Introduction
The development of atherosclerosis is a complex process involving endothelial dysfunction and arterial inflammation. Additionally, systemic inflammation likely exacerbates atherogenesis as several studies have shown a relationship between cardiovascular disease (CVD) and chronic inflammatory disorders such as systemic lupus erythematosus and rheumatoid arthritis [1, 2]. Chronic HIV is also considered a state of persistent inflammation that likely plays a role in accelerated biologic aging and development of conditions such as CVD. With increased use of combination antiretroviral therapy (cART), especially in resource-rich countries, CVD has in fact become one of the leading causes of morbidity and mortality in patients with HIV [3], and the proportionate CVD-related mortality rate may be rising over time despite declines seen in the general population and those with inflammatory polyarthropathies [4]. Moreover, several epidemiologic studies in HIV-infected patients have shown an increased risk of myocardial infarction and stroke that persists even when controlling for traditional CVD risk factors [5–11], suggesting contributory effects of non-traditional risk factors such as immune activation. As a result, investigators are now evaluating interventions that can reduce inflammation and lower CVD in people living with HIV. Additionally, more aggressive treatment with existing therapies such as lipid lowering medications and modern cART regimens may substantially reduce the risk of MI in HIV [12]
Vascular Immunobiology of Atherosclerosis
Although all three layers of the artery and several different cell types are involved in atherosclerotic plaque development, the actions of macrophages, T-cells, and smooth muscle cells at the tunica intima have best been studied to date. A simplified model is presented here and in Figure 1. Activation of the overlying endothelium is a critical first step in atherogenesis. Normally, the endothelium helps prevent thrombus formation and is a regulator of vascular permeability, inflammation, and tone. In response to proatherogenic stimuli, the endothelium alters production of nitric oxide (NO), which affects vascular tone, increases permeability, allowing entry of lipids, and expresses chemokines like monocyte chemoattractant protein-1 (MCP-1) and adhesion molecules like vascular cell adhesion molecule-1 (VCAM-1), which results in leukocyte transendothelial migration [13, 14]. These effects are observable as a thickening of the tunica intima associated with the presence of subendothelial pools and leukocytes in the vessel wall [15].
Figure 1.
Effects of HIV Viral Proteins on the Development of Atherosclerosis. In vitro studies have shown the following proatherogenic effects of HIV viral proteins: A) Tat and gp120 induces expression of adhesion molecules [179, 180]; B) Nef and gp120 reduce endothelial NO production [181, 182]; C) Nef increases inflammatory cytokine release including IL-6 and TNF-alpha from macrophages [183]; D) Nef promotes MCP-1 secretion from endothelial cells [184]; E) Tat stimulates MCP-1 mediated monocyte transmigration [185]; F) Nef inhibits ABCA1-dependent CEC of macrophages [76]; G) Nef and gp120 may induce endothelial apoptosis, which could promote fibroatheroma rupture/erosion, resulting in formation of an acute thrombus [186, 187]; H) Tat stimulates IL-6 production from peripheral blood monocytes [188]. In vitro studies have also shown potential proatherogenic effects of specific ARTs including increased leukocyte adhesion to endothelial cells with efavirenz [189]; increased platelet reactivity with abacavir [190]; reduced cholesterol efflux from macrophages, decreased endothelial nitric oxide production, and endothelial cytotoxicity with ritonavir [191–193]; and CD36-dependent cholesterol accumulation in macrophages with certain protease inhibitors [194]. ABCA1 = ATP-binding cassette transporter-A1, CEC = cholesterol efflux capacity, RCT = reverse cholesterol transport, TF = tissue factor, HDL = high-density lipoprotein, LDL = low-density lipoprotein, oxLDL = oxidized LDL, MCP-1 = monocyte chemoattractant protein-1.
Once in the arterial wall, scavenger receptors on macrophages recognize modified forms of low-density lipoprotein (LDL) such as oxidized LDL (oxLDL), resulting in differentiation to foam cells that are characterized by intracellular accumulation of lipids and production of pro-inflammatory cytokines and matrix metalloproteinases (MMP). Fragments of LDL are also likely presented to naive T-cells that can later enter the vessel wall and secrete pro-inflammatory molecules, leading to changes in macrophages, endothelial cells, and smooth muscle cells that promote further progression of the atherosclerotic plaque [16].
Eventually, the area of intimal thickening may progress into a fibroatheroma, characterized by the presence of a necrotic (or lipid) core. The necrotic core is largely defined by two features. The first involves the presence of dead or dying cells, largely macrophages, releasing intracellular lipids into the lipid core. The second features a dearth of extracellular matrix proteins, likely as a result of MMPs and other peptidases that contribute to structural instability of the arterial wall. Other notable components of fibroatheromas are intra-plaque calcifications and the overlying fibrous cap. The fibrous cap lends structural support to the growing plaque, but unfortunately, some fibrous caps are vulnerable to erosion or rupture, resulting in thrombus formation and potentially myocardial infarction, stroke, or sudden cardiac death [15].
Insights from Cardiac Imaging into the Unique Pathophysiology of Cardiovascular Disease in HIV
Although the relative risk of CVD is increased in patients with HIV, one limitation in studying CVD in this generally young patient population has been the relatively low absolute event rate in terms of CVD deaths [17]. As a result, several investigators have utilized cardiac imaging to document the burden of subclinical CVD, better understand the underlying pathophysiological mechanisms, and follow response to treatment interventions.
Carotid Ultrasound
One of the most widely utilized cardiac imaging modalities has been ultrasound of the carotid arteries, measuring various endpoints including carotid intima-media thickness (CIMT), presence of plaque, and progression or regression of CIMT. In the general population, CIMT has been associated with increased risk of MI and stroke [18, 19]. In patients with HIV, studies involving carotid ultrasound have often differed in terms of patient population, study design, and ultrasound technique, likely accounting for some of the conflicting results. The weight of evidence, however, suggests that CIMT and presence of plaque are greater in patients with HIV than uninfected controls, independent of traditional CVD risk factors [20, 21].
Cardiac Computed Tomography
Computed tomography has also become a useful tool in assessing subclinical CVD. Coronary artery calcium (CAC) scoring is an independent predictor of CVD risk in the general population [22, 23]. In a meta-analysis, however, CAC score was not significantly higher in patients with HIV compared with uninfected controls [20]. The CAC score though may underestimate atherosclerotic burden in patients with HIV, especially compared to other imaging modalities. Among patients with undetectable CAC, more patients with HIV have increased CIMT [24] and non-calcified plaque on cardiac computed tomography angiography (CCTA) [25] compared to patients without HIV. In addition to non-calcified plaque, CCTA can also detect high-risk morphologic features of coronary atherosclerosis such as plaque volume, low attenuation, positive remodeling, spotty calcification, and the napkin ring sign, which in the general population have been associated with culprit lesions in acute coronary syndrome [26]. In patients with HIV, several studies have reported an increased prevalence of non-calcified, low attenuation, and positively remodeled plaque compared with uninfected controls matched for traditional CVD risk factors [27–29]. Together, these data suggest that HIV infection is associated with accelerated development of atherosclerosis and possibly an altered plaque morphology that is more prone to rupture, findings that are congruent with the increased risk of MI and stroke reported in observational studies [30].
Functional Imaging with 18FDG PET-CT
Ultrasound and computed tomography, however, do not directly characterize the underlying biological process occurring within the atheroma. 18-Fluorodeoxyglucose (18FDG) coupled with positron emission tomography-computed tomography (PET-CT) is a molecular imaging modality that identifies sites of increased glucose metabolism such as malignancies and inflammatory processes. Given what is known about the dynamic inflammation involved in atherosclerosis, 18FDG PET-CT has also been utilized in research settings to study CVD and arterial inflammation at sites such as the carotid artery or aorta. In the general population, arterial inflammation on 18FDG PET-CT has been shown to independently predict cardiovascular events [31, 32]. In patients with HIV without known CVD, Subramanian and colleagues showed that aortic inflammation on 18FDG PET-CT was significantly increased compared to Framingham Risk Score-matched uninfected controls; that the degree of 18FDG uptake equaled those observed in uninfected subjects with known CVD; and that 18FDG PET-CT activity was associated with soluble CD163 (sCD163), a marker of monocyte/macrophage activation [33]. Additionally, higher levels of aortic 18FDG uptake have been associated with an increased prevalence of high-risk coronary plaque features on CCTA in patients with HIV [34]. Thus, these studies describe a significant relationship between systemic immune activation, arterial inflammation, and rupture-prone coronary lesions.
Chronic Inflammation and Endothelial Dysfunction in Patients with HIV
Chronic Inflammation in Untreated Patients with HIV
Untreated HIV infection leads to generalized activation of the immune system. Non-HIV specific T-cell activation has been observed and the frequency of activated CD8+ T-cells is greater than the frequency of HIV-specific T-cells [35, 36]. Moreover, this widespread stimulation of T-cells probably occurs through several different mechanisms, one of which likely includes activation of innate immunity. Studies have shown that sCD163 and inflammatory (CD14+CD16+) monocytes correlate with higher levels of CD8+ T-cell activation [37]. Therefore, immune activation in untreated HIV is pervasive, involving both HIV-specific and non-specific responses as well as both the adaptive and innate immune systems.
Chronic Inflammation in cART-Treated Patients with HIV
Viral suppression with cART alleviates but does not normalize immune activation in most patients with HIV. For example, cART reduces T-cell activation [38] (see Table 1), but levels remain elevated compared to uninfected controls [39, 40]. Additionally, other markers of inflammation and monocyte activation, including interleukin-6 (IL-6), high sensitivity C-reactive protein (hs-CRP), D-dimer, sCD163, and soluble CD14 (sCD14) decrease with cART [38, 41], but remain higher compared to HIV-uninfected individuals [37, 42–45]. It is important to note that studies differ regarding which markers decline and to what degree, with some even normalizing to levels seen in the general population. Although the reasons for this variable response are still under investigation, one potential factor is the timing of cART initiation. In some studies, earlier commencement is associated with greater improvements in markers of immune activation such as sCD163 levels [37], activated T-cells [46], IL-6, and hs-CRP [47]. Thus, treatment with cART at higher CD4+ T-cell counts may further reduce immune activation but whether this translates into reductions in CVD remains under investigation.
Table 1.
