Skip to main content

Immune correlates of cardiovascular co-morbidity in HIV infected participants from South India

Abstract

Background

Understanding the immune correlates of cardiovascular disease (CVD) risk in HIV infection is an important area of investigation in the current era of aging with HIV infection. Less is known about CVD risk and HIV infection in developing nations where additional risk factors may be playing a role in the CVD development. In this study, we assessed the effects of systemic inflammation, microbial translocation (MT), T cell immune activation (IA), and nadir CD4 counts on cardiac function and arterial stiffness as markers of subclinical atherosclerosis in HIV-infected individuals.

Methods

People with HIV (PWH) who were ART naïve (n = 102) or virally suppressed on ART (n = 172) were stratified on nadir CD4 counts and compared to HIV-uninfected controls (n = 64). Determination was made of cardiac function via radial pulse wave and carotid intima thickness (C-IMT) measurements. Plasma biomarkers of inflammation and MT by ELISA or multiplex assays, and immune activation (IA) of T cells based HLA-DR and CD38 expression were investigated by flow cytometry. T-test, Mann–Whitney U test, and Spearman correlation were used to analyze study parameters.

Results

Reduction in cardiac function with lower cardiac ejection time (p < 0.001), stroke volume (p < 0.001), cardiac output (p = 0.007), higher arterial stiffness (p < 0.05) were identified in ART-naïve participants, compared to PWH on ART (p < 0.05). No significant difference in C-IMT values were noted. Higher inflammatory and MT markers were found in the ART-naïve group compared to treated group who were comparable to uninfected participants, except for having higher TNF-α (p < 0.001) and sCD14 (p < 0.001). Immune activation of CD4 and CD8 T-cells was greater in ART-naïve participants compared to ART-treated and uninfected controls (p < 0.05). Lower nadir CD4 counts, higher inflammation, and higher MT predicted poor cardiac measures in the ART-naïve with nadir CD4 < 200cells/mm3 manifesting the highest arterial stiffness, and lowest cardiac function, whereas ART-treated, even with nadir < 200 cells/mm3 were similar to uninfected in these measures.

Conclusions

In HIV-infected individuals, initiation of ART even at nadir of < 200 cells/mm3 may prevent or reverse cardiovascular disease outcomes that are easily measurable in low income countries.

Peer Review reports

Background

Understanding risk and mechanism of Cardiovascular disease (CVD) in HIV infection is under intense investigation in the developed world, but less is known about this comorbidity in developing nations where additional risk factors may be playing a role [1,2,3]. The present study was focused on people with HIV (PWH) from the Indian subcontinent who in general are more predisposed to CVD, with its occurrence at least a decade earlier in the general population than in people of European ancestry [4, 5]. Risk of mortality due to CVD before the age of 70 years is 52% among Indians as compared to 23% in Western populations [6]. The impact of HIV infection on CVD in people from India is understudied. Overall, the risk of CVD in PWH on ART has been found to be twice that of HIV-negative persons [7] but other factors can further alter these dynamics. This issue is particularly important in the “treat all” era where the risk for comorbidities persist despite virus suppression with antiretroviral therapy (ART) [8,9,10] and decline in AIDS-related mortality [11, 12]. However in developing nations, patients often do not enter into care until late in the disease, and the drugs used in ART regimens may vary and thereby influence patient outcomes [13,14,15,16]. The strategies for management of antiretroviral therapy (SMART) study that included people of various backgrounds clearly established the importance of continuous ART in reducing risk of CVD compared to intermittent ART [17], but the veterans aging cohort (VAC) Study using data collected only on veterans found that ART was associated with an increased risk of acute MI [18]. As our understanding of the potential impact of HIV infection on CVD evolves, more data are needed to delineate the detrimental effects of HIV on cardiac function in culturally and ethnically diverse populations from various parts of the world including low resource countries.

Among pathogenic factors that influence CVD the role of immune activation and inflammation in HIV-associated atherosclerosis has received much attention. A majority of published studies have focused on the role of the innate immune system, such as markers of monocyte and macrophage activation [19,20,21] on HIV-associated atherosclerosis. Increased inflammatory response has been implicated in CVD for the general population and may be partly driving the association between HIV and CVD, contributing to the persistent risk noted after adjusting for traditional CVD risk factors [15, 22,23,24]. For example, the inflammatory cytokine IL-6 was found to be elevated in progressive HIV infection [25,26,27,28,29] and to predict mortality in HIV infected participants [30, 31]. In addition, elevations in soluble inflammatory receptors, such as Tumor Necrosis Factor Receptor (TNFR) I and II, have been implicated in myocardial dysfunction after acute coronary syndromes, as well as in recurrent myocardial infarction (MI) and cardiac death [32, 33]. An association of gut-associated microbial translocation (MT) based on soluble CD14 (sCD14) and plasma lipopolysaccharide (LPS) levels and CVD risk has been reported in PWH [34].

Arterial elasticity along with carotid-intima media thickness (C-IMT) measurements are valuable tools to identify early vascular functional and structural abnormalities. These measures are easily adapted to outpatient facilities and have provided valuable information in studies of the influence of ART regimens on traditional CVD risk factors [34,35,36,37]. Torriani et al.reported improved brachial flow measures within 4 weeks of initial ART use, which was associated with a decline in HIV RNA levels [38]. Delayed entry into care can lead to initiation of ART at lower nadir CD4 counts, which have been associated with higher C-IMT [39, 40] and MI events [41] but this is not well established and controversies exist [40, 42]. Adopting measures such as C-IMT in outpatient facilities in LRC can potentially facilitate implementation of CVD prevention strategies in PWH [39, 43,44,45,46]. Data are needed to fill in the gaps in understanding how C-IMT and arterial stiffness are affected by nadir CD4 counts as well as the role of gut-associated MT. The present study analyzed the functional and structural changes of arterial vasculature using practical tools in an outpatient setting in treatment-naïve and ART-experienced HIV + participants from South India across different CD4 nadirs and their associations with T-cell immune activation, inflammation, and microbial translocation (MT). Our findings show that adverse impact of HIV on these measures of cardiac function in patients with low CD4 nadirs can potentially be reversed with ART initiation.

