Skip to main content

Circulating biomarker correlates of left atrial size and myocardial extracellular volume fraction among persons living with and without HIV

Abstract

Background

Infection with human immunodeficiency virus (HIV) is associated with higher risk for myocardial disease despite modern combination antiretroviral therapy (cART). Factors contributing to this excess risk, however, remain poorly characterized. We aimed to assess cross-sectional relationships between elevations of left atrial volume index (LAVI) and myocardial extracellular volume (ECV) fraction that have been reported in persons living with HIV and levels of circulating biomarkers of inflammation, fibrosis, and myocyte stretch among persons living with and without HIV (PLWH, PLWOH).

Methods

Participants from three cohorts of PLWH and PLWOH underwent cardiovascular magnetic resonance imaging for measurement of LAVI and ECV. Levels of circulating proteins (IL-6, sCD14, galectin-3, NT-proBNP, GDF-15, TIMP-2, MMP-2, and hsTnI) were measured using immunoassays. Associations were assessed using logistic and linear regression, adjusting for demographics, substance use, and clinical characteristics.

Results

Among 381 participants with and without HIV, median age (IQR) was 55.1 (51.2, 58.4) years, 28% were female, 69% were Black, and 46% were current smokers. Sixty-two percent were PLWH (n = 235), of whom 88% were receiving cART and 72% were virally suppressed. PLWH had higher levels of sCD14 (p = < 0.001), GDF-15 (p = < 0.001), and NT-proBNP (p = 0.03) compared to PLWOH, while levels of other biomarkers did not differ by HIV serostatus, including IL-6 (p = 0.84). Among PLWH, higher sCD14, GDF-15, and NT-proBNP were also associated with lower CD4 + cell count, and higher NT-proBNP was associated with detectable HIV viral load. NT-proBNP was associated with elevated LAVI (OR: 1.79 [95% CI: 1.31, 2.44]; p < 0.001) with no evidence of effect measure modification by HIV serostatus. Other associations between HIV-associated biomarkers and LAVI or ECV were small or imprecise.

Conclusions

Our findings suggest that elevated levels of sCD14, GDF-15, and NT-proBNP among PLWH compared to PLWOH observed in the current cART era may only minimally reflect HIV-associated elevations in LAVI and ECV. Future studies of excess risk of myocardial disease among contemporary cohorts of PLWH should investigate mechanisms other than those connoted by the studied biomarkers.

Peer Review reports

Introduction

Combination antiretroviral therapy (cART) use among persons living with HIV (PLWH) has resulted in longer life expectancy and a shift in causes of morbidity and mortality from acquired immunodeficiency syndrome (AIDS)-defining conditions to chronic aging-related non-communicable diseases, such as cardiovascular disease (CVD) [1, 2]. Extensive evidence suggests that HIV infection in the current cART era is an independent risk factor for myocardial disease [3], including incident heart failure (HF) with both reduced (HFrEF) and preserved (HFpEF) ejection fraction [4,5,6] and sudden cardiac death [7, 8]. Factors contributing to this excess risk, however, remain poorly characterized.

Advanced HIV disease is associated with systolic dysfunction and dilated cardiomyopathy, but with the advent of effective cART came a transition from this phenotype to one characterized more by subclinical differences in cardiac structure and diastolic abnormalities which may ultimately contribute to higher risk of incident clinical HFpEF observed in the current treatment era [9, 10]. Among a contemporary population of cART-treated PLWH compared to persons living without HIV (PLWOH) in the same setting, we recently reported a higher left atrial volume index (LAVI) and myocardial extracellular volume (ECV) fraction—a measure of fibrosis, edema, or inflammation—using cardiac magnetic resonance (CMR) imaging [11], both of which may presage diastolic dysfunction and HFpEF. No other cardiac structural or functional characteristics differed significantly by HIV serostatus within that study population, and similar associations between HIV serostatus and both measures of myocardial fibrosis and diastolic abnormalities have been described in the United States, Europe, and Africa [12,13,14,15,16,17,18].

Contemporary data are needed to understand the pathogenesis of HIV-associated myocardial injury and remodeling. Several biomarkers have been studied for associations with cardiac structure and function among the general population, including those of inflammation, cardiac stress and injury, and extracellular matrix remodeling and fibrosis. However, these relationships have been understudied in the context of HIV infection, where they may differ due to immune dysregulation and chronically elevated systemic inflammation that persists even in the context of cART.