Atherosclerosis-related immune markers and associated treatment in HIV-infected individuals
Immune Marker | Subclinical Atherosclerosis | Cardiovascular Disease | Immune Marker Reducing Therapy |
---|---|---|---|
hs-CRP | CIMT progression[57] | MI and major CVD event[58, 59] | Probiotics[149], Lisinopril[171], Panobinostat[172], ART [38] |
IL-6 | MI, stroke, and major CVD event[59, 60] | Probiotics[149], Panobinostat[172], Fish Oil[166], Hydroxychloroquine [153], ART [38] | |
D-dimer | MI, stroke, and major CVD event[58–60] | Probiotics [150], rhIL-7 [146], ART [38] | |
LPS | CIMT progression[71] | Hydroxychloroquine [153], ART [41] | |
MCP-1 | CIMT, stenosis ≥ 50%, CAC, coronary segments with plaque[27, 43, 173] | ||
oxLDL | CIMT[173] | Rosuvastatin[160], Atorvastatin[161], Sevelamer[145] | |
sCD163 | CAC; vulnerable, total, non-calcified, mixed, and calcified plaque; coronary stenosis ≥ 50%[29, 43, 75] | Aprepitant[174], ART [38] | |
sCD14 | Non-calcified plaque, CIMT and coronary stenosis ≥ 50%[43, 71, 73, 75] | Highest quartile sCD14 w/CVD death,[72] MI, and stroke[60] | Rosuvastatin[157], rhIL-7 [146] Prebiotics [175], Aspirin [66], ART [41] |
CD14+CD16+ monocytes | CAC progression[70] | Panobinostat[172], ART [38] | |
CD4+CD38+HLADR+ T-cells | CIMT[80] | Probiotics[149], Rosuvastatin[157], Atorvastatin [176] Aspirin [66], ART [38] | |
CD8+CD38+HLADR+ T-cells | CIMT[79, 80] | Probiotics[149], Rosuvastatin[157], Atorvastatin [177], Chloroquine [152] Leflunomide [178], ART [38] |
hs-CRP = High sensitivity C-reactive protein, LPS = lipopolysaccharide, IL-6 = interleukin-6, MCP-1 = monocyte chemoattractant protein-1, oxLDL = oxidized low density lipoprotein, sCD163 = soluble CD163, sCD14 = soluble CD14, CIMT = carotid intima media thickness, CAC = coronary artery calcium, MI = myocardial infarction, CVD = cardiovascular disease, ART = antiretroviral therapy, rhIL-7 = recombinant human interleukin 7
Endothelial Dysfunction and Activation in Patients with HIV
Markers of endothelial cell activation and dysfunction are also altered in patients with HIV. Brachial artery reactivity testing to measure flow mediated dilation (FMD) is a measure of endothelial function and is a predictor of CVD in the general population [48]. In HIV, case-control studies have shown varying results regarding the effect of HIV on FMD, likely due to small sample sizes and differences in imaging techniques [49–51]. However, in a prospective, randomized trial, cART improved FMD [52], suggesting a relationship between viremia and endothelial dysfunction. Moreover, soluble VCAM-1 (sVCAM-1), a marker of endothelial activation, is higher in HIV-infected patients, declines with cART treatment, but remains elevated compared with HIV-uninfected patients [53, 54]. These studies also showed a relationship between sVCAM-1, inflammatory cytokines, and CIMT, supporting potential relationships between inflammation, endothelial activation, and CVD in HIV.
Immune Activation and CVD in Patients with HIV
Inflammation and Coagulation
Several biomarkers of immune activation have been related to both subclinical and clinical CVD in patients with HIV (see Table 1). Circulating markers of general inflammation such IL-6 and hs-CRP are well known independent predictors of CVD in the general population [55, 56]. In patients with HIV, hs-CRP has been associated with CIMT progression [57], and both hs-CRP and IL-6 increase the risk of CVD and mortality [58–62]. Given the known relationship between inflammation and coagulation, it is not surprising that D-dimer, a product of the coagulation cascade, is also increased in HIV [42] and has been strongly associated with mortality and CVD in patients with HIV [59–63]. It remains unclear, however, whether the relationship between coagulation and CVD is causal or simply a marker of heightened inflammation. Theoretically, a procoagulable state could contribute to the risk of CVD as myocardial infarction and stroke are acute thrombotic events. Furthermore, platelets appear to function abnormally in patients with HIV as studies have shown increased tendencies for platelet aggregation, activation, and expression of tissue factor (TF), a pro-coagulant molecule [64–66].
Monocytes/Macrophages
Given the central role of monocyte-derived macrophages in atherogenesis, it is not surprising that several lines of evidence suggest a relationship between macrophage activation and CVD in HIV. Monocytes are generally divided into three different subsets based on their expression of CD14 and CD16 as classical (CD14++CD16−), inflammatory (CD14++CD16+), and patrolling (CD14+CD16++). In the general population, the percentage of inflammatory monocytes independently predicts clinical CVD events [67], and patients with acute coronary syndrome have increased percentages of both inflammatory and patrolling monocytes along with TF expression [68]. Thus, some monocytes, especially those with reduced cellular CD14 expression and increased CD16 expression may be proatherogenic. Untreated HIV-infected patients also have increased percentages of inflammatory and patrolling monocytes with tissue factor expression, similar to uninfected patients suffering acute coronary syndrome [68]. This suggests that HIV infection may create an environment that promotes the development of proatherogenic monocytes, possibly as a direct result of HIV itself or potentially through indirect mechanisms such as increased presence of other microbial agents. Additionally, reduced cellular expression of CD14 has been associated with CIMT [69], and CD16 expression on monocytes appears to independently predict progression of coronary artery calcium [70]. Beyond cellular markers, a relationship between soluble markers of monocyte activation and CVD has also been established. Soluble CD14, a receptor for lipopolysaccharide (LPS) and a marker of macrophage activation, independently predicts all-cause mortality and has been associated with both subclinical and clinical CVD [43, 71–74]. Furthermore, sCD163 has been shown to be related to arterial inflammation on 18FDG PET-CT and coronary plaque on CCTA [29, 33, 43, 75].
In addition to activation, HIV infection may also lead to macrophage dysfunction in cholesterol handling, which could contribute to the development of atherosclerosis. Cholesterol can be moved from macrophages to high-density lipoprotein in a process known as reverse cholesterol transport, which is thought to protect against atherosclerosis. One of the initial steps involves macrophages and their cholesterol efflux capacity (CEC). In animal studies, HIV impairs ATP-binding cassette transporter A1 (ABCA1) dependent CEC in murine macrophages [76]. In human studies, patients with HIV have reduced ABCA1-dependent CEC compared to uninfected controls [77], and initiation of cART during acute HIV infection restores CEC [78].
T-cells
In contrast to monocytes/macrophages, a relationship between T-cell activation and CVD in patients with HIV is less certain. In cross sectional studies, the percentage of activated T-cells (based on CD38 and HLA-DR expression) was associated with prevalence of CIMT [79, 80] and carotid artery stiffness [81]. On the other hand, two case-control studies found no relationship between activated T-cells and clinical cardiovascular events [60, 63], and one cross-sectional study showed no correlation between T-cell activation and CIMT [82]. Thus, further research is needed to determine whether a relationship exists and if so, whether the relationship is causal or represents a marker for some other closely related process such as immunodeficiency, as low CD4+ T-cell counts have been linked to CVD along with nadir CD4+ T-cell counts in several [7, 28, 83–87] but not all studies [88].
Net Benefits of cART on Immune Activation and CVD
Early Initiation of cART
Current and nadir CD4+ T-cell counts are related to delayed initiation of cART, which in conjunction with observations that some CVD-related markers of immune activation such as sCD163 may normalize when cART is started during acute infection, suggests that earlier initiation of cART may improve CVD by reducing immune stimulation. This hypothesis was explored in the Strategic Timing of Antiretroviral Treatment (START) trial, which randomized patients with HIV to early initiation of cART at CD4+ T-cell counts > 500 cells/uL versus delayed initiation at CD4+ T-cell counts > 350 cells/uL [89]. The trial was stopped early after an average follow-up time of 3 years as an interim analysis showed a significant decrease in serious AIDS and non-AIDS related events and death in the early treatment group (HR 0·43 [95% CI 0·3–0·62]). CVD event rates, however, were not different between the two groups, possibly as a result of lower event rates than predicted due to the younger age of the population (median age was 36 years-old) and the early termination of the study.
Cardiometabolic Risk with cART
Large observational studies have shown that some anti-retroviral therapies are associated with an increased risk of MI [90–93] that may be related to metabolic side effects of cART such as dyslipidemia [91], glucose intolerance [94, 95], and lipodystrophy [96, 97]. Furthermore, based on in vitro studies, some antiretroviral medications may have direct proatherogenic effects on vascular endothelium, macrophages, and platelets [98]. The protease inhibitor, ritonavir, reduces cholesterol efflux from macrophages, decreases endothelial nitric oxide production, and is cytotoxic to endothelial cells. A number of protease inhibitors have also been shown to promote CD36-dependent cholesterol accumulation in macrophages. Additionally, efavirenz may promote leukocyte adhesion to endothelial cells, and abacavir could increase platelet reactivity. Further in vivo studies of these agents are important, as the proatherogenic properties of these specific medications have largely been established from in vitro studies and beneficial effects on viremia and inflammation may contribute to clinical benefits on CVD.
Newer antiretroviral therapies, on the other hand, may have fewer cardiometabolic complications [99]. Thus, some modern antiretroviral therapies may not be associated with an increased risk of MI [100]. This absence of CVD risk with newer antiretrovirals, however, may simply be a result of insufficient observational time for complications to have developed or aggressive treatment of traditional CVD risk factors in the modern era.
Lessons from Treatment Interruption and Elite Controllers
Clues regarding the relative influence of inflammation versus cART-related cardiometabolic toxicity on CVD in HIV come from treatment interruption trials and studies of elite controllers. The Strategic Management of Antiretroviral Therapy (SMART) trial randomized patients to a drug conservation group with intermittent cART to maintain CD4+ T-cell counts > 350 cells/uL or a viral suppression group with continuous cART. They showed an increase in mortality and CVD [101] in the drug conservation group, which was partially related to increased markers of inflammation [59, 61]. Thus, cART appears to have a net benefit in reducing CVD as any potential cardiometabolic toxicity appears to be outweighed by decreased immune activation and viral suppression.
Studies with elite controllers, a rare subset of patients with HIV who maintain undetectable viral loads without cART, provide additional support to the greater role of immune activation over cART toxicity in the development of atherosclerosis in virally suppressed HIV-infected patients. In cross-sectional studies, elite controllers have been found to have an increased prevalence of CIMT on carotid ultrasound and coronary plaque on CCTA compared with uninfected controls in conjunction with elevated levels of C-reactive protein, sCD14, and sCD163 [102, 103]. Thus, these patients with HIV without exposure to cART also have accelerated atherosclerosis, which may be a result of heightened immune activation.
Causes of Chronic Inflammation in Patients with HIV
Several reasons likely contribute to ongoing immune activation in cART-treated HIV-infected patients (see Figure 2). Although our understanding is still incomplete, possible explanations include microbial translocation, co-infections, and continued presence of HIV RNA at low levels below the detection of clinical assays.
Figure 2.
Pathways Involved in the Development of Immune Activation and Atherosclerosis in HIV. Arrows indicate a contributory effect. Terminal lines indicate an inhibitory effect. Dotted Lines indicate a potential yet uncertain relationship. cART = combined antiretroviral therapy, RCT = reverse cholesterol transport, CEC = cholesterol efflux capacity, GI = gastrointestinal, LDL = low-density lipoprotein.
Microbial Translocation
GI tract structural integrity is influenced by the underlying mucosal immune system and likely also the microbiota in the gut itself. One of the key events of early HIV infection is mucosal inflammation and a massive depletion of CD4+ T-cells in the intestinal lymphoid tissue, which is not fully reversed with cART [104–107], resulting in alterations to expression of genes related to GI barrier function [108]. Furthermore, the gut microbiome is altered in patients with HIV, and these alterations have been linked to mucosal and systemic inflammation [109].