Methods

Study setting and subjects

Recruitment of participants for this study was conducted during 2014–2016 at YRG CARE, a tertiary care center in Chennai located in South India providing patient care to more than 20,000 PWH. The study enrolled 274 male and female participants with chronic HIV infection (HIV +) and 64 HIV-uninfected healthy controls (HC) at age > 18 yr. Among the HIV + participants, 102, with 50 males and 52 females were ART-naïve with no pre-exposure to any ART (Group 1). 172 with 114 males and 58 females were on ART for > 12 months viral suppression with plasma viral load of < 40 copies/mL on two consecutive measurements and CD4 counts > 500/μL (Group 2). HC were categorized as Group 3 with 18 males and 46 females. All the HC participants were from the same socio-demographic background as the HIV + participants. HC on pre-exposure prophylaxis, and pregnant women were excluded. In Groups 1 and 2, HIV + participants were stratified based on nadir CD4 counts of < 200 cells/µL (groups 1a and 2a); 200–350 cells/µL (groups 1b and 2b) and > 350 cells/µL (groups 1c and 2c). The study was approved by both YRG CARE and University of Miami institutional review boards, with written informed consent obtained from all enrolled participants. A detailed interview was conducted at time of enrollment to collect demographic information along with screening tests for HBV and HCV co-infections and cardiovascular assessment including a complete lipid profile. All measures of cardia function were obtained in the outpatient setting coupled to a clinic visit.

Sample processing and storage

40 ml venous blood was collected in EDTA containing sterile vacutainer tubes (BD Biosciences) at the visit. Plasma collection was performed by centrifuging the blood at 400 g for 10 min and plasma was collected and further centrifuged at 1000 g for 15 min to remove the platelets. Plasma was stored at −80 °C until further use. Peripheral blood mononuclear cells (PBMC) were collected from the whole blood by density gradient method and cells were cryopreserved in liquid N2. All the laboratory procedures used sterile endotoxin free tubes.

Carotid Intima-media Thickness (C-IMT) measurement

Doppler study of carotid and vertebral arteries was performed by B-mode ultrasound for all participants. Thickness of intima-media complex was measured in both the right and left common carotid arteries.

Measurement of cardiac functions and arterial stiffness

Stiffness in the arterial system was estimated by pulse-wave velocity (PWV) using the HDI/PulseWave CR-2000 (Hypertension Diagnostics, Inc., Eagan, MN), a diagnostic tool that was previously applied in the International Network for Strategic Initiatives in Global HIV Trials (INSIGHT) Strategic Timing of Anti Retroviral Treatment (START) arterial stiffness sub-study [47]. Along with Large Artery Elasticity index (LAE) and Small Artery Elasticity (SAE) index measures, systemic vascular resistance (SVR) and total vascular impedance (TVI) were measured as arterial stiffness parameters with measurements of pulse rate, stroke volume, stroke volume index, cardiac output, cardiac index, and cardiac ejection time to ascertain cardiac functioning.

Quantification of inflammatory and MT markers

The plasma inflammatory and MT markers TNFR-I, TNFR-II and sCD14 were measured using RayBio Tech ELISA kits, (RayBiotech, Inc., GA) after appropriate dilution of the plasma according to manufacturer’s protocol. Cytokines IL-1b, IL-2, IL-6, IL-8, IL-10, IL-12 (p70), IL-17, IFN-α2, IFN-γ and TNF-α levels were determined in undiluted plasma using Milliplex cytokine magnetic bead panel in the Magpix instrument (Luminex Corporation). These markers were selected based on their known association with systemic inflammation and immune activation. Median fluorescent intensities (MFI) were analyzed and cytokine levels were expressed as pg/mL. LPS was measured in plasma samples using the Limulus amebocyte lysate chromogenic endpoint assay (Lonza, MD, USA).

Analysis of markers of immune activation and exhaustion

Thawed cryopreserved PBMC were rested overnight. After overnight rest, cell recovery was between 75 and 85% with a viability of > 90% for all the samples. 1 × 106 cells were stained with antibodies against different cell surface markers in the dark at room temperature along with live-dead discriminator (Aqua). Cells were acquired on a BD LSRFortessa (BD Bioscience, San Jose, CA). Data were analyzed using FlowJo software (TreeStar V10.02, Ashland, OR). Frequencies of desired subsets were determined in gated live (Aqua) cell populations. Total CD4 + and CD8 + T-cells were analyzed for immune activation based on the co-expression of HLA-DR and CD38  [48].

Statistical analysis

Descriptive statistics such as percentages, means and standard deviation, and median and interquartile ranges were used to describe the demographic characteristics of the study population. Levene’s statistic was used to test for homogeneity of variances followed by independent samples T-test, Mann–Whitney U-test, ANOVA and Welch ANOVA if homogeneity of variances was not attained. Tukey or Games Howell post-hoc tests were used to compare various quantitative variables between the groups. Fold change over uninfected controls was calculated by dividing the HIV infected group data by uninfected controls data. In Fig. 1, uninfected controls are plotted as ‘1’ and values above and below 1 indicate fold change that is greater and lower respectively. Spearman correlation was used to identify associations of cardiac functional and structural parameters with inflammatory indices, microbial translocation, T-cell activation markers, and CD4 counts.

Fig. 1
figure 1

Comparison of cardiac functions and arterial stiffness parameters between the study groups with the uninfected control group: All the parameters were converted to fold changes with uninfected controls, while fold change for uninfected controls was taken as ‘1’. *Indicates statistical significance with p < 0.05

Results

Demographic characteristics of study population

Age was matched for all groups in the study. At the time of enrollment, 140 participants on ART were on first-line reverse transcriptase inhibitors (RTI; AZT/D4T/TDF + 3TC/FTC + EFV/NVP), while 32 were on PI-based (RTV-boosted LPV) second-line therapy. When stratified based on smoking habits, 8% of Group 1, 13% of Group 2 and 8% of the Group 3 were found to be current smokers, 24.6% of Group 1, 30% of Group 2 and 21% of the Group 3 were found to be past smokers, and 67.5% of Group 1 and 57% of Group 2 were found to be never smokers. Of the 274 HIV-infected participants, two were found to be HBV and HCV-positive. Characteristics of the study groups are shown in Table 1.