Inflammation is a hallmark of chronic HIV infection and is intimately involved in the pathology of tissue remodeling and fibrosis. When inflammatory responses to tissue insult are imbalanced, further tissue damage can occur via prolonged insult by the original injurious stimuli or persistent activation of inflammatory pathways [19]. Epidemiologic evidence also suggests higher circulating levels of inflammation and immune activation markers are associated with fibrosis across organ systems, including within the myocardium [16, 20, 21]. However, in the context of cART-treated HIV infection, the characterization of inflammatory markers that may contribute to myocardial injury and remodeling remains limited.

The objective of this study was to evaluate the cross-sectional relationships of biomarkers of inflammation, ventricular and cellular stress, tissue remodeling, and tissue injury with previously identified HIV-associated subclinical alterations in cardiac structure. We studied PLWH and PLWOH who were enrolled in one of three established cohort studies—the Multicenter AIDS Cohort Study (MACS), the Women’s Interagency HIV Study (WIHS), and the AIDS Linked to the IntraVenous Experience (ALIVE) study—and we focused our analyses to biomarkers and the CMR structural metrics of LAVI and ECV, which were associated with HIV serostatus within this study population.

Methods

Study population

Active participants of MACS, WIHS, and ALIVE—each described in detail previously—were recruited and consented into the current study for CMR imaging under identical, standardized protocols from 2015 to 2018 [11]. Briefly, the MACS is a prospective cohort study of men who have sex with men living with and without HIV infection across four cities in the United States; the WIHS is a prospective cohort of women living with and without HIV across ten US cities; and the ALIVE study is a community-based prospective cohort of persons with a history of injection drug use enrolled in Baltimore, MD. By nature of the parent cohort designs, sex at birth and substance use differed across cohorts—i.e., MACS participants were 100% male, WIHS participants were 100% female, and ALIVE participants had higher reported usage of opioids and stimulants. As previously reported, there were differences in other major factors across cohorts, such as race/ethnicity, with 47%, 5%, and 3% self-reported white participants in MACS, WIHS, and ALIVE, respectively [11]. Distributions of age, HIV seropositivity, and HIV-related factors such as cART use were similar across parent cohorts. Distributions of most important characteristics were similar between PWOH and PLWH—including age, sex, race/ethnicity, and traditional cardiovascular risk factors [11].

Participants of the current study were aged 40–70 years and actively enrolled in MACS, WIHS, or ALIVE at Chicago, Baltimore, or Washington, DC study sites. Exclusion criteria were any contraindications to CMR or contrast administration, including estimated glomerular filtration rate (eGFR) < 45 mL/min/1.73 m2, weight > 350 pounds, known claustrophobia, or known gadolinium contrast allergy.

Clinical data

Under protocols of each of the included parent cohorts, participants underwent structured semi-annual study visits, which included standardized interviews, physical examinations, and plasma collection and storage. Data collected include demographics, education, smoking status, systolic and diastolic blood pressures, body mass index (BMI), fasting glucose; total, low-density lipoprotein (LDL), and high-density lipoprotein (HDL) cholesterol levels; serum creatinine, eGFR, hematocrit, and hepatitis C viral (HCV) infection status. Substance use variables were derived from data collected over the 5 years preceding CMR; any reported substance use during that time period is reported as use, and pack-years were calculated over this 5 year time period, not lifetime cumulative. In PLWH, measures of HIV disease activity included plasma HIV RNA concentrations (quantified down to 50 copies/mL, Roche ultrasensitive assay), current and nadir CD4 + T cell counts/µL, history of AIDS-defining malignancy or opportunistic infection, and use of cART.

Participants enrolled in the current study underwent a single visit between March 2015 and February 2018 where CMR was performed and a structured interview was administered, collecting further data on prescribed medications, history of cardiovascular disease, and substance use—including alcohol, marijuana, opiates, stimulants, prescribed or recreational use of erectile dysfunction agents, and prescribed or recreational use of nitrates.