Studies have demonstrated an increase in epithelial apoptosis in the small intestine and decreased expression of tight junction proteins in the large intestine of HIV-infected individuals [110, 111], likely resulting in increased GI permeability [110]. Furthermore, animal studies with pathologic SIV infection and in vitro studies with HIV have shown that the structural breakdown of the GI tight epithelial barrier leads to in situ translocation of microbes from the GI lumen into the body [112, 113]. Additionally, microbial byproducts can be detected in the circulation. One study showed that patients with HIV have elevated levels of LPS, a component of gram-negative bacteria. Initiation of cART resulted in a decline in LPS concentrations that remained elevated compared to uninfected controls [114]. Once microbial products have entered the host, they can be recognized by the immune system, resulting in immune activation, which in turn could result in CVD. Plasma levels of LPS and bacterial 16S rDNA, for example, have been related to markers of inflammation, monocyte activation, and T-cell activation [114–116]. Furthermore, LPS levels in cART-treated patients with HIV have also been directly linked to progression of CIMT [71].
Co-Infections
Patients with HIV are often co-infected with various microbes, especially other chronic viral infections. This additional infectious burden may lead to increased immune activation and thus CVD. In HIV, Hepatitis C (HCV) co-infection, for example, results in higher levels of sCD163 [53, 117], activated T-cells [118] and increased risk of CVD [7, 119, 120], although it is unknown whether HCV directly contributes to CVD risk or is a surrogate marker for another potential risk factor such as intravenous drug use. Some evidence suggests that it may be more than a surrogate marker as suppression of HCV with PEGylated alpha interferon and ribavirin reduces T-cell activation and markers of vascular inflammation in co-infected individuals [121, 122]. However, no studies to date have shown that treatment of HCV results in improvements in subclinical or clinical CVD. Furthermore, newer, more effective agents to treat HCV have been developed and investigating their impact on reducing inflammation and CVD will be an important area for future research.
Cytomegalovirus (CMV) is highly prevalent (75–90%) in patients with HIV [123] and for unknown reasons, the percentage of CMV-specific CD8+ T-cells in cART-treated patients may be twice that of HIV-uninfected controls [124], suggesting an increased immune response associated with CMV co-infection. In the general population, CMV has been associated with both subclinical and clinical CVD [125, 126]. In HIV, CMV antibody levels are associated with increased IL-6 and sCD14 levels [127], sCD163 [75], coronary plaque burden on CCTA [27], and prevalence of carotid artery lesions [128]. Additionally, higher CMV-specific T-cell responses in co-infected patients have been associated with increased CIMT in some [129] but not all studies [82]. In a large, prospective observational study of patients with HIV, CMV seropositivity was associated with non-AIDS morbidity and mortality including CVD [123]. Moreover, short-term treatment of CMV with valgancyclovir [130] in patients with HIV reduced CD8+ T-cell activation, but whether treatment could be sustained long enough to result in improvements in atherosclerotic disease is uncertain.
HIV Viremia
Several in vitro studies have shown potential mechanistic links between HIV viral proteins and processes of immune activation and atherogenesis (see Figure 1) [98]. The viral protein, Nef, for example, has been shown to reduce endothelial NO production, promote secretion of endothelial-cell derived MCP-1, induce endothelial cell apoptosis, increase inflammatory cytokine release from macrophages, and inhibit macrophage cholesterol efflux capacity. It remains uncertain, however, whether these in vitro observations occur in vivo.
In addition, clinical data has supported an association between viremia and CVD. In an observational study, HIV replication has been associated with myocardial infarction [87], and reduction of viremia with initiation of cART improves vascular function [52]. Furthermore, in the SMART study, continuous cART resulted in less cardiovascular events compared with intermittent or delayed therapy [101, 131]. Together, these data link viremia with atherosclerosis and highlight the importance of viral suppression with modern cART regimens to reduce CVD.
Antiretroviral therapy, however, cannot eradicate HIV from the body. Even when patients have undetectable viral loads based on current clinical assays, ultrasensitive methods can still detect HIV RNA in the plasma [132, 133]. Although it is theoretically possible that residual viremia stimulates the immune system and contributes to CVD, these relationships have yet to be proven. Furthermore, determining the source of residual viremia remains under investigation but could lead to additional therapeutic strategies for CVD and chronic inflammation. Residual viremia may be the result of low levels of ongoing replication in active HIV reservoirs or may represent release of non-productive virus from latent reservoirs. If active replication is occurring and contributing to persistent inflammation, intensification therapy with additional antiretroviral medications in theory should further decrease viral loads and immune activation. Studies to date, however, have shown no significant reductions in viremia and most, but not all, studies have been unable to detect a decline in markers of inflammation [134–139]. One potential explanation is that some antiretroviral therapies may not achieve therapeutic levels at all sites with active replication [140]. Thus, intensification therapy may be ineffective in suppressing the active viral reservoir, or active replication may not be a major contributor to residual viremia and inflammation.
Novel Use of Interventions to Reduce Immune Activation and CVD in HIV
The mounting evidence connecting increased risk of CVD and immune activation suggests that anti-inflammatory drugs may provide benefit in patients with HIV (see Table 1). Although some medications such as pentoxifylline [141] and salsalate [142, 143] have had disappointing results in reducing immune activation markers and/or subclinical CVD endpoints, other therapies are currently being investigated. A pilot study using methotrexate in cART-treated patients with HIV is underway and will assess safety and efficacy on inflammation and endothelial function (NCT0194911). Furthermore, biologic agents such as the IL-6 receptor antagonist tocilizumab (NCT02049437) and the IL-1 antagonist canakinumab (NCT02272946) are also being investigated for effects on endothelial dysfunction and arterial inflammation.
Targeting Microbial Translocation and Intestinal Inflammation
In addition to anti-inflammatory medications, investigators are also targeting the underlying processes that promote chronic immune activation. Trials involving intensification therapy for residual viremia and therapies for HCV and CMV have been discussed above. Interventions aimed at reducing microbial translocation have taken several different approaches that have been met with varying results. Mesalamine, an agent used for intestinal inflammation in ulcerative colitis, failed to reduce circulating markers of inflammation as well as T-cell activation in intestinal tissue and the periphery [144]. Sevelamer, a phosphate binding agent that also has a high affinity for LPS, did not decrease markers of immune activation and microbial translocation [145]. On the other hand, investigational IL-7 administration in humans [146] and IL-21 administration in SIV-infected rhesus macaques [147], given in addition to cART, showed improvements in intestinal immunity and peripheral markers of immune activation. Probiotics, which may alter the GI microbiota, have been shown to decrease markers of microbial translocation, inflammation, coagulation, and T-cell activation [148–150] with some markers being reduced to levels seen in HIV-uninfected patients. These studies were small and effects of probiotics on individual markers differed between studies. Together, however, these data suggest that altering the microbiome may reduce chronic inflammation but whether this translates into cardiovascular benefit remains undetermined. Quinolines such as chloroquine and hydroxychloroquine have also been tested in patients with HIV with inconclusive results regarding effects on microbial translocation and inflammation [151–155], possibly due to differences in the dose of medication used or whether patients were treated with cART. Recent studies with these two medications are either ongoing or have yet to publish final results (NCT01232660 and NCT00819390). Finally, teduglutide—a glucagon-like-peptide-2 analog thought to promote intestinal epithelial function—is currently being studied in a randomized, controlled trial for effects on microbial translocation, immune activation, arterial inflammation, and coronary plaque on CCTA (NCT02431325).
Traditional Cardiometabolic Therapies to Reduce Immune Activation in HIV
One interesting study has shown that currently available therapies may reduce the risk of CVD substantially. In a large, retrospective study of patients with HIV, Klein and colleagues reported a declining risk of MI over time in conjunction with increased prescriptions for lipid lowering and anti-hypertensive treatment and better control of HIV with cART. Importantly, the risk of MI was equivalent between HIV-infected and uninfected individuals in the most recent year of the study (2010–2011), during which time HIV-infected patients had a lower Framingham Risk score [12]. Thus, although these findings require further validation, they suggest that current cardiometabolic therapies could potentially have a profound impact in ameliorating CVD, especially those that have additional effects on inflammation.
Effects of Statins on Inflammation and CVD in HIV
Statins are known for the ability to improve lipids and prevent CVD [156]. They are also believed to possess anti-inflammatory effects. In patients with HIV, statins reduce several markers of immune activation and improve subclinical CVD. In the SATURN-HIV study, rosuvastatin was associated with decreases in sCD14, percent of non-classical monocytes with tissue factor expression, and percent of activated T-cells [157]. The rosuvastatin group also had a slower progression of CIMT and the greatest benefit was seen in those with higher levels of inflammatory markers at baseline [158]. In another randomized, placebo-controlled trial, atorvastatin resulted in reductions in coronary plaque volume and number of lesions with high risk morphology on CCTA [159]. The improvements in atherosclerosis observed in these studies may be partially related to reductions in plasma oxLDL [160, 161], and one in vitro study showed that infusion of oxLDL increases macrophage activation and promotes development of proatherogenic monocytes [162]. Thus, oxLDL may represent one mechanism through which statins exert their anti-inflammatory effects.
In general, randomized clinical trials of statins in patients with HIV have been limited to small sample sizes or subclinical cardiovascular endpoints. As a result, a large, multi-center, randomized, placebo-controlled trialed named REPRIEVE has been initiated to determine potential benefits of statin therapy in preventing CVD in HIV-infected individuals. This large 6,500 person trial will assess effects on major adverse cardiovascular events (MACE) and will include a mechanistic study to also assess effects on plaque and immune function (NCT02344290).
Effects of Other Cardiometabolic Therapies on Inflammation and CVD in HIV
There are several other medications regularly used in treatment of cardiometabolic diseases whose effects on immune activation have been investigated in patients with HIV. Other than statins, other medications for dyslipidemia such as fish oil have failed to reduce markers of inflammation in most [163–165] but not all studies[166], although the type and dose of fish oil used varied. Recently, extended release niacin and fenofibrate showed no effect on reducing hs-CRP, IL-6, or D-dimer [167]. In addition to treatment for dyslipidemia, treatments for platelet inhibition and hyperglycemia have been or are currently being studied. Although a short, one-week study showed that low-dose aspirin attenuates sCD14 and activated T-cells in cART-treated patients with HIV [66], a longer 12-week randomized controlled trial showed no effects of high or low-dose aspirin on monocyte activation (sCD14, sCD163), proatherogenic monocyte subsets, T-cell activation, and endothelial dysfunction [168]. Furthermore, metformin is being used in a 24-week pilot study to determine potential effects on immune activation and coronary lesions on CCTA (NCT02383563).
Relationship between the Renin-Angiotensin-Aldosterone System, Inflammation and Cardiometabolic Disease in HIV
Additionally, the renin-angiotensin-aldosterone system (RAAS) may also be related to inflammation and cardiometabolic disease in patients with HIV. Aldosterone concentrations were found to be higher in HIV-infected patients compared to uninfected individuals in a RAAS activated state that was achieved by a low-sodium diet. Aldosterone concentrations were also associated with visceral adiposity and insulin resistance [169]. The relationship between RAAS activation in HIV may in part be related to RAAS activation in adipocytes as a result of antiretroviral therapy, particularly ritonavir-boosted PIs [170]. Furthermore, in those with HIV, RAAS activation in those on a low sodium diet resulted in elevated hs-CRP and IL-6 levels [169], implicating a relationship between the RAAS system and inflammation in HIV-infected patients. Moreover, the angiotensin converting enzyme (ACE) inhibitor, lisinopril, reduces levels of hs-CRP and TNF-alpha [171]. Currently, several studies are evaluating the effects of blocking the RAAS system with eplerenone, telmisartan, and losartan on inflammation, visceral adipose tissue, insulin resistance, flow mediated dilation, and progression of CIMT (NCT01928927, NCT01529749, NCT02049307, NCT0185294, NCT01405456).
