CoA reductase inhibitor as adjunct treatment for persons with HIV infection: a feasibility randomized trial. PLoS One. 2012;7:e46894. doi: 10.1371/journal.pone.0046894 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331.Marconi VC, Moser C, Gavegnano C, Deeks SG, Lederman MM, Overton ET, Tsibris A, Hunt PW, Kantor A, Sekaly RP, et al. Randomized trial of ruxolitinib in antiretroviral-treated adults with human immunodeficiency virus. Clin Infect Dis. 2022;74:95–104. doi: 10.1093/cid/ciab212 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 332.Kelly KM, Tocchetti CG, Lyashkov A, Tarwater PM, Bedja D, Graham DR, Beck SE, Metcalf Pate KA, Queen SE, Adams RJ, et al. CCR5 inhibition prevents cardiac dysfunction in the SIV/macaque model of HIV. J Am Heart Assoc. 2014;3:e000874. doi: 10.1161/JAHA.114.000874 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 333.Bowman E, Cichon M, Reidl K, et al. Tocilizumab alters lipids in HIV+ individuals in a randomized, double-blind study. https://www.croiconference.org/abstract/tocilizumab-alters-lipids-hiv-individuals-randomized-double-blind-study/ [Google Scholar]
- 334.Funderburg NT, Shive CL, Chen Z, Tatsuoka C, Bowman ER, Longenecker CT, McComsey GA, Clagett BM, Dorazio D, Freeman ML, et al. Interleukin 6 blockade with tocilizumab diminishes indices of inflammation that are linked to mortality in treated human immunodeficiency virus infection. Clin Infect Dis. 2023;77:272–279. doi: 10.1093/cid/ciad199 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335.Hsue PY, Li D, Ma Y, Ishai A, Manion M, Nahrendorf M, Ganz P, Ridker PM, Deeks SG, Tawakol A. IL-1β inhibition reduces atherosclerotic inflammation in HIV infection. J Am Coll Cardiol. 2018;72:2809–2811. doi: 10.1016/j.jacc.2018.09.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336.d’Ettorre G, Ceccarelli G, Giustini N, Serafino S, Calantone N, De Girolamo G, Bianchi L, Bellelli V, Ascoli-Bartoli T, Marcellini S, et al. Probiotics reduce inflammation in antiretroviral treated, HIV-infected individuals: results of the “Probio-HIV” clinical trial. PLoS One. 2015;10:e0137200. doi: 10.1371/journal.pone.0137200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 337.Aberg JA, Sponseller CA, Ward DJ, Kryzhanovski VA, Campbell SE, Thompson MA. Pitavastatin versus pravastatin in adults with HIV-1 infection and dyslipidaemia (INTREPID): 12 week and 52 week results of a phase 4, multicentre, randomised, double-blind, superiority trial. Lancet HIV. 2017;4:e284–e294. doi: 10.1016/S2352-3018(17)30075-9 [DOI] [PubMed] [Google Scholar]
- 338.Toribio M, Fitch KV, Sanchez L, Burdo TH, Williams KC, Sponseller CA, McCurdy Pate M, Aberg JA, Zanni MV, Grinspoon SK. Effects of pitavastatin and pravastatin on markers of immune activation and arterial inflammation in HIV. AIDS. 2017;31:797–806. doi: 10.1097/QAD.0000000000001427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339.Funderburg NT, Jiang Y, Debanne SM, Labbato D, Juchnowski S, Ferrari B, Clagett B, Robinson J, Lederman MM, McComsey GA. Rosuvastatin reduces vascular inflammation and T-cell and monocyte activation in HIV-infected subjects on antiretroviral therapy. J Acquir Immune Defic Syndr. 2015;68:396–404. doi: 10.1097/QAI.0000000000000478 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 340.Eckard AR, Jiang Y, Debanne SM, Funderburg NT, McComsey GA. Effect of 24 weeks of statin therapy on systemic and vascular inflammation in HIV-infected subjects receiving antiretroviral therapy. J Infect Dis. 2014;209:1156–1164. doi: 10.1093/infdis/jiu012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 341.Funderburg NT, Jiang Y, Debanne SM, Storer N, Labbato D, Clagett B, Robinson J, Lederman MM, McComsey GA. Rosuvastatin treatment reduces markers of monocyte activation in HIV-infected subjects on antiretroviral therapy. Clin Infect Dis. 2014;58:588–595. doi: 10.1093/cid/cit748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 342.Nixon DE, Bosch RJ, Chan ES, Funderburg NT, Hodder S, Lake JE, Lederman MM, Klingman KL, Aberg JA; AIDS Clinical Trials Group Study A5275 Team. Effects of atorvastatin on biomarkers of immune activation, inflammation, and lipids in virologically suppressed, human immunodeficiency virus-1-infected individuals with low-density lipoprotein cholesterol <130 mg/dL (AIDS Clinical Trials Group Study A5275). J Clin Lipidol. 2017;11:61–69. doi: 10.1016/j.jacl.2016.09.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 343.Nou E, Lu MT, Looby SE, Fitch KV, Kim EA, Lee H, Hoffmann U, Grinspoon SK, Lo J. Serum oxidized low-density lipoprotein decreases in response to statin therapy and relates independently to reductions in coronary plaque in patients with HIV. AIDS. 2016;30:583–590. doi: 10.1097/QAD.0000000000000946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 344.Leucker TM, Gerstenblith G, Schär M, Brown TT, Jones SR, Afework Y, Weiss RG, Hays AG. Evolocumab, a PCSK9-monoclonal antibody, rapidly reverses coronary artery endothelial dysfunction in people living with HIV and people with dyslipidemia. J Am Heart Assoc. 2020;9:e016263. doi: 10.1161/JAHA.120.016263 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 345.Abbate A, Kontos MC, Abouzaki NA, Melchior RD, Thomas C, Van Tassell BW, Oddi C, Carbone S, Trankle CR, Roberts CS, et al. Comparative safety of interleukin-1 blockade with anakinra in patients with ST-segment elevation acute myocardial infarction (from the VCU-ART and VCU-ART2 pilot studies). Am J Cardiol. 2015;115:288–292. doi: 10.1016/j.amjcard.2014.11.003 [DOI] [PubMed] [Google Scholar]