Cardiac magnetic resonance imaging

CMR was performed at Johns Hopkins Hospital (Baltimore, MD, USA) or Northwestern Memorial Hospital (Chicago, IL, USA) on 1.5T Siemens Avanto or Aera scanners (Erlangen, Germany) using a standardized protocol, described in detail previously and identical across these two sites [11]. Briefly, short- and long-axis cines (30 phases/cardiac cycle) were acquired using a steady-state free precession sequence; two-dimensional late gadolinium enhancement short- and long-axis images were acquired using an inversion-recovery fast gradient-echo sequence 15 min post-intravenous administration of 0.2 mmol/kg of gadobutrol contrast (Gadavist®, Bayer, Montville, NJ, USA); and T1 mapping was ascertained using a modified Look-Locker inversion (MOLLI) recovery sequence both before and after contrast administration. All CMR analyses were performed blinded to HIV serostatus and other participant characteristics using Segment v2.0 (http://segment.heiberg.se), Multimodality Tissue Tracking software (MTT; version 6.0, Toshiba, Japan), and MRmap version 1.2 (Charite University Medicine, Berlin, Germany).

LAVI and ECV fraction were selected as outcomes for the current analysis because they have been previously shown to differ by HIV serostatus in this study population [11]. Maximum LA volume was measured using the LA volume curve generated by the Simpson’s method from the four-chamber and two-chamber views and indexed by body surface area [22, 23]. Tracing of the LA borders excluded pulmonary veins and the left atrial appendage [22]. ECV fraction (%) was calculated as ECV = 100 × partition coefficient × [1 − hematocrit]), where the partition coefficient was derived from the slope of the linear relationship of 1/T1myocardium veruss 1/T1blood pre- and post-contrast [24].

Protein biomarkers

Blood was drawn from participants concurrent with CMR, and samples were processed for serum and plasma on the same day using standard protocols and stored at − 80 °C until assays were performed. Biomarkers of inflammation (interleukin-6 [IL-6], soluble cluster of differentiation 14 [sCD14], galectin-3), ventricular and cellular stress (N-terminal prohormone of brain natriuretic peptide [NT-proBNP], growth differentiation factor-15 [GDF-15]), tissue remodeling (tissue inhibitor of metalloproteinase-2 [TIMP-2], matrix metalloproteinase-2 [MMP-2]) and tissue injury (high-sensitivity troponin I [hsTnI]) were measured in serum and plasma at central laboratories. Serum levels of IL-6 were measured by electrochemiluminescence (Meso Scale Discovery V-PLEX), and the following markers were measured by enzyme-linked immunosorbent assay: sCD14 (R&D Systems Quantikine), GDF-15 (R&D Systems Quantikine), TIMP-2 (R&D Systems Quantikine), MMP-2 (R&D Systems Quantikine), and NT-proBNP (Abbott Architect i2000sR) in serum, and galectin-3 (Abbott Architect i2000sR) and hsTnI (Abbott Architect i2000sR) in plasma.

Statistics

Biomarkers were winsorized to their 97th percentiles to limit the effect of outliers in modeling procedures and standardized for interpretability. Cardiac phenotypes of interest were dichotomized at normative thresholds as previously described [11]. Specifically, elevated left atrial volume was defined as ≥ 40 mL/m2 following indexation by body surface area [25] and elevated ECV fraction was defined as ≥ 30% among women and ≥ 28% among men [26].

Participant characteristics, CMR imaging parameters (continuous and dichotomized LAVI and ECV fraction), and laboratory measures were summarized by median (interquartile range [IQR]) or proportion (count). The distributions of participant characteristics, including circulating biomarker levels and CMR parameters, were compared by HIV serostatus using the Wilcoxon rank-sum test for continuous variables and Pearson’s χ2 test for categorical variables. In order to minimize multiple testing, only biomarkers that were associated with HIV serostatus were evaluated in subsequent analyses.

Among PLWH, analyses were performed using multivariable linear regression to estimate the associations between measures of HIV disease severity and biomarkers that differed by HIV serostatus (identified in the previous analytic step), adjusting for age, sex, race/ethnicity, education, pack-years of smoking over the 5 years preceding CMR, and hazardous alcohol use defined by an Alcohol Use Disorders Identification Test (AUDIT) score > 8 at the time of CMR. For these analyses, biomarker responses were modeled on the natural log scale. Exponentiated β coefficients are therefore the ratios of the geometric mean biomarker concentrations among those exposed versus unexposed to the HIV clinical factor.