Conclusion
Chronic HIV infection results in a state of persistent inflammation and immune activation, which in turn promotes CVD, a significant contributor to morbidity and mortality. Combined antiretroviral therapy reduces but does not normalize immune activation, which may provide cardioprotection. The use of cART, however, is evolving and whether changes such as earlier initiation lead to cardiovascular benefit remains to be seen. The reason for chronic inflammation, even with cART treatment, is likely multifactorial, including microbial translocation, co-infections, and possibly ongoing low-levels of viremia. Currently available cardiometabolic medications such as statins can have a profound impact on CVD, partly as a result of beneficial reductions in immune activation, but their safety and efficacy in this patient population requires validation. Furthermore, investigators are also studying novel therapies that reduce inflammation directly by dampening the immune system or indirectly by targeting underlying causes that may drive chronic inflammation. CVD is likely to worsen as this patient population ages, and thus, understanding the various risk factors for CVD in this unique cohort and developing effective therapeutic strategies are paramount in reducing morbidity and mortality for patients living with HIV.
Acknowledgments
EN did the initial literature review and wrote the first draft of the manuscript. JL and SKG assisted with additional literature review and revised the manuscript. The authors would like to thank Arch MacInnes for his help in creating Figure 1.
Funding: This work was supported by the National Institutes of Health [5T32DK007028-40 to E.N., R01HL123351 to J.L, and U01HL123336 and P30 DK040561 to S.K.G.]. Funding sources had no role in the writing of the manuscript.
Footnotes
Conflicts of Interest: S.K.G has consulted with Navidea, AstraZeneca, NovoNordisk, Theratechnologies, Bristol Myers Squibb, Merck, and Gilead, and received grant support from Gilead, Amgen, KOWA Pharmaceuticals, Navidea, and Theratechnologies, unrelated to this manuscript. J.L. has consulted with Gilead, unrelated to this manuscript. E.N. declares no competing interests.
References
- 1.Roman MJ, Shanker BA, Davis A, Lockshin MD, Sammaritano L, Simantov R, et al. Prevalence and correlates of accelerated atherosclerosis in systemic lupus erythematosus. N Engl J Med. 2003;349:2399–2406. doi: 10.1056/NEJMoa035471. [DOI] [PubMed] [Google Scholar]
- 2.Avina-Zubieta JA, Choi HK, Sadatsafavi M, Etminan M, Esdaile JM, Lacaille D. Risk of cardiovascular mortality in patients with rheumatoid arthritis: a meta-analysis of observational studies. Arthritis Rheum. 2008;59:1690–1697. doi: 10.1002/art.24092. [DOI] [PubMed] [Google Scholar]
- 3.Miller CJ, Baker JV, Bormann AM, Erlandson KM, Huppler Hullsiek K, Justice AC, et al. Adjudicated Morbidity and Mortality Outcomes by Age among Individuals with HIV Infection on Suppressive Antiretroviral Therapy. PLoS One. 2014;9:e95061. doi: 10.1371/journal.pone.0095061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Feinstein MJ, Bahiru E, Achenbach C, Longenecker CT, Hsue P, So-Armah K, et al. Patterns of Cardiovascular Mortality for HIV-Infected Adults in the United States: 1999 to 2013. Am J Cardiol. 2016;117:214–220. doi: 10.1016/j.amjcard.2015.10.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Currier JS, Taylor A, Boyd F, Dezii CM, Kawabata H, Burtcel B, et al. 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]
- 6.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]
- 7.Freiberg MS, Chang CC, Kuller LH, Skanderson M, Lowy E, Kraemer KL, 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]
- 8.Paisible AL, Chang CC, So-Armah KA, Butt AA, Leaf DA, Budoff M, et al. HIV infection, cardiovascular disease risk factor profile, and risk for acute myocardial infarction. J Acquir Immune Defic Syndr. 2015;68:209–216. doi: 10.1097/QAI.0000000000000419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chow FC, Regan S, Feske S, Meigs JB, Grinspoon SK, Triant VA. Comparison of ischemic stroke incidence in HIV-infected and non-HIV-infected patients in a US health care system. J Acquir Immune Defic Syndr. 2012;60:351–358. doi: 10.1097/QAI.0b013e31825c7f24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Marcus JL, Leyden WA, Chao CR, Chow FC, Horberg MA, Hurley LB, et al. HIV infection and incidence of ischemic stroke. AIDS. 2014;28:1911–1919. doi: 10.1097/QAD.0000000000000352. [DOI] [PubMed] [Google Scholar]
- 11.Sico JJ, Chang CC, So-Armah K, Justice AC, Hylek E, Skanderson M, et al. HIV status and the risk of ischemic stroke among men. Neurology. 2015;84:1933–1940. doi: 10.1212/WNL.0000000000001560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Klein DB, Leyden WA, Xu L, Chao CR, Horberg MA, Towner WJ, et al. 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]
- 13.Chistiakov DA, Orekhov AN, Bobryshev YV. Endothelial Barrier and Its Abnormalities in Cardiovascular Disease. Front Physiol. 2015;6:365. doi: 10.3389/fphys.2015.00365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lo J, Plutzky J. The biology of atherosclerosis: general paradigms and distinct pathogenic mechanisms among HIV-infected patients. J Infect Dis. 2012;205(Suppl 3):S368–374. doi: 10.1093/infdis/jis201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Yahagi K, Kolodgie FD, Otsuka F, Finn AV, Davis HR, Joner M, et al. Pathophysiology of native coronary, vein graft, and in-stent atherosclerosis. Nat Rev Cardiol. 2016;13:79–98. doi: 10.1038/nrcardio.2015.164. [DOI] [PubMed] [Google Scholar]
- 16.Hansson GK, Libby P, Tabas I. Inflammation and plaque vulnerability. J Intern Med. 2015;278:483–493. doi: 10.1111/joim.12406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ingle SM, May MT, Gill MJ, Mugavero MJ, Lewden C, Abgrall S, et al. Impact of risk factors for specific causes of death in the first and subsequent years of antiretroviral therapy among HIV-infected patients. Clin Infect Dis. 2014;59:287–297. doi: 10.1093/cid/ciu261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Den Ruijter HM, Peters SA, Anderson TJ, Britton AR, Dekker JM, Eijkemans MJ, et al. Common carotid intima-media thickness measurements in cardiovascular risk prediction: a meta-analysis. JAMA. 2012;308:796–803. doi: 10.1001/jama.2012.9630. [DOI] [PubMed] [Google Scholar]
- 19.Stein JH, Korcarz CE, Hurst RT, Lonn E, Kendall CB, Mohler ER, et al. Use of carotid ultrasound to identify subclinical vascular disease and evaluate cardiovascular disease risk: a consensus statement from the American Society of Echocardiography Carotid Intima-Media Thickness Task Force. Endorsed by the Society for Vascular Medicine. J Am Soc Echocardiogr. 2008;21:93–111. doi: 10.1016/j.echo.2007.11.011. quiz 189–190. [DOI] [PubMed] [Google Scholar]
- 20.Hulten E, Mitchell J, Scally J, Gibbs B, Villines TC. HIV positivity, protease inhibitor exposure and subclinical atherosclerosis: a systematic review and meta-analysis of observational studies. Heart. 2009;95:1826–1835. doi: 10.1136/hrt.2009.177774. [DOI] [PubMed] [Google Scholar]
- 21.Grunfeld C, Delaney JA, Wanke C, Currier JS, Scherzer R, Biggs ML, et al. Preclinical atherosclerosis due to HIV infection: carotid intima-medial thickness measurements from the FRAM study. AIDS. 2009;23:1841–1849. doi: 10.1097/QAD.0b013e32832d3b85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Detrano R, Guerci AD, Carr JJ, Bild DE, Burke G, Folsom AR, et al. Coronary calcium as a predictor of coronary events in four racial or ethnic groups. N Engl J Med. 2008;358:1336–1345. doi: 10.1056/NEJMoa072100. [DOI] [PubMed] [Google Scholar]
- 23.Goff DC, Jr, Lloyd-Jones DM, Bennett G, Coady S, D’Agostino RB, Gibbons R, et al. 2013 ACC/AHA guideline on the assessment of cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation. 2014;129:S49–73. doi: 10.1161/01.cir.0000437741.48606.98. [DOI] [PubMed] [Google Scholar]
- 24.Hsue PY, Ordovas K, Lee T, Reddy G, Gotway M, Schnell A, et al. Carotid intima-media thickness among human immunodeficiency virus-infected patients without coronary calcium. Am J Cardiol. 2012;109:742–747. doi: 10.1016/j.amjcard.2011.10.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Metkus TS, Brown T, Budoff M, Kingsley L, Palella FJ, Jr, Witt MD, et al. HIV infection is associated with an increased prevalence of coronary noncalcified plaque among participants with a coronary artery calcium score of zero: Multicenter AIDS Cohort Study (MACS) HIV Med. 2015;16:635–639. doi: 10.1111/hiv.12262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Maurovich-Horvat P, Ferencik M, Voros S, Merkely B, Hoffmann U. Comprehensive plaque assessment by coronary CT angiography. Nat Rev Cardiol. 2014;11:390–402. doi: 10.1038/nrcardio.2014.60. [DOI] [PubMed] [Google Scholar]
- 27.Lo J, Abbara S, Shturman L, Soni A, Wei J, Rocha-Filho JA, et al. Increased prevalence of subclinical coronary atherosclerosis detected by coronary computed tomography angiography in HIV-infected men. AIDS. 2010;24:243–253. doi: 10.1097/QAD.0b013e328333ea9e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Post WS, Budoff M, Kingsley L, Palella FJ, Jr, Witt MD, Li X, et al. 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]
- 29.Zanni MV, Abbara S, Lo J, Wai B, Hark D, Marmarelis E, et al. Increased coronary atherosclerotic plaque vulnerability by coronary computed tomography angiography in HIV-infected men. AIDS. 2013;27:1263–1272. doi: 10.1097/QAD.0b013e32835eca9b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Boccara F, Lang S, Meuleman C, Ederhy S, Mary-Krause M, Costagliola D, et al. HIV and coronary heart disease: time for a better understanding. J Am Coll Cardiol. 2013;61:511–523. doi: 10.1016/j.jacc.2012.06.063. [DOI] [PubMed] [Google Scholar]
- 31.Figueroa AL, Abdelbaky A, Truong QA, Corsini E, MacNabb MH, Lavender ZR, et al. Measurement of arterial activity on routine FDG PET/CT images improves prediction of risk of future CV events. JACC Cardiovasc Imaging. 2013;6:1250–1259. doi: 10.1016/j.jcmg.2013.08.006. [DOI] [PubMed] [Google Scholar]
- 32.Marnane M, Merwick A, Sheehan OC, Hannon N, Foran P, Grant T, et al. Carotid plaque inflammation on 18F-fluorodeoxyglucose positron emission tomography predicts early stroke recurrence. Ann Neurol. 2012;71:709–718. doi: 10.1002/ana.23553. [DOI] [PubMed] [Google Scholar]
- 33.Subramanian S, Tawakol A, Burdo TH, Abbara S, Wei J, Vijayakumar J, 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]