Table 1 Characteristics of the study cohorts

Cardiac function parameters are altered in treatment naïve participants

HIV infection may increase the risk of CVD, with previous studies showing increased risk of subclinical atherosclerosis [49]. The data in Table 2 shows that Group 1 had significantly lower cardiac ejection time (p < 0.001), stroke volume (p < 0.001), and cardiac output (p = 0.008) than Group 2. Group 3 had superior cardiac function compared to Group 1 in all of the same cardiac function parameters. Groups 2 and 3, however, did not vary significantly in cardiac function except for a significantly lower vascular impedance in group 2 (Table 2). Thus, ART treatment was found to potentially be cardioprotective, with CVD risk factors in Group 2 being comparable to healthy participants. Low CD4 counts have shown variable results in terms of CVD risk [50]. Among the subgroups, group 1a had the lowest cardiac functions of all three subgroups (Additional File 1: Table S1). Compared to uninfected controls, the fold change in cardiac measures were significantly lower in the group 1a (Fig. 1). Their cardiac ejection time, stroke volume, stroke volume index, cardiac output and cardiac index were significantly lower than groups 1b and 1c (Additional File 1:Table S1). Fold changes in cardiac function parameters in group 1c were comparable to uninfected controls with the exception of a lower stroke volume and stroke volume index (Fig. 1). Interestingly, Group 2a who had initiated ART at CD4 nadirs < 200 cells/mm3 showed minimal differences in Cardiac function measures from Group 3. These results show that PWH on ART who achieve CD4 reconstitution are near normal to HC in cardiac measures employed in this study, but may show some residual abnormalities e.g. in vascular impedance amongst patients who started ART at a low nadir CD4 count of < 200 cells/mm3.

Table 2 Measures of cardiac functions, and arterial stiffness in HIV + treatmentnaïve, on ART, and HIV uninfected participants

Higher arterial stiffness and vascular resistance in treatment naïve participants

Progression of HIV infection and declining CD4 counts have shown to increase arterial stiffness [51]. We observed that Group 2 had higher LAE (p = 0.001) and SAE (p = 0.019) than Group 1 as well as lower SVR (p = 0.003) and total vascular impedance (p = 0.046), as shown in Table 2. Group 3 did not vary significantly in arterial stiffness parameters from Group 2 while right IMT was different between Group 3 and Group 1 (Table 2). Of all the study populations, treatment-naïve participants had the greatest arterial stiffness and vascular resistance. The results in Additional File 1: Table S1 show that group 1a had increased arterial stiffness based on lower LAE and higher SVR compared to other treatment naïve groups (p < 0.05). Participants in group 1c had comparable levels of cardiac functions and arterial elasticity to uninfected controls, while 1a had altered cardiac measures (Fig. 1). Among the virologically suppressed participants in Group 2, those in groups 2a (p = 0.021) and 2c (p = 0.068) had longer duration of treatment than group 2b (Table 1). From the data in Additional File 1: Table S1, no significant difference in arterial stiffness was noted between treated groups initiating ART in different nadir CD4 groups. Thus, ART may preserve arterial elasticity regardless of starting nadir CD4 counts.

Higher nadir CD4 counts correlated with better cardiac function

Lower nadir CD4 count is a marker of advanced disease and of more virus-induced CD4 T-cell destruction [52]. In the group 1, higher nadir CD4 counts correlated positively with better cardiac function, including higher cardiac ejection time, higher stroke volume, higher stroke volume index, higher cardiac output, higher cardiac index, LAE, SAE, and lower SVR, (p < 0.05; Fig. 2; Table 3). Even though the significance noted was not very strong, the findings suggest that severity of immune compromise in HIV + persons increases the risk for CVD, underscoring the importance of initiating ART as early as possible.

Fig. 2
figure 2

Association of nadir CD4 counts with cardiac functioning and arterial stiffness in naïve participants. A Association of nadir CD4 counts with estimated cardiac ejection time. B Association of nadir CD4 counts with estimated stroke volume. C Association of nadir CD4 counts with estimated stroke volume index. D Association of nadir CD4 counts with estimated cardiac output. E Association of nadir CD4 counts with estimated cardiac index. F Association of nadir CD4 counts with large artery elasticity index. G Association of nadir CD4 counts with small artery elasticity index. H Association of nadir CD4 counts with systemic vascular resistance

Table 3 Correlation of CD4 T-cell counts with sub-clinical CVD markers in the HIV + treatment naïve group

T-cell immune activation markers in treatment naïve and ART groups associated with poor cardiac outcomes

Chronic immune activation and inflammation during HIV infection are thought to increase CVD progression [53, 54]. Our results revealed that CD4 and CD8 T-cell immune activation measured by co-expression of HLA-DR and CD38  was higher in Group 1 compared to Group 2 (p < 0.001) and Group 3 (p < 0.001; Table S2; Fig. 3). In the context of nadir CD4, groups 1a & 1b had higher immune activation in both CD4 and CD8 T-cells (p < 0.05) than group 1c. Group 1c also showed higher T-cell immune activation (p < 0.001) compared to group 3. Among group 2, CD4 T-cell immune activation was lowest in the group with the highest CD4 nadir (group 2c) compared to 2a (p = 0.001) and 2b (p = 0.03) and the levels were still higher than uninfected controls (Fig. 3a). CD8 T-cell immune activation did not differ between different nadir CD4 in group 2 (Fig. 3b). Comparing respective CD4 nadirs in groups 1 and 2, CD4 and CD8 T-cell immune activation was lower in group 2a, b, c than in groups 1a, b, c respectively (p < 0.01). In group 1, an inverse correlation was noted between CD4 and CD8 T-cell immune activation and cardiac ejection time, stroke volume, cardiac output, and cardiac index (p < 0.05; Table 4). In group 2 as well CD8 T-cell immune activation was inversely correlated with large and small artery elasticity (p < 0.05; Table 4). These results imply that lowering of both CD4 and CD8 T-cell  immune activation by ART may be cardioprotective.