The cross-sectional associations between HIV-associated biomarkers (identified in the previous analytic step) and HIV-associated cardiac phenotypes (high LAVI and high ECV fraction) were estimated per standard deviation increment in biomarker concentration using multivariable logistic regression. These regression models were adjusted for HIV serostatus, age, sex, race/ethnicity, education level, and traditional cardiovascular risk factors—specifically history of cardiovascular disease, average continuous systolic blood pressure measured over 5 years preceding CMR, antihypertensive medication, dyslipidemia (use of lipid-lowering medication, fasting total cholesterol level ≥ 200 mg/dL, LDL-cholesterol level ≥ 130 mg/dL, HDL-cholesterol level < 40 mg/dL, or serum triglyceride level ≥ 150 mg/dL), diabetes (use of hypoglycemic medication or fasting serum glucose levels ≥ 126 mg/dL at the cohort study visit closest to the time of the CMR study; hemoglobin A1C level < 6.5% was used to exclude diabetes if a participant was not using hypoglycemic medication and fasting glucose levels were unavailable), pack-years of smoking over 5 years preceding CMR, and hazardous alcohol use at the time of CMR. We selected this model based on hypothesized confounders and did not adjust for parent cohort due to collinearity with sex, i.e., WIHS enrolled only women and MACS enrolled only men.

To assess heterogeneity of the associations between biomarkers and outcomes by HIV serostatus, we stratified analyses by HIV serostatus and tested a multiplicative interaction term for each biomarker (HIV × biomarker) in separate regression models. The stratified models included the same covariates as in the unstratified analyses, with the exception of cardiovascular disease history and hypertension medication due to small cell counts. We also assessed whether inclusion of biomarkers attenuated the association between HIV serostatus and either high ECV or high LAVI by comparing HIV effect estimates from multivariable models with and without individual outcome-associated biomarkers included as covariates. Finally, post-hoc sensitivity analyses were performed to evaluate the impact of HCV infection and additional recreational drug use covariates—specifically marijuana, opioid, stimulant, erectile dysfunction drug, and nitrate use. All analyses were conducted using Stata 16.1 and SAS 9.4 with a type I error rate of 0.05.

Results

A total of 468 persons were initially enrolled across the three cohorts, of whom 381 had complete covariate data and either LAVI (n = 380) or ECV fraction (n = 319) acquired on CMR (Additional file 1: Figure S1). Demographic and clinical characteristics are presented in Table 1. Overall median age (IQR) was 55.1 (51.2, 58.4) years, 28% were female, 69% were Black, and 46% were current smokers. Sixty-two percent were PLWH, 88% of whom were receiving cART, and 72% of whom were virally suppressed. On average, PLWH had significantly higher levels of circulating sCD14 (p = < 0.001), GDF-15 (p =  < 0.001), and NT-proBNP (p = 0.03) compared to PLWOH; whereas there was no statistically significant difference by HIV serostatus in IL-6 (p = 0.84), galectin-3 (p = 0.13), TIMP-2 (p = 0.93), MMP-2 (p = 0.59), or hsTnI (p = 0.31) (Table 2). Only sCD14, GDF-15, and NT-proBNP were evaluated in subsequent analyses.

Table 1 Participant characteristics by HIV serostatus (n = 381)
Table 2 Circulating biomarkers of inflammation, fibrosis, and myocyte stretch stratified by HIV serostatus (n = 381)

Among PLWH, some measures of greater HIV disease severity were associated with higher circulating concentrations of sCD14, GDF-15 and NT-proBNP, independent of demographics and substance use (Additional file 1: Table S1). Associations were observed between CD4 + cell count < 500 cells/µL (yes vs. no) and plasma concentration of sCD14 (mean percent difference in sCD14: 9% higher [95% CI: 2–16], p = 0.01; GDF-15: 15% higher [2–31], p = 0.03; and NT-proBNP: 30% higher [2–65], p = 0.03). Higher NT-proBNP was also associated with detectable HIV viral load (30% higher [1–68]; p = 0.05).

Figure 1 presents adjusted estimates of the association between biomarkers that differed by HIV serostatus in this study population—sCD14, NT-proBNP, and GDF-15—and high LAVI and ECV fraction [11], overall and by HIV serostatus. Among all participants, NT-proBNP was associated with elevated LAVI (odds ratio [OR]: 1.79 per SD increment in NT-proBNP [95% CI: 1.31–2.44]; p < 0.001), following adjustment for age, sex at birth, race/ethnicity, education, pack-years of smoking in 5 years preceding CMR, hazardous alcohol use, dyslipidemia, systolic blood pressure, blood pressure-lowering therapy, diabetes, history of CVD, and HIV infection. No HIV × biomarker interaction term was statistically significant, although subgroup estimates were imprecise due to rare outcomes and relatively small sample sizes (e.g., only 8 cases with elevated LAVI among those without HIV). Other associations between HIV-associated biomarkers and HIV-associated cardiac structural characteristics were either small or imprecise, both overall and within subgroups defined by HIV serostatus.