- 34.Tawakol A, Lo J, Zanni MV, Marmarelis E, Ihenachor EJ, MacNabb M, et al. Increased Arterial Inflammation Relates to High-Risk Coronary Plaque Morphology in HIV-Infected Patients. J Acquir Immune Defic Syndr. 2014;66:164–171. doi: 10.1097/QAI.0000000000000138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Doisne JM, Urrutia A, Lacabaratz-Porret C, Goujard C, Meyer L, Chaix ML, et al. CD8+ T cells specific for EBV, cytomegalovirus, and influenza virus are activated during primary HIV infection. J Immunol. 2004;173:2410–2418. doi: 10.4049/jimmunol.173.4.2410. [DOI] [PubMed] [Google Scholar]
- 36.Papagno L, Spina CA, Marchant A, Salio M, Rufer N, Little S, et al. Immune activation and CD8+ T-cell differentiation towards senescence in HIV-1 infection. PLoS Biol. 2004;2:E20. doi: 10.1371/journal.pbio.0020020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Burdo TH, Lentz MR, Autissier P, Krishnan A, Halpern E, Letendre S, et al. 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]
- 38.Kelesidis T, Tran TT, Stein JH, Brown TT, Moser C, Ribaudo HJ, et al. Changes in Inflammation and Immune Activation With Atazanavir-, Raltegravir-, Darunavir-Based Initial Antiviral Therapy: ACTG 5260s. Clin Infect Dis. 2015;61:651–660. doi: 10.1093/cid/civ327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Valdez H, Connick E, Smith KY, Lederman MM, Bosch RJ, Kim RS, et al. Limited immune restoration after 3 years’ suppression of HIV-1 replication in patients with moderately advanced disease. AIDS. 2002;16:1859–1866. doi: 10.1097/00002030-200209270-00002. [DOI] [PubMed] [Google Scholar]
- 40.Hunt PW, Martin JN, Sinclair E, Bredt B, Hagos E, Lampiris H, et al. 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]
- 41.Vesterbacka J, Nowak P, Barqasho B, Abdurahman S, Nystrom J, Nilsson S, et al. Kinetics of microbial translocation markers in patients on efavirenz or lopinavir/r based antiretroviral therapy. PLoS One. 2013;8:e55038. doi: 10.1371/journal.pone.0055038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Neuhaus J, Jacobs DR, Jr, Baker JV, Calmy A, Duprez D, La Rosa A, 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]
- 43.McKibben RA, Margolick JB, Grinspoon S, Li X, Palella FJ, Jr, Kingsley LA, 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]
- 44.Rudy BJ, Kapogiannis BG, Worrell C, Squires K, Bethel J, Li S, et al. Immune Reconstitution but Persistent Activation After 48 Weeks of Antiretroviral Therapy in Youth With Pre-Therapy CD4 >350 in ATN 061. J Acquir Immune Defic Syndr. 2015;69:52–60. doi: 10.1097/QAI.0000000000000549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hattab S, Guiguet M, Carcelain G, Fourati S, Guihot A, Autran B, et al. Soluble biomarkers of immune activation and inflammation in HIV infection: impact of 2 years of effective first-line combination antiretroviral therapy. HIV Med. 2015;16:553–562. doi: 10.1111/hiv.12257. [DOI] [PubMed] [Google Scholar]
- 46.Jain V, Hartogensis W, Bacchetti P, Hunt PW, Hatano H, Sinclair E, et al. Antiretroviral therapy initiated within 6 months of HIV infection is associated with lower T-cell activation and smaller HIV reservoir size. J Infect Dis. 2013;208:1202–1211. doi: 10.1093/infdis/jit311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ghislain M, Bastard JP, Meyer L, Capeau J, Fellahi S, Gerard L, et al. Late Antiretroviral Therapy (ART) Initiation Is Associated with Long-Term Persistence of Systemic Inflammation and Metabolic Abnormalities. PLoS One. 2015;10:e0144317. doi: 10.1371/journal.pone.0144317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Yeboah J, Crouse JR, Hsu FC, Burke GL, Herrington DM. Brachial flow-mediated dilation predicts incident cardiovascular events in older adults: the Cardiovascular Health Study. Circulation. 2007;115:2390–2397. doi: 10.1161/CIRCULATIONAHA.106.678276. [DOI] [PubMed] [Google Scholar]
- 49.van Wijk JP, de Koning EJ, Cabezas MC, Joven J, op’t Roodt J, Rabelink TJ, et al. Functional and structural markers of atherosclerosis in human immunodeficiency virus-infected patients. J Am Coll Cardiol. 2006;47:1117–1123. doi: 10.1016/j.jacc.2005.09.073. [DOI] [PubMed] [Google Scholar]
- 50.Solages A, Vita JA, Thornton DJ, Murray J, Heeren T, Craven DE, et al. Endothelial function in HIV-infected persons. Clin Infect Dis. 2006;42:1325–1332. doi: 10.1086/503261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Blanco JJ, Garcia IS, Cerezo JG, de Rivera JM, Anaya PM, Raya PG, et al. Endothelial function in HIV-infected patients with low or mild cardiovascular risk. J Antimicrob Chemother. 2006;58:133–139. doi: 10.1093/jac/dkl190. [DOI] [PubMed] [Google Scholar]
- 52.Torriani FJ, Komarow L, Parker RA, Cotter BR, Currier JS, Dube MP, et al. 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]
- 53.Beltran LM, Munoz Hernandez R, de Pablo Bernal RS, Garcia Morillo JS, Egido J, Noval ML, et al. Reduced sTWEAK and increased sCD163 levels in HIV-infected patients: modulation by antiretroviral treatment, HIV replication and HCV co-infection. PLoS One. 2014;9:e90541. doi: 10.1371/journal.pone.0090541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ross AC, Rizk N, O’Riordan MA, Dogra V, El-Bejjani D, Storer N, et al. 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]
- 55.Ridker PM, Rifai N, Rose L, Buring JE, Cook NR. Comparison of C-Reactive Protein and Low-Density Lipoprotein Cholesterol Levels in the Prediction of First Cardiovascular Events 10.1056/NEJMoa021993. N Engl J Med. 2002;347:1557–1565. doi: 10.1056/NEJMoa021993. [DOI] [PubMed] [Google Scholar]
- 56.Danesh J, Kaptoge S, Mann AG, Sarwar N, Wood A, Angleman SB, et al. Long-term interleukin-6 levels and subsequent risk of coronary heart disease: two new prospective studies and a systematic review. PLoS Med. 2008;5:e78. doi: 10.1371/journal.pmed.0050078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Hileman CO, Longenecker CT, Carman TL, McComsey GA. C-reactive protein predicts 96-week carotid intima media thickness progression in HIV-infected adults naive to antiretroviral therapy. J Acquir Immune Defic Syndr. 2014;65:340–344. doi: 10.1097/QAI.0000000000000063. [DOI] [PubMed] [Google Scholar]
- 58.Triant VA, Meigs JB, Grinspoon SK. Association of C-reactive protein and HIV infection with acute myocardial infarction. J Acquir Immune Defic Syndr. 2009;51:268–273. doi: 10.1097/QAI.0b013e3181a9992c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Duprez DA, Neuhaus J, Kuller LH, Tracy R, Belloso W, De Wit S, et al. 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]
- 60.Tenorio AR, Zheng Y, Bosch RJ, Krishnan S, Rodriguez B, Hunt PW, et al. Soluble markers of inflammation and coagulation but not T-cell activation predict non-AIDS-defining morbid events during suppressive antiretroviral treatment. J Infect Dis. 2014;210:1248–1259. doi: 10.1093/infdis/jiu254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kuller LH, Tracy R, Belloso W, De Wit S, Drummond F, Lane HC, et al. 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]
- 62.Vos AG, Idris NS, Barth RE, Klipstein-Grobusch K, Grobbee DE. Pro-Inflammatory Markers in Relation to Cardiovascular Disease in HIV Infection. A Systematic Review. PLoS One. 2016;11:e0147484. doi: 10.1371/journal.pone.0147484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Ford ES, Greenwald JH, Richterman AG, Rupert A, Dutcher L, Badralmaa Y, et al. Traditional risk factors and D-dimer predict incident cardiovascular disease events in chronic HIV infection. Aids. 2010;24:1509–1517. doi: 10.1097/QAD.0b013e32833ad914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mayne E, Funderburg NT, Sieg SF, Asaad R, Kalinowska M, Rodriguez B, et al. Increased platelet and microparticle activation in HIV infection: upregulation of P-selectin and tissue factor expression. J Acquir Immune Defic Syndr. 2012;59:340–346. doi: 10.1097/QAI.0b013e3182439355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Holme PA, Muller F, Solum NO, Brosstad F, Froland SS, Aukrust P. Enhanced activation of platelets with abnormal release of RANTES in human immunodeficiency virus type 1 infection. FASEB J. 1998;12:79–89. doi: 10.1096/fasebj.12.1.79. [DOI] [PubMed] [Google Scholar]
- 66.O’Brien M, Montenont E, Hu L, Nardi MA, Valdes V, Merolla M, 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]
- 67.Rogacev KS, Cremers B, Zawada AM, Seiler S, Binder N, Ege P, 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]
- 68.Funderburg NT, Zidar DA, Shive C, Lioi A, Mudd J, Musselwhite LW, et al. Shared monocyte subset phenotypes in HIV-1 infection and in uninfected subjects with acute coronary syndrome. Blood. 2012;120:4599–4608. doi: 10.1182/blood-2012-05-433946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Barbour JD, Jalbert EC, Chow DC, Gangcuangco LM, Norris PJ, Keating SM, 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]
- 70.Baker JV, Hullsiek KH, Singh A, Wilson E, Henry K, Lichtenstein K, et al. Immunologic predictors of coronary artery calcium progression in a contemporary HIV cohort. AIDS. 2014;28:831–840. doi: 10.1097/QAD.0000000000000145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.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]
- 72.Sandler NG, Wand H, Roque A, Law M, Nason MC, Nixon DE, et al. Plasma levels of soluble CD14 independently predict mortality in HIV infection. J Infect Dis. 2011;203:780–790. doi: 10.1093/infdis/jiq118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Merlini E, Luzi K, Suardi E, Barassi A, Cerrone M, Martinez JS, et al. 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]
- 74.Siedner MJ, Kim JH, Nakku RS, Bibangambah P, Hemphill L, Triant VA, et al. Persistent Immune Activation and Carotid Atherosclerosis in HIV-Infected Ugandans Receiving Antiretroviral Therapy. J Infect Dis. 2016;213:370–378. doi: 10.1093/infdis/jiv450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Burdo TH, Lo J, Abbara S, Wei J, DeLelys ME, Preffer F, et al. Soluble CD163, a novel marker of activated macrophages, is elevated and associated with noncalcified coronary plaque in HIV-infected patients. J Infect Dis. 2011;204:1227–1236. doi: 10.1093/infdis/jir520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Mujawar Z, Rose H, Morrow MP, Pushkarsky T, Dubrovsky L, Mukhamedova N, 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]