Fig. 3
figure 3

CD4 and CD8 T-cell immune activation between treatment naïve and ART-treated participants stratified by nadir CD4 T-cell counts in comparison to the uninfected control group. CD4 and CD8 T cell immune activation were analyzed by Flow cytometry based on the surface expression of HLA-DR and CD38 dual positive CD4 and CD8 T cells. A, DR+38+: CD4 T-cell activation; B, CD8+DR+38+: CD8 T-cell activation; group 1a: Untreated participants   with nadir CD4 T-cell counts < 200 cells/mm3; group 1b: Untreated participants  with nadir CD4 T-cell counts 200–350 cells/mm3; group 1c: Untreated participants with nadir CD4 T-cell counts > 350 cells/mm3; group 2a: On ART participants with nadir CD4 T-cell counts < 200 cells/mm3; group 2b: On ART participants with nadir CD4 T-cell counts 200–350 cells/mm3; group 2c: On ART patients with nadir CD4 T-cell counts > 350 cells/mm3. Lines with asterix (*) show the significant difference (p < 0.05) between indicated groups with *p < 0.05, **p < 0.01, and ***p < 0.001

Table 4 Correlation coefficient of cardiac functional and arterial stiffness markers with T-cell immune activation in HIV + treatment naïve and on ART participants

Plasma inflammatory markers in naïve and ART groups and their association with sub-clinical CVD progression

Various markers of inflammation have been associated with CVD and increased mortality during HIV infection [55,56,57,58,59]. We found that Group 1 had higher amounts of certain plasma inflammatory cytokines compared to Groups 2 (p < 0.05) and 3 (p < 0.05), including IFNα2, IL-10, IL-6, TNFα, TNFR-1, and TNFR-2 (Table S2). Groups 2 and 3 had similar plasma inflammatory cytokines with the exceptions of TNF, and IL-12 which were higher in Group 2 (p = 0.001). Not only did Group 1 have the greatest T-cell immune activation, but also the highest amounts of inflammatory cytokines. Elevated IL-6 levels in naïve participants were associated with altered cardiac measures, including lower stroke volume and stroke volume index, lower cardiac output, lower LAE, and higher SVR (p < 0.05; Table 5). Higher TNFR1 and TNFR2 were also associated with poor cardiac measures parameters in group 1 (p < 0.05; Table 5). In the absence of ART, elevated markers of inflammation appear to have a detrimental role on CVD progression.

Table 5 Correlation coefficient of cardiac functional and arterial stiffness markers of inflammation and microbial translocation in HIV + treatment naïve participants

Plasma gut microbial translocation markers in treatment naïve and ART groups and their association with sub-clinical CVD progression

Bacterial translocation may lead to increased immune activation in HIV infection causing CVD progression [15, 34, 60]. Group 1 had higher plasma LPS compared to Groups 2 (p < 0.001) and 3 (p < 0.001), while plasma LPS levels in Groups 2 and 3 were not different (Table S2). Group 1 also had higher sCD14 compared to Group 3 (p = 0.004) but not to Group 2. In addition, sCD14 was higher in Group 2 compared to Group 3 (p < 0.001; Table S2). Our results show that the markers of microbial translocation are highest among treatment-naïve participants, with partial elevations seen even with ART treatment.

Similar to elevated IL-6 levels, elevated LPS levels in group 1 were associated with poor cardiac measures, including lower stroke volume and stroke volume index, lower cardiac output and cardiac index, lower LAE, and higher SVR (p < 0.05; Table 5). Likewise, elevated sCD14 was associated with lower cardiac ejection time, lower stroke volume and stroke volume index, lower cardiac output and cardiac index, and lower LAE (p < 0.05; Table 5). Overall, Similar to inflammatory biomarkers, microbial translocation was also correlated with poor cardiac function.

Discussion

We assessed sub-clinical CVD progression and cardiac health in terms of functional and structural cardiac changes in HIV-infected participants from South India. Our study identified decreased cardiac functioning and elevated inflammatory and T-cell immune activation markers in ART-naïve participants; however, it did not identify significant increases in C-IMT measurements during HIV-infection. These results indicate that ART-induced viral suppression may have cardio-protective effects, as seen with superior cardiac function and reduced arterial stiffness in ART-treated participants. In addition, lower nadir CD4 count was identified as a major factor associated with poor cardiac function in treatment-naïve participants.

The effects of HIV infection on the risk of CVD have shown conflicting data, with some studies showing increased risk of subclinical atherosclerosis [49], and others finding no association between HIV infection and CVD risk [18, 61, 62]. Many studies in people living with HIV have shown an effect of low CD4 + T-cell count on MI risk [63, 64], carotid atherosclerosis [40, 65], arterial stiffness [51], and endothelial dysfunction [21, 50, 66,67,68]. Our results demonstrated that low nadir CD4 counts were associated with reduced cardiac function, especially in ART-naïve participants, compared to those with nadir CD4 counts > 350cells/mm3. In addition, participants who initiated ART and had achieved viral suppression had better cardiac function parameters. The initiation of ART in HIV + participants was associated with reduced risk of CVD compared to naïve participants [18, 69]. Thus, our data highlights the importance of higher CD4 counts and early ART initiation as people who started ART at higher nadir CD4 counts were associated with decreased CVD risk in both naïve and ART-treated groups.

HIV infection has been shown to increase the risk of coronary artery disease through increasing plaque formation and arterial stiffness [45, 46, 49, 70, 71]. In our group 1, C-IMT levels did not vary with differences in nadir CD4; however, reduced LAE and SAE and elevated SVR and vascular impedance were noted with treatment naïve low nadir CD4 group. LAE and SAE as measures of arterial stiffness are effective markers of CVD, with central measures being able to independently predict future clinical events [72,73,74]. Changes in arterial stiffness was found to precede the development of atherosclerosis, which could explain why early changes in C-IMT were not found [75]. Other traditional risk factors, such as smoking, also contribute to atherosclerotic disease; however, reduced LAE and SAE [72] and impaired carotid and femoral arterial stiffness [76] was still seen in naïve participants even after adjusting for these risk factors in these studies. In addition, it was found that recently HIV-infected participants were not at increased risk of atherosclerotic disease when compared to uninfected controls [77], so well-treated HIV-seropositive participants, such as our ART treated group, may not vary significantly from healthy controls, as shown in our study.

Currently, the increased immune activation, increased expression of immune checkpoint inhibitors and chronic inflammation among HIV-infected individuals is thought to increase atherosclerotic progression and arterial stiffness [53, 54, 78]. We previously published that in ART naïve group, cardiac function was decreased with evidence of increased vascular resistance and that LAG3, PD1 or LAG3 plus PD1 expressing CD4 T cells were inversely correlated with cardiac function while being directly correlated with vascular resistance [78]. Our results show that both CD4 and CD8 T-cell activation was highest in treatment- naïve participants, and that this high immune activation correlated with lower cardiac function, such as lower stroke volume and cardiac output. Previous studies have shown that CD8 T-cell immune activation was more predictive of carotid plaque, while CD4 T-cell activation was associated with arterial stiffness [79, 80]. In our study, higher CD8 T-cell activation rather than CD4 T-cell activation was associated with markers of arterial stiffness, such as lower LAE and SAE. It has been demonstrated, however, that both activated CD4 + and CD8 + T cells can be associated with decreased arterial distensibility, likely due to both subtypes secreting pro-inflammatory mediators [80, 81]. Association of CD8 T cell immune activation with small and large artery elasticity in group 2 may be more associated with a role of the persistent immune activation on CVD outcomes over time. In group 1 ongoing viral replication and shorter duration of infection may not be able to capture the relationship of immune activation and arterial elasticity.