- 346.Van Tassell BW, Arena R, Biondi-Zoccai G, Canada JM, Oddi C, Abouzaki NA, Jahangiri A, Falcao RA, Kontos MC, Shah KB, et al. Effects of interleukin-1 blockade with anakinra on aerobic exercise capacity in patients with heart failure and preserved ejection fraction (from the D-HART pilot study). Am J Cardiol. 2014;113:321–327. doi: 10.1016/j.amjcard.2013.08.047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 347.Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT; Anti-TNF Therapy Against Congestive Heart Failure Investigators. Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-alpha, in patients with moderate-to-severe heart failure: results of the anti-TNF Therapy Against Congestive Heart Failure (ATTACH) trial. Circulation. 2003;107:3133–3140. doi: 10.1161/01.CIR.0000077913.60364.D2 [DOI] [PubMed] [Google Scholar]
- 348.Mann DL, McMurray JJV, Packer M, Swedberg K, Borer JS, Colucci WS, Djian J, Drexler H, Feldman A, Kober L, et al. Targeted anticytokine therapy in patients with chronic heart failure: results of the Randomized Etanercept Worldwide Evaluation (RENEWAL). Circulation. 2004;109:1594–1602. doi: 10.1161/01.CIR.0000124490.27666.B2 [DOI] [PubMed] [Google Scholar]
- 349.Aoki T, Narumiya S. Prostaglandins and chronic inflammation. Trends Pharmacol Sci. 2012;33:304–311. doi: 10.1016/j.tips.2012.02.004 [DOI] [PubMed] [Google Scholar]
- 350.O’Brien M, Montenont E, Hu L, Nardi MA, Valdes V, Merolla M, Gettenberg G, Cavanagh K, Aberg JA, Bhardwaj N, et al. Aspirin attenuates platelet activation and immune activation in HIV-1-infected subjects on antiretroviral therapy: a pilot study. J Acquir Immune Defic Syndr. 2013;63:280–288. doi: 10.1097/QAI.0b013e31828a292c [DOI] [PMC free article] [PubMed] [Google Scholar]
- 351.Klein DB, Leyden WA, Xu L, Chao CR, Horberg MA, Towner WJ, Hurley LB, Marcus JL, Quesenberry CP, Silverberg MJ. Declining relative risk for myocardial infarction among HIV-positive compared with HIV-negative individuals with access to care. Clin Infect Dis. 2015;60:1278–1280. doi: 10.1093/cid/civ014 [DOI] [PubMed] [Google Scholar]
- 352.Metkus TS, Timpone J, Leaf D, Bidwell Goetz M, Harris WS, Brown TT. Omega-3 fatty acid therapy reduces triglycerides and interleukin-6 in hypertriglyeridemic HIV patients. HIV Med. 2013;14:530–539. doi: 10.1111/hiv.12046 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 353.Longenecker CT, Eckard AR, McComsey GA. Statins to improve cardiovascular outcomes in treated HIV infection. Curr Opin Infect Dis. 2016;29:1–9. doi: 10.1097/QCO.0000000000000223 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 354.Srichatrapimuk S, Wongsa A, Sungkanuparph S, Kiertiburanakul S, Tassaneetrithep B, Phuphuakrat A. Effects of pitavastatin on atherosclerotic-associated inflammatory biomarkers in people living with HIV with dyslipidemia and receiving ritonavir-boosted atazanavir: a randomized, double-blind, crossover study. AIDS Res Ther. 2023;20:13. doi: 10.1186/s12981-023-00506-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 355.Ferri N, Tibolla G, Pirillo A, Cipollone F, Mezzetti A, Pacia S, Corsini A, Catapano AL. Proprotein convertase subtilisin kexin type 9 (PCSK9) secreted by cultured smooth muscle cells reduces macrophages LDLR levels. Atherosclerosis. 2012;220:381–386. doi: 10.1016/j.atherosclerosis.2011.11.026 [DOI] [PubMed] [Google Scholar]
- 356.Leander K, Mälarstig A, Van’t Hooft FM, Hyde C, Hellénius ML, Troutt JS, Konrad RJ, Öhrvik J, Hamsten A, de Faire U. Circulating proprotein convertase subtilisin/kexin type 9 (PCSK9) predicts future risk of cardiovascular events independently of established risk factors. Circulation. 2016;133:1230–1239. doi: 10.1161/CIRCULATIONAHA.115.018531 [DOI] [PubMed] [Google Scholar]
- 357.Harb T, Ziogos E, Schär M, Brown TT, Lai S, Gerstenblith G, Hays AG, Leucker TM. Rapid improvement of coronary endothelial function with PCSK9 inhibition in people with HIV is associated with reduced lipoprotein (a) and not LDL-cholesterol. Circ Cardiovasc Imaging. 2023;16:e015693. doi: 10.1161/CIRCIMAGING.123.015693 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 358.Giugliano RP, Sabatine MS. Are PCSK9 inhibitors the next breakthrough in the cardiovascular field? J Am Coll Cardiol. 2015;65:2638–2651. doi: 10.1016/j.jacc.2015.05.001 [DOI] [PubMed] [Google Scholar]