- 77.El Khoury P, Ghislain M, Villard EF, Le Goff W, Lascoux-Combe C, Yeni P, et al. Plasma cholesterol efflux capacity from human THP-1 macrophages is reduced in HIV-infected patients: impact of HAART. J Lipid Res. 2015;56:692–702. doi: 10.1194/jlr.M054510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Lo J, Rosenberg ES, Fitzgerald ML, Bazner SB, Ihenachor EJ, Hawxhurst V, et al. High-density lipoprotein-mediated cholesterol efflux capacity is improved by treatment with antiretroviral therapy in acute human immunodeficiency virus infection. Open Forum Infect Dis. 2014;1:ofu108. doi: 10.1093/ofid/ofu108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Longenecker CT, Funderburg NT, Jiang Y, Debanne S, Storer N, Labbato DE, et al. 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]
- 80.Kaplan RC, Sinclair E, Landay AL, Lurain N, Sharrett AR, Gange SJ, 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]
- 81.Karim R, Mack WJ, Kono N, Tien PC, Anastos K, Lazar J, 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]
- 82.Goulenok T, Boyd A, Larsen M, Fastenackels S, Boccara F, Meynard JL, et al. Increased carotid intima-media thickness is not associated with T-cell activation nor with cytomegalovirus in HIV-infected never-smoker patients. AIDS. 2015;29:287–293. doi: 10.1097/QAD.0000000000000539. [DOI] [PubMed] [Google Scholar]
- 83.Silverberg MJ, Leyden WA, Xu L, Horberg MA, Chao CR, Towner WJ, et al. Immunodeficiency and risk of myocardial infarction among HIV-positive individuals with access to care. J Acquir Immune Defic Syndr. 2014;65:160–166. doi: 10.1097/QAI.0000000000000009. [DOI] [PubMed] [Google Scholar]
- 84.Baker JV, Peng G, Rapkin J, Abrams DI, Silverberg MJ, MacArthur RD, et al. CD4+ count and risk of non-AIDS diseases following initial treatment for HIV infection. AIDS. 2008;22:841–848. doi: 10.1097/QAD.0b013e3282f7cb76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Lichtenstein KA, Armon C, Buchacz K, Chmiel JS, Buckner K, Tedaldi EM, et al. Low CD4+ T cell count is a risk factor for cardiovascular disease events in the HIV outpatient study. Clin Infect Dis. 2010;51:435–447. doi: 10.1086/655144. [DOI] [PubMed] [Google Scholar]
- 86.Triant VA, Regan S, Lee H, Sax PE, Meigs JB, Grinspoon SK. Association of immunologic and virologic factors with myocardial infarction rates in a US healthcare system. J Acquir Immune Defic Syndr. 2010;55:615–619. doi: 10.1097/QAI.0b013e3181f4b752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Lang S, Mary-Krause M, Simon A, Partisani M, Gilquin J, Cotte L, et al. HIV replication and immune status are independent predictors of the risk of myocardial infarction in HIV-infected individuals. Clin Infect Dis. 2012;55:600–607. doi: 10.1093/cid/cis489. [DOI] [PubMed] [Google Scholar]
- 88.Sabin CA, Ryom L, De Wit S, Mocroft A, Phillips AN, Worm SW, et al. Associations between immune depression and cardiovascular events in HIV infection. AIDS. 2013;27:2735–2748. doi: 10.1097/01.aids.0000432457.91228.f3. [DOI] [PubMed] [Google Scholar]
- 89.Lundgren JD, Babiker AG, Gordin F, Emery S, Grund B, Sharma S, et al. 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]
- 90.Friis-Moller N, Reiss P, Sabin CA, Weber R, Monforte A, El-Sadr W, et al. Class of antiretroviral drugs and the risk of myocardial infarction. N Engl J Med. 2007;356:1723–1735. doi: 10.1056/NEJMoa062744. [DOI] [PubMed] [Google Scholar]
- 91.Worm SW, Sabin C, Weber R, Reiss P, El-Sadr W, Dabis F, et al. Risk of myocardial infarction in patients with HIV infection exposed to specific individual antiretroviral drugs from the 3 major drug classes: the data collection on adverse events of anti-HIV drugs (D:A:D) study. J Infect Dis. 2010;201:318–330. doi: 10.1086/649897. [DOI] [PubMed] [Google Scholar]
- 92.Klein D, Hurley LB, Quesenberry CP, Jr, Sidney S. Do protease inhibitors increase the risk for coronary heart disease in patients with HIV-1 infection? J Acquir Immune Defic Syndr. 2002;30:471–477. doi: 10.1097/00126334-200208150-00002. [DOI] [PubMed] [Google Scholar]
- 93.Desai M, Joyce V, Bendavid E, Olshen RA, Hlatky M, Chow A, et al. Risk of Cardiovascular Events Associated With Current Exposure to HIV Antiretroviral Therapies in a US Veteran Population. Clin Infect Dis. 2015;61:445–452. doi: 10.1093/cid/civ316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Brown TT, Cole SR, Li X, Kingsley LA, Palella FJ, Riddler SA, et al. Antiretroviral therapy and the prevalence and incidence of diabetes mellitus in the multicenter AIDS cohort study. Arch Intern Med. 2005;165:1179–1184. doi: 10.1001/archinte.165.10.1179. [DOI] [PubMed] [Google Scholar]
- 95.Brown TT, Li X, Cole SR, Kingsley LA, Palella FJ, Riddler SA, et al. Cumulative exposure to nucleoside analogue reverse transcriptase inhibitors is associated with insulin resistance markers in the Multicenter AIDS Cohort Study. Aids. 2005;19:1375–1383. doi: 10.1097/01.aids.0000181011.62385.91. [DOI] [PubMed] [Google Scholar]
- 96.Hadigan C, Meigs JB, Corcoran C, Rietschel P, Piecuch S, Basgoz N, et al. Metabolic abnormalities and cardiovascular disease risk factors in adults with human immunodeficiency virus infection and lipodystrophy. Clin Infect Dis. 2001;32:130–139. doi: 10.1086/317541. [DOI] [PubMed] [Google Scholar]
- 97.Mallon PW, Miller J, Cooper DA, Carr A. Prospective evaluation of the effects of antiretroviral therapy on body composition in HIV-1-infected men starting therapy. AIDS. 2003;17:971–979. doi: 10.1097/00002030-200305020-00005. [DOI] [PubMed] [Google Scholar]
- 98.Zanni MV, Schouten J, Grinspoon SK, Reiss P. Risk of coronary heart disease in patients with HIV infection. Nat Rev Cardiol. 2014;11:728–741. doi: 10.1038/nrcardio.2014.167. [DOI] [PubMed] [Google Scholar]
- 99.Srinivasa S, Grinspoon SK. Metabolic and body composition effects of newer antiretrovirals in HIV-infected patients. Eur J Endocrinol. 2014;170:R185–202. doi: 10.1530/EJE-13-0967. [DOI] [PubMed] [Google Scholar]
- 100.Monforte A, Reiss P, Ryom L, El-Sadr W, Dabis F, De Wit S, et al. Atazanavir is not associated with an increased risk of cardio- or cerebrovascular disease events. AIDS. 2013;27:407–415. doi: 10.1097/QAD.0b013e32835b2ef1. [DOI] [PubMed] [Google Scholar]
- 101.El-Sadr WM, Lundgren JD, Neaton JD, Gordin F, Abrams D, Arduino RC, et al. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med. 2006;355:2283–2296. doi: 10.1056/NEJMoa062360. [DOI] [PubMed] [Google Scholar]
- 102.Hsue PY, Hunt PW, Schnell A, Kalapus SC, Hoh R, Ganz P, et al. Role of viral replication, antiretroviral therapy, and immunodeficiency in HIV-associated atherosclerosis. AIDS. 2009;23:1059–1067. doi: 10.1097/QAD.0b013e32832b514b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Pereyra F, Lo J, Triant VA, Wei J, Buzon MJ, Fitch KV, 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]
- 104.Gordon SN, Cervasi B, Odorizzi P, Silverman R, Aberra F, Ginsberg G, et al. Disruption of intestinal CD4+ T cell homeostasis is a key marker of systemic CD4+ T cell activation in HIV-infected individuals. J Immunol. 2010;185:5169–5179. doi: 10.4049/jimmunol.1001801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Guadalupe M, Reay E, Sankaran S, Prindiville T, Flamm J, McNeil A, et al. Severe CD4+ T-cell depletion in gut lymphoid tissue during primary human immunodeficiency virus type 1 infection and substantial delay in restoration following highly active antiretroviral therapy. J Virol. 2003;77:11708–11717. doi: 10.1128/JVI.77.21.11708-11717.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Mehandru S, Poles MA, Tenner-Racz K, Horowitz A, Hurley A, Hogan C, et al. Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J Exp Med. 2004;200:761–770. doi: 10.1084/jem.20041196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Brenchley JM, Schacker TW, Ruff LE, Price DA, Taylor JH, Beilman GJ, 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]
- 108.Sankaran S, George MD, Reay E, Guadalupe M, Flamm J, Prindiville T, et al. Rapid onset of intestinal epithelial barrier dysfunction in primary human immunodeficiency virus infection is driven by an imbalance between immune response and mucosal repair and regeneration. J Virol. 2008;82:538–545. doi: 10.1128/JVI.01449-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Dillon SM, Lee EJ, Kotter CV, Austin GL, Dong Z, Hecht DK, et al. An altered intestinal mucosal microbiome in HIV-1 infection is associated with mucosal and systemic immune activation and endotoxemia. Mucosal Immunol. 2014;7:983–994. doi: 10.1038/mi.2013.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Chung CY, Alden SL, Funderburg NT, Fu P, Levine AD. Progressive proximal-to-distal reduction in expression of the tight junction complex in colonic epithelium of virally-suppressed HIV+ individuals. PLoS Pathog. 2014;10:e1004198. doi: 10.1371/journal.ppat.1004198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Epple HJ, Schneider T, Troeger H, Kunkel D, Allers K, Moos V, et al. Impairment of the intestinal barrier is evident in untreated but absent in suppressively treated HIV-infected patients. Gut. 2009;58:220–227. doi: 10.1136/gut.2008.150425. [DOI] [PubMed] [Google Scholar]
- 112.Nazli A, Chan O, Dobson-Belaire WN, Ouellet M, Tremblay MJ, Gray-Owen SD, et al. Exposure to HIV-1 directly impairs mucosal epithelial barrier integrity allowing microbial translocation. PLoS Pathog. 2010;6:e1000852. doi: 10.1371/journal.ppat.1000852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Estes JD, Harris LD, Klatt NR, Tabb B, Pittaluga S, Paiardini M, et al. Damaged Intestinal Epithelial Integrity Linked to Microbial Translocation in Pathogenic Simian Immunodeficiency Virus Infections. PLoS Pathog. 2010;6:e1001052. doi: 10.1371/journal.ppat.1001052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12:1365–1371. doi: 10.1038/nm1511. [DOI] [PubMed] [Google Scholar]
- 115.Jiang W, Lederman MM, Hunt P, Sieg SF, Haley K, Rodriguez B, 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]
- 116.Cassol E, Malfeld S, Mahasha P, van der Merwe S, Cassol S, Seebregts C, et al. Persistent microbial translocation and immune activation in HIV-1-infected South Africans receiving combination antiretroviral therapy. J Infect Dis. 2010;202:723–733. doi: 10.1086/655229. [DOI] [PubMed] [Google Scholar]
- 117.Williams B, Livak B, Bahk M, Keating SM, Adeyemi OM. Short Communication: SCD14 and SCD163 Levels Are Correlated with VACS Index Scores: Initial Data from the Blunted Immune Recovery in CORE Patients with HIV (BIRCH) Cohort. AIDS Res Hum Retroviruses. 2016;32:144–147. doi: 10.1089/aid.2015.0012. [DOI] [PubMed] [Google Scholar]
- 118.Kovacs A, Al-Harthi L, Christensen S, Mack W, Cohen M, Landay A. CD8(+) T cell activation in women coinfected with human immunodeficiency virus type 1 and hepatitis C virus. J Infect Dis. 2008;197:1402–1407. doi: 10.1086/587696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.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]