In ART-treated participants, the cardiac function was normalized, and no associations were seen with T-cell activation. This may be due to lower T-cell activation and improved cardiac functions seen in participants on ART.

The role of inflammatory mechanisms in the initiation and progression of CVD along with the rupture of atheromatous plaques has recently become more appreciated [55]. Various inflammatory markers, such as IL-6, TNFR-1, and TNFR-2, have been associated with CVD and increased mortality in HIV-positive participants [25, 27, 55,56,57,58,59,60]. In our study, IL-6 was associated with poor cardiac measures, including lower stroke volume and cardiac output, increased arterial stiffness, and higher SVR in naïve participants. Similarly, TNFR-1, TNFR-2, and to a lesser extent TNFα were also associated with markers of CVD. While still lower than naïve participants, immune activation of CD4 T-cells was higher in ART-treated compared to healthy participants, which could reflect ongoing immune activation even with successful suppression of HIV replication [25]. However, the risk of CVD is likely less severe given the lower immune activation of ART-treated participants compared to naïve. Inflammatory markers such as IL-6 not only correlated with CVD parameters, but also markers of microbial translocation, such as sCD14 [56].

Bacterial translocation has been implicated as a possible cause of immune activation leading to CVD progression [15, 34, 60]. In previous studies, elevated LPS and sCD14 were associated with increased coronary artery calcification and mortality in HIV infection, while in healthy individuals no association was found [21, 56, 59, 82, 83]. The naïve participants in our cohort had higher LPS and sCD14 markers, and both of these markers were associated with poor cardiovascular parameters, such as lower stroke volume and cardiac output, as well as increased arterial stiffness. LPS and sCD14 levels were lowest among participants that were ART-treated and healthy, likely due to the cardioprotective reduction in inflammatory markers and improved immune function [71, 84, 85].

Conclusions

Although it was previously thought that virologic suppression came at the expense of possible pro-atherogenic side effects of ART, our data suggest that the overall benefits of treatment in terms of virologic suppression with concomitant reduction in inflammatory markers and improvement of cardiac function are likely to be cardioprotective, ART treated patients had improved cardiac function, with Group 2 being comparable to healthy participants. Our data does not indicate how long they have to be on ART to start manifesting improvement in cardiac measures as the median duration of their treatment was 43 months (range 22–72 months). As ART regimens have improved dramatically and continue to improve in rapidity of achieving virus suppression, this is a moving target. Although the data were adjusted for all of the known risk factors, lack of information about other potential confounders such as other co-infections, like CMV may present among our study populations, and that could be a limitation with any cross-sectional study. Longitudinal studies are necessary to further explore and confirm the deterioration or improvement of arterial stiffness in participants on ART. Taken together, our data points towards the detrimental effects of persistent immune activation in CD4 T-cell populations on cardiac outcomes. Early ART initiation irrespective of CD4 counts may prevent adverse CVD outcomes by lowering immune activation, and thus limiting complications and morbidity in HIV-infected individuals.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on request.

Abbreviations

PWH:

People with HIV

MT:

Microbial translocation

ART:

Antiretroviral therapy

C-IMT:

Carotid intima-media thickness

IA:

Immune activation

CVD:

Cardiovascular disease

SMART:

Strategies for management of antiretroviral therapy

VAC:

Veterans aging cohort

TNFR:

Tumor necrosis factor receptor

sCD14:

Soluble CD14

LPS:

Lipopolysaccharide

PWV:

Pulse-wave velocity

INSIGHT:

International network for strategic initiatives in Global HIV trials

START:

Strategic timing of anti-retroviral treatment

LAE:

Large artery elasticity index

SAE:

Small artery elasticity index

SVR:

Systemic vascular resistance

TVI:

Total vascular impedance

References

  1. Palella FJ Jr, Baker RK, Moorman AC, Chmiel JS, Wood KC, Brooks JT, et al. Mortality in the highly active antiretroviral therapy era: changing causes of death and disease in the HIV outpatient study. J Acquir Immune Defic Syndr. 2006;43(1):27–34.

    Article  CAS  PubMed  Google Scholar 

  2. Bonnet F, Chene G, Thiebaut R, Dupon M, Lawson-Ayayi S, Pellegrin JL, et al. Trends and determinants of severe morbidity in HIV-infected patients: the ANRS CO3 Aquitaine Cohort, 2000–2004. HIV Med. 2007;8(8):547–54.

    Article  CAS  PubMed  Google Scholar 

  3. Achhra AC, Amin J, Law MG, Emery S, Gerstoft J, Gordin FM, et al. Immunodeficiency and the risk of serious clinical endpoints in a well studied cohort of treated HIV-infected patients. AIDS. 2010;24(12):1877–86.

    Article  PubMed  Google Scholar 

  4. Joshi P, Islam S, Pais P, Reddy S, Dorairaj P, Kazmi K, et al. Risk factors for early myocardial infarction in South Asians compared with individuals in other countries. JAMA. 2007;297(3):286–94.

    Article  CAS  PubMed  Google Scholar 

  5. Xavier D, Pais P, Devereaux PJ, Xie C, Prabhakaran D, Reddy KS, et al. Treatment and outcomes of acute coronary syndromes in India (CREATE): a prospective analysis of registry data. Lancet. 2008;371(9622):1435–42.

    Article  PubMed  Google Scholar 

  6. Leeder S, Raymond S, Greenberg H, Liu H, Esson K. A race against time: the challenge of cardiovascular diseases in developing economies: Trustees of Columbia University in the City of New York, pp. 1–95. 2004

  7. Mathabire Rucker SC, Tayea A, Bitilinyu-Bangoh J, Bermudez-Aza EH, Salumu L, Quiles IA, et al. High rates of hypertension, diabetes, elevated low-density lipoprotein cholesterol, and cardiovascular disease risk factors in HIV-infected patients in Malawi. AIDS. 2018;32(2):253–60.