- 120.Fernandez-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]
- 121.Gonzalez VD, Falconer K, Blom KG, Reichard O, Morn B, Laursen AL, et al. High levels of chronic immune activation in the T-cell compartments of patients coinfected with hepatitis C virus and human immunodeficiency virus type 1 and on highly active antiretroviral therapy are reverted by alpha interferon and ribavirin treatment. J Virol. 2009;83:11407–11411. doi: 10.1128/JVI.01211-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Masia M, Robledano C, Lopez N, Escolano C, Gutierrez F. Treatment for hepatitis C virus with pegylated interferon-alpha plus ribavirin induces anti-atherogenic effects on cardiovascular risk biomarkers in HIV-infected and -uninfected patients. J Antimicrob Chemother. 2011;66:1861–1868. doi: 10.1093/jac/dkr202. [DOI] [PubMed] [Google Scholar]
- 123.Lichtner M, Cicconi P, Vita S, Cozzi-Lepri A, Galli M, Lo Caputo S, et al. 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]
- 124.Naeger DM, Martin JN, Sinclair E, Hunt PW, Bangsberg DR, Hecht F, et al. Cytomegalovirus-specific T cells persist at very high levels during long-term antiretroviral treatment of HIV disease. PLoS One. 2010;5:e8886. doi: 10.1371/journal.pone.0008886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.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]
- 126.Nieto FJ, Adam E, Sorlie P, Farzadegan H, Melnick JL, Comstock GW, et al. 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]
- 127.Lurain NS, Hanson BA, Hotton AL, Weber KM, Cohen MH, Landay AL. The Association of Human Cytomegalovirus with Biomarkers of Inflammation and Immune Activation in HIV-1-Infected Women. AIDS Res Hum Retroviruses. 2016;32:134–143. doi: 10.1089/aid.2015.0169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Parrinello CM, Sinclair E, Landay AL, Lurain N, Sharrett AR, Gange SJ, 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]
- 129.Hsue PY, Hunt PW, Sinclair E, Bredt B, Franklin A, Killian M, 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]
- 130.Hunt PW, Martin JN, Sinclair E, Epling L, Teague J, Jacobson MA, et al. 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]
- 131.Emery S, Neuhaus JA, Phillips AN, Babiker A, Cohen CJ, Gatell JM, et al. Major clinical outcomes in antiretroviral therapy (ART)-naive participants and in those not receiving ART at baseline in the SMART study. J Infect Dis. 2008;197:1133–1144. doi: 10.1086/586713. [DOI] [PubMed] [Google Scholar]
- 132.Dornadula G, Zhang H, VanUitert B, Stern J, Livornese L, Jr, Ingerman MJ, et al. Residual HIV-1 RNA in blood plasma of patients taking suppressive highly active antiretroviral therapy. JAMA. 1999;282:1627–1632. doi: 10.1001/jama.282.17.1627. [DOI] [PubMed] [Google Scholar]
- 133.Maldarelli F, Palmer S, King MS, Wiegand A, Polis MA, Mican J, et al. ART suppresses plasma HIV-1 RNA to a stable set point predicted by pretherapy viremia. PLoS Pathog. 2007;3:e46. doi: 10.1371/journal.ppat.0030046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Dinoso JB, Kim SY, Wiegand AM, Palmer SE, Gange SJ, Cranmer L, et al. Treatment intensification does not reduce residual HIV-1 viremia in patients on highly active antiretroviral therapy. Proc Natl Acad Sci U S A. 2009;106:9403–9408. doi: 10.1073/pnas.0903107106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Gandhi RT, Zheng L, Bosch RJ, Chan ES, Margolis DM, Read S, et al. The effect of raltegravir intensification on low-level residual viremia in HIV-infected patients on antiretroviral therapy: a randomized controlled trial. PLoS Med. 2010;7 doi: 10.1371/journal.pmed.1000321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Rusconi S, Vitiello P, Adorni F, Colella E, Foca E, Capetti A, et al. Maraviroc as intensification strategy in HIV-1 positive patients with deficient immunological response: an Italian randomized clinical trial. PLoS One. 2013;8:e80157. doi: 10.1371/journal.pone.0080157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hunt PW, Shulman NS, Hayes TL, Dahl V, Somsouk M, Funderburg NT, et al. The immunologic effects of maraviroc intensification in treated HIV-infected individuals with incomplete CD4+ T-cell recovery: a randomized trial. Blood. 2013;121:4635–4646. doi: 10.1182/blood-2012-06-436345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Ananworanich J, Chomont N, Fletcher JL, Pinyakorn S, Schuetz A, Sereti I, et al. Markers of HIV reservoir size and immune activation after treatment in acute HIV infection with and without raltegravir and maraviroc intensification. J Virus Erad. 2015;1:116–122. doi: 10.1016/S2055-6640(20)30482-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.van Lelyveld SF, Drylewicz J, Krikke M, Veel EM, Otto SA, Richter C, et al. Maraviroc Intensification of cART in Patients with Suboptimal Immunological Recovery: A 48-Week, Placebo-Controlled Randomized Trial. PLoS One. 2015;10:e0132430. doi: 10.1371/journal.pone.0132430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Sahu GK. Potential implication of residual viremia in patients on effective antiretroviral therapy. AIDS Res Hum Retroviruses. 2015;31:25–35. doi: 10.1089/aid.2014.0194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Gupta SK, Mi D, Dube MP, Saha CK, Johnson RM, Stein JH, et al. Pentoxifylline, inflammation, and endothelial function in HIV-infected persons: a randomized, placebo-controlled trial. PLoS One. 2013;8:e60852. doi: 10.1371/journal.pone.0060852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Gupta SK, Johnson RM, Saha C, Mather KJ, Greenwald ML, Waltz JS, et al. Improvement in HIV-related endothelial dysfunction using the anti-inflammatory agent salsalate: a pilot study. AIDS. 2008;22:653–655. doi: 10.1097/QAD.0b013e3282f470d2. [DOI] [PubMed] [Google Scholar]
- 143.Hileman CO, Carman TL, Gripshover BM, O’Riordan M, Storer NJ, Harrill DE, et al. Salsalate is poorly tolerated and fails to improve endothelial function in virologically suppressed HIV-infected adults. AIDS. 2010;24:1958–1961. doi: 10.1097/QAD.0b013e32833c3251. [DOI] [PubMed] [Google Scholar]
- 144.Somsouk M, Dunham RM, Cohen M, Albright R, Abdel-Mohsen M, Liegler T, et al. The immunologic effects of mesalamine in treated HIV-infected individuals with incomplete CD4+ T cell recovery: a randomized crossover trial. PLoS One. 2014;9:e116306. doi: 10.1371/journal.pone.0116306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Sandler NG, Zhang X, Bosch RJ, Funderburg NT, Choi AI, Robinson JK, et al. Sevelamer does not decrease lipopolysaccharide or soluble CD14 levels but decreases soluble tissue factor, low-density lipoprotein (LDL) cholesterol, and oxidized LDL cholesterol levels in individuals with untreated HIV infection. J Infect Dis. 2014;210:1549–1554. doi: 10.1093/infdis/jiu305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Sereti I, Estes JD, Thompson WL, Morcock DR, Fischl MA, Croughs T, et al. Decreases in colonic and systemic inflammation in chronic HIV infection after IL-7 administration. PLoS Pathog. 2014;10:e1003890. doi: 10.1371/journal.ppat.1003890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Micci L, Ryan ES, Fromentin R, Bosinger SE, Harper JL, He T, et al. Interleukin-21 combined with ART reduces inflammation and viral reservoir in SIV-infected macaques. J Clin Invest. 2015;125:4497–4513. doi: 10.1172/JCI81400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Villar-Garcia J, Hernandez JJ, Guerri-Fernandez R, Gonzalez A, Lerma E, Guelar A, et al. Effect of probiotics (Saccharomyces boulardii) on microbial translocation and inflammation in HIV-treated patients: a double-blind, randomized, placebo-controlled trial. J Acquir Immune Defic Syndr. 2015;68:256–263. doi: 10.1097/QAI.0000000000000468. [DOI] [PubMed] [Google Scholar]
- 149.d’Ettorre G, Ceccarelli G, Giustini N, Serafino S, Calantone N, De Girolamo G, et al. Probiotics Reduce Inflammation in Antiretroviral Treated, HIV-Infected Individuals: Results of the “Probio-HIV” Clinical Trial. PLoS One. 2015;10:e0137200. doi: 10.1371/journal.pone.0137200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Stiksrud B, Nowak P, Nwosu FC, Kvale D, Thalme A, Sonnerborg A, et al. Reduced Levels of D-dimer and Changes in Gut Microbiota Composition After Probiotic Intervention in HIV-Infected Individuals on Stable ART. J Acquir Immune Defic Syndr. 2015;70:329–337. doi: 10.1097/QAI.0000000000000784. [DOI] [PubMed] [Google Scholar]
- 151.Sperber K, Louie M, Kraus T, Proner J, Sapira E, Lin S, et al. Hydroxychloroquine treatment of patients with human immunodeficiency virus type 1. Clin Ther. 1995;17:622–636. doi: 10.1016/0149-2918(95)80039-5. [DOI] [PubMed] [Google Scholar]
- 152.Murray SM, Down CM, Boulware DR, Stauffer WM, Cavert WP, Schacker TW, et al. Reduction of immune activation with chloroquine therapy during chronic HIV infection. J Virol. 2010;84:12082–12086. doi: 10.1128/JVI.01466-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Piconi S, Parisotto S, Rizzardini G, Passerini S, Terzi R, Argenteri B, et al. Hydroxychloroquine drastically reduces immune activation in HIV-infected, antiretroviral therapy-treated immunologic nonresponders. Blood. 2011;118:3263–3272. doi: 10.1182/blood-2011-01-329060. [DOI] [PubMed] [Google Scholar]
- 154.Paton NI, Goodall RL, Dunn DT, Franzen S, Collaco-Moraes Y, Gazzard BG, et al. Effects of hydroxychloroquine on immune activation and disease progression among HIV-infected patients not receiving antiretroviral therapy: a randomized controlled trial. JAMA. 2012;308:353–361. doi: 10.1001/jama.2012.6936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Routy JP, Angel JB, Patel M, Kanagaratham C, Radzioch D, Kema I, et al. Assessment of chloroquine as a modulator of immune activation to improve CD4 recovery in immune nonresponding HIV-infected patients receiving antiretroviral therapy. HIV Med. 2015;16:48–56. doi: 10.1111/hiv.12171. [DOI] [PubMed] [Google Scholar]
- 156.Stone NJ, Robinson JG, Lichtenstein AH, Bairey Merz CN, Blum CB, Eckel RH, et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;63:2889–2934. doi: 10.1016/j.jacc.2013.11.002. [DOI] [PubMed] [Google Scholar]
- 157.Funderburg NT, Jiang Y, Debanne SM, Labbato D, Juchnowski S, Ferrari B, et al. Rosuvastatin reduces vascular inflammation and T-cell and monocyte activation in HIV-infected subjects on antiretroviral therapy. J Acquir Immune Defic Syndr. 2015;68:396–404. doi: 10.1097/QAI.0000000000000478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Longenecker CT, Jiang Y, Debanne SM, DE L, B K, NJ S, et al. Rosuvastatin arrests progression of carotid intima-media thickness in treated HIV. Conference on Retroviruses and Opportunistic Infections (CROI); Seattle, WA. 2015. [Google Scholar]
- 159.Lo J, Lu MT, Ihenachor EJ, Wei J, Looby SE, Fitch KV, et al. Effects of statin therapy on coronary artery plaque volume and high-risk plaque morphology in HIV-infected patients with subclinical atherosclerosis: a randomised, double-blind, placebo-controlled trial. The Lancet HIV. 2015;2:e52–e63. doi: 10.1016/S2352-3018(14)00032-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Hileman CO, Turner R, N TF, Semba RD, McComsey GA. Changes in oxidized lipids drive the improvement in monocyte activation and vascular disease after statin therapy in HIV. AIDS. 2016;30:65–73. doi: 10.1097/QAD.0000000000000885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Nou E, Lu MT, Looby SE, Fitch KV, Kim EA, Lee H, et al. 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]
- 162.Zidar DA, Juchnowski S, Ferrari B, Clagett B, Pilch-Cooper HA, Rose S, et al. Oxidized LDL Levels Are Increased in HIV Infection and May Drive Monocyte Activation. J Acquir Immune Defic Syndr. 2015;69:154–160. doi: 10.1097/QAI.0000000000000566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Thusgaard M, Christensen JH, Morn B, Andersen TS, Vige R, Arildsen H, et al. Effect of fish oil (n-3 polyunsaturated fatty acids) on plasma lipids, lipoproteins and inflammatory markers in HIV-infected patients treated with antiretroviral therapy: a randomized, double-blind, placebo-controlled study. Scand J Infect Dis. 2009;41:760–766. doi: 10.1080/00365540903168056. [DOI] [PubMed] [Google Scholar]
- 164.Oliveira JM, Rondo PH, Lima LR, Fortuna ES, Yudkin JS. Effects of a Low Dose of Fish Oil on Inflammatory Markers of Brazilian HIV-Infected Adults on Antiretroviral Therapy: A Randomized, Parallel, Placebo-Controlled Trial. Nutrients. 2015;7:6520–6528. doi: 10.3390/nu7085294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Hileman CO, Carman TL, Storer NJ, Labbato DE, White CA, McComsey GA. Omega-3 fatty acids do not improve endothelial function in virologically suppressed HIV-infected men: a randomized placebo-controlled trial. AIDS Res Hum Retroviruses. 2012;28:649–655. doi: 10.1089/aid.2011.0088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.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]
- 167.Dube MP, Komarow L, Fichtenbaum CJ, Cadden JJ, Overton ET, Hodis HN, et al. Extended-Release Niacin Versus Fenofibrate in HIV-Infected Participants With Low High-Density Lipoprotein Cholesterol: Effects on Endothelial Function, Lipoproteins, and Inflammation. Clin Infect Dis. 2015;61:840–849. doi: 10.1093/cid/civ385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.O’Brien M, Kitch D, Hunt P, Klingman KL, Stein JH, Funderburg NT, et al. Aspirin Fails to Impact Immune Activation or Endothelial Function in Treated HIV. Conference on Retroviruses and Opportunistic Infections (CROI); Boston, MA. 2016. [Google Scholar]
- 169.Srinivasa S, Fitch KV, Wong K, Torriani M, Mayhew C, Stanley T, et al. RAAS Activation Is Associated With Visceral Adiposity and Insulin Resistance Among HIV-infected Patients. J Clin Endocrinol Metab. 2015;100:2873–2882. doi: 10.1210/jc.2015-1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Boccara F, Auclair M, Cohen A, Lefevre C, Prot M, Bastard JP, et al. HIV protease inhibitors activate the adipocyte renin angiotensin system. Antivir Ther. 2010;15:363–375. doi: 10.3851/IMP1533. [DOI] [PubMed] [Google Scholar]
- 171.Baker JV, Huppler Hullsiek K, Prosser R, Duprez D, Grimm R, Tracy RP, et al. 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]
- 172.Hogh Kolbaek Kjaer AS, Brinkmann CR, Dinarello CA, Olesen R, Ostergaard L, Sogaard OS, et al. The histone deacetylase inhibitor panobinostat lowers biomarkers of cardiovascular risk and inflammation in HIV patients. AIDS. 2015;29:1195–1200. doi: 10.1097/QAD.0000000000000678. [DOI] [PubMed] [Google Scholar]
- 173.Parra S, Coll B, Aragones G, Marsillach J, Beltran R, Rull A, et al. Nonconcordance between subclinical atherosclerosis and the calculated Framingham risk score in HIV-infected patients: relationships with serum markers of oxidation and inflammation. HIV Med. 2010;11:225–231. doi: 10.1111/j.1468-1293.2009.00766.x. [DOI] [PubMed] [Google Scholar]
- 174.Tebas P, Spitsin S, Barrett JS, Tuluc F, Elci O, Korelitz JJ, et al. Reduction of soluble CD163, substance P, programmed death 1 and inflammatory markers: phase 1B trial of aprepitant in HIV-1-infected adults. AIDS. 2015;29:931–939. doi: 10.1097/QAD.0000000000000638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Gori A, Rizzardini G, Van’t Land B, Amor KB, van Schaik J, Torti C, et al. Specific prebiotics modulate gut microbiota and immune activation in HAART-naive HIV-infected adults: results of the “COPA” pilot randomized trial. Mucosal Immunol. 2011;4:554–563. doi: 10.1038/mi.2011.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Nakanjako D, Ssinabulya I, Nabatanzi R, Bayigga L, Kiragga A, Joloba M, et al. Atorvastatin reduces T-cell activation and exhaustion among HIV-infected cART-treated suboptimal immune responders in Uganda: a randomised crossover placebo-controlled trial. Trop Med Int Health. 2015;20:380–390. doi: 10.1111/tmi.12442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Ganesan A, Crum-Cianflone N, Higgins J, Qin J, Rehm C, Metcalf J, et al. High dose atorvastatin decreases cellular markers of immune activation without affecting HIV-1 RNA levels: results of a double-blind randomized placebo controlled clinical trial. J Infect Dis. 2011;203:756–764. doi: 10.1093/infdis/jiq115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Read SW, DeGrezia M, Ciccone EJ, DerSimonian R, Higgins J, Adelsberger JW, et al. The effect of leflunomide on cycling and activation of T-cells in HIV-1-infected participants. PLoS One. 2010;5:e11937. doi: 10.1371/journal.pone.0011937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Dhawan S, Puri RK, Kumar A, Duplan H, Masson JM, Aggarwal BB. Human immunodeficiency virus-1-tat protein induces the cell surface expression of endothelial leukocyte adhesion molecule-1, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1 in human endothelial cells. Blood. 1997;90:1535–1544. [PubMed] [Google Scholar]
- 180.Ren Z, Yao Q, Chen C. HIV-1 envelope glycoprotein 120 increases intercellular adhesion molecule-1 expression by human endothelial cells. Lab Invest. 2002;82:245–255. doi: 10.1038/labinvest.3780418. [DOI] [PubMed] [Google Scholar]
- 181.Duffy P, Wang X, Lin PH, Yao Q, Chen C. HIV Nef protein causes endothelial dysfunction in porcine pulmonary arteries and human pulmonary artery endothelial cells. J Surg Res. 2009;156:257–264. doi: 10.1016/j.jss.2009.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Jiang J, Fu W, Wang X, Lin PH, Yao Q, Chen C. HIV gp120 induces endothelial dysfunction in tumour necrosis factor-alpha-activated porcine and human endothelial cells. Cardiovasc Res. 2010;87:366–374. doi: 10.1093/cvr/cvq013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Olivetta E, Percario Z, Fiorucci G, Mattia G, Schiavoni I, Dennis C, et al. HIV-1 Nef induces the release of inflammatory factors from human monocyte/macrophages: involvement of Nef endocytotic signals and NF-kappa B activation. J Immunol. 2003;170:1716–1727. doi: 10.4049/jimmunol.170.4.1716. [DOI] [PubMed] [Google Scholar]
- 184.Wang T, Green LA, Gupta SK, Kim C, Wang L, Almodovar S, et al. Transfer of intracellular HIV Nef to endothelium causes endothelial dysfunction. PLoS One. 2014;9:e91063. doi: 10.1371/journal.pone.0091063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Weiss JM, Nath A, Major EO, Berman JW. HIV-1 Tat induces monocyte chemoattractant protein-1-mediated monocyte transmigration across a model of the human blood-brain barrier and up-regulates CCR5 expression on human monocytes. J Immunol. 1999;163:2953–2959. [PubMed] [Google Scholar]
- 186.Acheampong EA, Parveen Z, Muthoga LW, Kalayeh M, Mukhtar M, Pomerantz RJ. Human Immunodeficiency virus type 1 Nef potently induces apoptosis in primary human brain microvascular endothelial cells via the activation of caspases. J Virol. 2005;79:4257–4269. doi: 10.1128/JVI.79.7.4257-4269.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Green LA, Yi R, Petrusca D, Wang T, Elghouche A, Gupta SK, et al. HIV envelope protein gp120-induced apoptosis in lung microvascular endothelial cells by concerted upregulation of EMAP II and its receptor, CXCR3. Am J Physiol Lung Cell Mol Physiol. 2014;306:L372–382. doi: 10.1152/ajplung.00193.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Zauli G, Furlini G, Re MC, Milani D, Capitani S, La Placa M. Human immunodeficiency virus type 1 (HIV-1) tat-protein stimulates the production of interleukin-6 (IL-6) by peripheral blood monocytes. New Microbiol. 1993;16:115–120. [PubMed] [Google Scholar]
- 189.Orden S, De Pablo C, Rios-Navarro C, Martinez-Cuesta MA, Peris JE, Barrachina MD, et al. Efavirenz induces interactions between leucocytes and endothelium through the activation of Mac-1 and gp150,95. J Antimicrob Chemother. 2014;69:995–1004. doi: 10.1093/jac/dkt468. [DOI] [PubMed] [Google Scholar]
- 190.Baum PD, Sullam PM, Stoddart CA, McCune JM. Abacavir increases platelet reactivity via competitive inhibition of soluble guanylyl cyclase. AIDS. 2011;25:2243–2248. doi: 10.1097/QAD.0b013e32834d3cc3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Wang X, Mu H, Chai H, Liao D, Yao Q, Chen C. Human immunodeficiency virus protease inhibitor ritonavir inhibits cholesterol efflux from human macrophage-derived foam cells. Am J Pathol. 2007;171:304–314. doi: 10.2353/ajpath.2007.060965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Fu W, Chai H, Yao Q, Chen C. Effects of HIV protease inhibitor ritonavir on vasomotor function and endothelial nitric oxide synthase expression. J Acquir Immune Defic Syndr. 2005;39:152–158. [PubMed] [Google Scholar]
- 193.Zhong DS, Lu XH, Conklin BS, Lin PH, Lumsden AB, Yao Q, et al. HIV protease inhibitor ritonavir induces cytotoxicity of human endothelial cells. Arterioscler Thromb Vasc Biol. 2002;22:1560–1566. doi: 10.1161/01.atv.0000034707.40046.02. [DOI] [PubMed] [Google Scholar]
- 194.Dressman J, Kincer J, Matveev SV, Guo L, Greenberg RN, Guerin T, 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]