    Article  PubMed  CAS  Google Scholar 

  8. Mary-Krause M, Cotte L, Simon A, Partisani M, Costagliola D. Clinical Epidemiology Group from the French Hospital D. Increased risk of myocardial infarction with duration of protease inhibitor therapy in HIV-infected men. AIDS. 2003;17(17):2479–86.

    Article  PubMed  Google Scholar 

  9. Group DADS, Friis-Moller N, Reiss P, Sabin CA, Weber R, Monforte A, et al. Class of antiretroviral drugs and the risk of myocardial infarction. N Engl J Med. 2007;356(17):1723–35.

    Article  Google Scholar 

  10. 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(7):2506–12.

    Article  CAS  PubMed  Google Scholar 

  11. May MT, Sterne JA, Costagliola D, Sabin CA, Phillips AN, Justice AC, et al. HIV treatment response and prognosis in Europe and North America in the first decade of highly active antiretroviral therapy: a collaborative analysis. Lancet. 2006;368(9534):451–8.

    Article  PubMed  CAS  Google Scholar 

  12. Wada N, Jacobson LP, Cohen M, French A, Phair J, Munoz A. Cause-specific life expectancies after 35 years of age for human immunodeficiency syndrome-infected and human immunodeficiency syndrome-negative individuals followed simultaneously in long-term cohort studies, 1984–2008. Am J Epidemiol. 2013;177(2):116–25.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Holmberg SD, Moorman AC, Williamson JM, Tong TC, Ward DJ, Wood KC, et al. Protease inhibitors and cardiovascular outcomes in patients with HIV-1. Lancet. 2002;360(9347):1747–8.

    Article  CAS  PubMed  Google Scholar 

  14. Iloeje UH, Yuan Y, L’Italien G, Mauskopf J, Holmberg SD, Moorman AC, et al. Protease inhibitor exposure and increased risk of cardiovascular disease in HIV-infected patients. HIV Med. 2005;6(1):37–44.

    Article  CAS  PubMed  Google Scholar 

  15. Teer E, Essop MF. HIV and cardiovascular disease: role of immunometabolic perturbations. Physiology. 2018;33(1):74–82.

    Article  CAS  PubMed  Google Scholar 

  16. Mikuła T, Peller M, Balsam P, Suchacz MM, Sapula M, Koltowski L, Glowczynska R, Opolski G, Filipiak KJ, Wiercinska-Drapalo A. High platelet count and high low-density lipoprotein level may be an independent marker of increased arterial stiffness in adult HIV-infected persons. HIV AIDS Rev Int J HIV-Relat Probl. 2019;18(1):14–8.

    Article  Google Scholar 

  17. Bozzette SA, Ake CF, Tam HK, Chang SW, Louis TA. Cardiovascular and cerebrovascular events in patients treated for human immunodeficiency virus infection. N Engl J Med. 2003;348(8):702–10.

    Article  CAS  PubMed  Google Scholar 

  18. 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(8):614–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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(8):1227–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Subramanian S, Tawakol A, Burdo TH, Abbara S, Wei J, Vijayakumar J, et al. Arterial inflammation in patients with HIV. JAMA. 2012;308(4):379–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Liang H, Xie Z, Shen T. Monocyte activation and cardiovascular disease in HIV infection. Cell Mol Immunol. 2017;14(12):960–2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Teer E, Joseph DE, Driescher N, Nell TA, Dominick L, Midgley N, et al. HIV and cardiovascular diseases risk: exploring the interplay between T-cell activation, coagulation, monocyte subsets, and lipid subclass alterations. Am J Physiol Heart Circ Physiol. 2019;316(5):H1146–57.

    Article  CAS  PubMed  Google Scholar 

  23. Libby P, Ridker PM, Hansson GK. Leducq Transatlantic Network on A. Inflammation in atherosclerosis: from pathophysiology to practice. J Am Coll Cardiol. 2009;54(23):2129–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Circulation. 2002;105(9):1135–43.

    Article  CAS  PubMed  Google Scholar 

  25. 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(12):1788–95.

    Article  CAS  PubMed  Google Scholar 

  26. Calmy A, Gayet-Ageron A, Montecucco F, Nguyen A, Mach F, Burger F, et al. HIV increases markers of cardiovascular risk: results from a randomized, treatment interruption trial. AIDS. 2009;23(8):929–39.

    Article  CAS  PubMed  Google Scholar 

  27. Baker J, Ayenew W, Quick H, Hullsiek KH, Tracy R, Henry K, et al. High-density lipoprotein particles and markers of inflammation and thrombotic activity in patients with untreated HIV infection. J Infect Dis. 2010;201(2):285–92.

    Article  CAS  PubMed  Google Scholar 

  28. 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(10):1509–17.

    Article  PubMed  Google Scholar 

  29. Baker JV, Sharma S, Grund B, Rupert A, Metcalf JA, Schechter M, et al. Systemic inflammation, coagulation, and clinical risk in the START trial. Open Forum Infect Dis. 2017;4(4):ofx262.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. 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(10): e203.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Tien PC, Choi AI, Zolopa AR, Benson C, Tracy R, Scherzer R, et al. Inflammation and mortality in HIV-infected adults: analysis of the FRAM study cohort. J Acquir Immune Defic Syndr. 2010;55(3):316–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ridker PM, Rifai N, Pfeffer M, Sacks F, Lepage S, Braunwald E. Elevation of tumor necrosis factor-alpha and increased risk of recurrent coronary events after myocardial infarction. Circulation. 2000;101(18):2149–53.

    Article  CAS  PubMed  Google Scholar 

  33. Nian M, Lee P, Khaper N, Liu P. Inflammatory cytokines and postmyocardial infarction remodeling. Circ Res. 2004;94(12):1543–53.

    Article  CAS  PubMed  Google Scholar 

  34. 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(10):1558–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Stein JH, Klein MA, Bellehumeur JL, McBride PE, Wiebe DA, Otvos JD, et al. Use of human immunodeficiency virus-1 protease inhibitors is associated with atherogenic lipoprotein changes and endothelial dysfunction. Circulation. 2001;104(3):257–62.

    Article  CAS  PubMed  Google Scholar 

  36. Charakida M, Donald AE, Green H, Storry C, Clapson M, Caslake M, et al. Early structural and functional changes of the vasculature in HIV-infected children: impact of disease and antiretroviral therapy. Circulation. 2005;112(1):103–9.

    Article  PubMed  Google Scholar 

  37. Johnsen S, Dolan SE, Fitch KV, Kanter JR, Hemphill LC, Connelly JM, et al. Carotid intimal medial thickness in human immunodeficiency virus-infected women: effects of protease inhibitor use, cardiac risk factors, and the metabolic syndrome. J Clin Endocrinol Metab. 2006;91(12):4916–24.

    Article  CAS  PubMed  Google Scholar 

  38. 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(7):569–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. 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(3):340–4.

    Article  CAS  PubMed  Google Scholar 

  40. Kaplan RC, Kingsley LA, Gange SJ, Benning L, Jacobson LP, Lazar J, et al. Low CD4+ T-cell count as a major atherosclerosis risk factor in HIV-infected women and men. AIDS. 2008;22(13):1615–24.

    Article  PubMed  Google Scholar 

  41. 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(17):2735–48.

    Article  PubMed  Google Scholar 

  42. Delaney JA, Scherzer R, Biggs ML, Shliplak MG, Polak JF, Currier JS, et al. Associations of antiretroviral drug use and HIV-specific risk factors with carotid intima-media thickness. AIDS. 2010;24(14):2201–9.

    Article  CAS  PubMed  Google Scholar 

  43. Lorenz MW, Markus HS, Bots ML, Rosvall M, Sitzer M. Prediction of clinical cardiovascular events with carotid intima-media thickness: a systematic review and meta-analysis. Circulation. 2007;115(4):459–67.

    Article  PubMed  Google Scholar 

  44. Hsue PY, Lo JC, Franklin A, Bolger AF, Martin JN, Deeks SG, et al. Progression of atherosclerosis as assessed by carotid intima-media thickness in patients with HIV infection. Circulation. 2004;109(13):1603–8.

    Article  PubMed  Google Scholar 

  45. Nanoudis S, Pikilidou M, Yavropoulou M, Gogou C, Papagianni M, Gogou A, et al. Markers of arterial stiffness and atherosclerosis in hiv-infected individuals. J Hypertens. 2018;1(36):e198.

    Google Scholar 

  46. Mosepele M, Hemphill LC, Moloi W, Moyo S, Nkele I, Makhema J, et al. Pre-clinical carotid atherosclerosis and sCD163 among virally suppressed HIV patients in Botswana compared with uninfected controls. PLoS ONE. 2017;12(6):e0179994.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Baker JV, Engen NW, Huppler Hullsiek K, Stephan C, Jain MK, Munderi P, et al. Assessment of arterial elasticity among HIV-positive participants with high CD4 cell counts: a substudy of the INSIGHT Strategic Timing of AntiRetroviral Treatment (START) trial. HIV Med. 2015;16(Suppl 1):109–18.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Alcaide ML, Parmigiani A, Pallikkuth S, Roach M, Freguja R, Della Negra M, et al. Immune activation in HIV-infected aging women on antiretrovirals–implications for age-associated comorbidities: a cross-sectional pilot study. PLoS ONE. 2013;8(5):e63804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. 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(7):458–67.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Ho JE, Scherzer R, Hecht FM, Maka K, Selby V, Martin JN, et al. The association of CD4+ T-cell counts and cardiovascular risk in treated HIV disease. AIDS. 2012;26(9):1115–20.

    Article  PubMed  Google Scholar 

  51. Ho JE, Deeks SG, Hecht FM, Xie Y, Schnell A, Martin JN, et al. Initiation of antiretroviral therapy at higher nadir CD4+ T-cell counts is associated with reduced arterial stiffness in HIV-infected individuals. AIDS. 2010;24(12):1897–905.

    Article  PubMed  Google Scholar 

  52. Negredo E, Massanella M, Puig J, Perez-Alvarez N, Gallego-Escuredo JM, Villarroya J, et al. Nadir CD4 T cell count as predictor and high CD4 T cell intrinsic apoptosis as final mechanism of poor CD4 T cell recovery in virologically suppressed HIV-infected patients: clinical implications. Clin Infect Dis. 2010;50(9):1300–8.

    Article  PubMed  Google Scholar 

  53. 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(3):349–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. 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(3):370–8.

    Article  CAS  PubMed  Google Scholar 

  55. 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(9):e44454.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sandler NG, Wand H, Roque A, Law M, Nason MC, Nixon DE, et al. Plasma levels of soluble CD14 independently predict mortality in HIV infection. J Infect Dis. 2011;203(6):780–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Borges AH, Silverberg MJ, Wentworth D, Grulich AE, Fatkenheuer G, Mitsuyasu R, et al. Predicting risk of cancer during HIV infection: the role of inflammatory and coagulation biomarkers. AIDS. 2013;27(9):1433–41.

    Article  CAS  PubMed  Google Scholar 

  58. 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(8):1248–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Longenecker CT, Jiang Y, Orringer CE, Gilkeson RC, Debanne S, Funderburg NT, Lederman MM, Storer N, Labbato DE, McComsey GA. Soluble CD14 is independently associated with coronary calcification and extent of subclinical vascular disease in treated HIV infection. AIDS 2014;28(7):969–77

  60. Subramanya V, McKay HS, Brusca RM, Palella FJ, Kingsley LA, Witt MD, et al. Inflammatory biomarkers and subclinical carotid atherosclerosis in HIV-infected and HIV-uninfected men in the Multicenter AIDS Cohort Study. PLoS ONE. 2019;14(4):e0214735.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Phillips AN, Neaton J, Lundgren JD. The role of HIV in serious diseases other than AIDS. AIDS. 2008;22(18):2409–18.

    Article  PubMed  Google Scholar 

  62. 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(4):600–7.

    Article  CAS  PubMed  Google Scholar 

  63. 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(4):435–47.

    Article  CAS  PubMed  Google Scholar 

  64. 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(2):160–6.

    Article  CAS  PubMed  Google Scholar 

  65. Guaraldi G, Zona S, Alexopoulos N, Orlando G, Carli F, Ligabue G, et al. Coronary aging in HIV-infected patients. Clin Infect Dis. 2009;49(11):1756–62.

    Article  PubMed  Google Scholar 

  66. Dube MP, Lipshultz SE, Fichtenbaum CJ, Greenberg R, Schecter AD, Fisher SD, et al. Effects of HIV infection and antiretroviral therapy on the heart and vasculature. Circulation. 2008;118(2):e36-40.

    Article  CAS  PubMed  Google Scholar 

  67. 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(9):1325–32.

    Article  PubMed  Google Scholar 

  68. Anand AR, Rachel G, Parthasarathy D. HIV proteins and endothelial dysfunction: implications in cardiovascular disease. Front Cardiovasc Med. 2018;5:185.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Group ISS, Lundgren JD, Babiker AG, Gordin F, Emery S, Grund B, et al. Initiation of antiretroviral therapy in early asymptomatic HIV infection. N Engl J Med. 2015;373(9):795–807.

    Article  CAS  Google Scholar 

  70. Ngatchou W, Lemogoum D, Ndobo P, Yagnigni E, Tiogou E, Nga E, et al. Increased burden and severity of metabolic syndrome and arterial stiffness in treatment-naive HIV+ patients from Cameroon. Vasc Health Risk Manag. 2013;9:509–16.

    Article  PubMed  PubMed Central  Google Scholar 

  71. O’Brien MP, Zafar MU, Rodriguez JC, Okoroafor I, Heyison A, Cavanagh K, et al. Targeting thrombogenicity and inflammation in chronic HIV infection. Sci Adv. 2019;5(6):eaav5463.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Baker JV, Duprez D, Rapkin J, Hullsiek KH, Quick H, Grimm R, et al. Untreated HIV infection and large and small artery elasticity. J Acquir Immune Defic Syndr. 2009;52(1):25–31.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Vlachopoulos C, Aznaouridis K, O’Rourke MF, Safar ME, Baou K, Stefanadis C. Prediction of cardiovascular events and all-cause mortality with central haemodynamics: a systematic review and meta-analysis. Eur Heart J. 2010;31(15):1865–71.

    Article  PubMed  Google Scholar 

  74. Schillaci G, De Socio GV, Pucci G, Mannarino MR, Helou J, Pirro M, et al. Aortic stiffness in untreated adult patients with human immunodeficiency virus infection. Hypertension. 2008;52(2):308–13.

    Article  CAS  PubMed  Google Scholar 

  75. Oren A, Vos LE, Uiterwaal CS, Grobbee DE, Bots ML. Aortic stiffness and carotid intima-media thickness: two independent markers of subclinical vascular damage in young adults? Eur J Clin Invest. 2003;33(11):949–54.

    Article  CAS  PubMed  Google Scholar 

  76. van Vonderen MG, Smulders YM, Stehouwer CD, Danner SA, Gundy CM, Vos F, et al. Carotid intima-media thickness and arterial stiffness in HIV-infected patients: the role of HIV, antiretroviral therapy, and lipodystrophy. J Acquir Immune Defic Syndr. 2009;50(2):153–61.

    Article  PubMed  Google Scholar 

  77. 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(3):287–93.

    Article  CAS  PubMed  Google Scholar 

  78. Pallikkuth S, Pahwa R, Kausalya B, Saravanan S, Pan L, Vignesh R, et al. Cardiac morbidity in HIV infection is associated with checkpoint inhibitor LAG-3 on CD4 T cells. PLoS ONE. 2018;13(10):e0206256.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. 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(4):452–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Kaplan RC, Sinclair E, Landay AL, Lurain N, Sharrett AR, Gange SJ, et al. T cell activation predicts carotid artery stiffness among HIV-infected women. Atherosclerosis. 2011;217(1):207–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Grome HN, Barnett L, Hagar CC, Harrison DG, Kalams SA, Koethe JR. Association of T cell and macrophage activation with arterial vascular health in HIV. AIDS Res Hum Retroviruses. 2017;33(2):181–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Morange PE, Tiret L, Saut N, Luc G, Arveiler D, Ferrieres J, et al. TLR4/Asp299Gly, CD14/C-260T, plasma levels of the soluble receptor CD14 and the risk of coronary heart disease: The PRIME Study. Eur J Hum Genet. 2004;12(12):1041–9.

    Article  CAS  PubMed  Google Scholar 

  83. 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(8):1219–28.

    Article  CAS  PubMed  Google Scholar 

  84. Triant VA. HIV infection and coronary heart disease: an intersection of epidemics. J Infect Dis. 2012;205(Suppl 3):S355–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Romero-Sanchez M, Gonzalez-Serna A, Pacheco YM, Ferrando-Martinez S, Machmach K, Garcia-Garcia M, et al. Different biological significance of sCD14 and LPS in HIV-infection: importance of the immunovirology stage and association with HIV-disease progression markers. J Infect. 2012;65(5):431–8.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to the clinical and laboratory staff at YRG Centre for AIDS Research and Education, VHS, Chennai, India, for assistance with the study, and for resources at the Laboratory Sciences Core at the Miami Center for AIDS Research (Miami CFAR) and the Flow Cytometry Core facility at the University of Miami Sylvester Comprehensive Cancer Center. We thank Margaret Roach, Maria Pallin and Varghese George for technical assistance and the participants for participation in this study.

Funding

This work was supported by National Institutes of Health Grant: R21AI106373 (to S.P.); Indian Council for Medical Research, New Delhi, India Grant ECD/NTF/17/2013–14 (to NK), and the Miami Center for AIDS Research (P30AI073961). Funders have no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

BK, sample collection, perform experiments, data analysis, clinical database, initial draft writing; SS, sample collection, clinical data; S. Palli, RP, and S. Pahwa provided intellectual input and contributed to the experimental design; S. Palli, perform experiments, data analysis and manuscript writing; RP, Data analysis, manuscript writing; SRS, manuscript revision; SI, KGM, performed experiment, data analysis, SSS: clinical data analysis; S Poongulali, Clinical data measures; NK, intellectual input, study design, manuscript editing; SP, Study conception and design, manuscript writing, final draft revision. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Savita Pahwa.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the Institutional Review Board of YRG CARE (IRB 00001423/FWA00000672) and University of Miami institutional review boards (IRB#20140606), with written informed consent obtained from all enrolled participants. All methods were carried out in accordance with relevant guidelines and regulations.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1.

Table S1. Measures of cardiac functioning, and arterial stiffness in naïve and ART-treated participants stratified by nadir CD4 counts. Table S2. Comparison of T-cell activation, plasma inflammatory markers, and MT markers in HIV-infected naïve participants, on ART participants, and HIV-uninfected control groups.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kausalya, B., Saravanan, S., Pallikkuth, S. et al. Immune correlates of cardiovascular co-morbidity in HIV infected participants from South India. BMC Immunol 23, 24 (2022). https://doi.org/10.1186/s12865-022-00498-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12865-022-00498-0

Keywords