Fig. 1
figure 1

Adjusted associations between biomarkers of inflammation, fibrosis, and myocyte stretch that differed by HIV serostatus and cardiac structural characteristics (ECV and LAVI), overall and by HIV serostatus. Odds ratio and 95% confidence interval for high left atrial volume index (≥ 40 mL/m2) and high extracellular volume fraction (≥ 30% among women and ≥ 28% among men) are reported per standard deviation increment in biomarker, estimated using logistic regression adjusting for HIV serostatus, age, sex, race/ethnicity, education level, history of cardiovascular disease, systolic blood pressure, blood pressure-lowering therapy, dyslipidemia, diabetes, pack-years of smoking in 5 years preceding CMR, and hazardous alcohol use in 5 years preceding CMR. Null hypothesis is that odds ratio = 1. Interaction p-value is for a multiplicative interaction parameter, biomarker × HIV serostatus, used to assess effect measure modification. ECV = extracellular volume; LAVI = left atrial volume index; PLWH = persons living with HIV; PLWOH = persons living without HIV; sCD14 = soluble cluster of differentiation 14; GDF-15 = growth differentiation factor 15; NT-proBNP = N-terminal prohormone of brain natriuretic peptide

Adding NT-proBNP levels to adjusted models estimating the association between HIV and LAVI attenuated the magnitude of the HIV effect estimate slightly but did not alter the statistical significance of the association (OR without adjustment for NT-proBNP: 2.85 [95% CI: 1.26–6.45], p = 0.01; OR with this adjustment: 2.54 [95% CI: 1.11–5.81], p = 0.03; 11% decrease).

In sensitivity analyses, parsimonious models excluding non-significant covariates yielded biomarker parameter estimates with consistent magnitude and statistical significance with those in our primary models. Further adjustment for HCV infection and recreational drug use also yielded negligible differences in biomarker parameter estimates, as did further adjustment for detectable HIV viral load in subgroup analyses among PLWH.

Discussion

Several lines of evidence now suggest PLWH in the current cART era have a higher prevalence of myocardial fibrosis, left atrial dilation, and diastolic abnormalities compared to PLWOH, including among participants within the current study [9,10,11,12,13,14,15,16,17,18, 27]. In this population of treated PLWH and PLWOH in the United States, positive HIV serostatus was associated with higher circulating levels of sCD14, GDF-15, and NT-proBNP, consistent with prior literature. When evaluating associations between levels of these three biomarkers and high LAVI or high ECV, there was evidence only of a relationship between NT-proBNP and LAVI, following adjustment for demographics and traditional CVD risk factors. Further, there was no evidence of modification of any effect estimates by HIV serostatus, assessed by HIV × biomarker interaction terms.

Contributors to excess risk of myocardial disease among PLWH are undoubtedly multifactorial, but immune dysfunction and persistent immune activation have been consistently implicated, even in the current cART era [28,29,30,31,32,33,34]. The interplay between inflammation, cardiac fibrosis, and cardiac stress is well-appreciated [19, 35], but remains poorly understood in HIV infection, where such relationships may be augmented or differ from those among the general population.

Higher circulating levels of sCD14, GDF-15, and NT-proBNP by HIV serostatus in this study are consistent with previous studies comparing cART-treated PLWH with PLWOH [16, 36,37,38,39]. CD14 is a pattern recognition receptor for ligands such as lipopolysaccharide [40]. It is integral to the activation of monocytes and macrophages and is involved in a variety of cellular processes, including host–pathogen interactions and cell differentiation [40]. A higher circulating level of sCD14 is a strong predictor of cardiovascular disease and mortality among both PLWOH and PLWH [41,42,43]. Consistent with our findings in both men and women, however, sCD14 was not associated with measures of myocardial fibrosis or diastolic dysfunction in a recent study among women living with well-controlled HIV in the United States [16]. The discrepant results in associations of sCD14 observed with clinical events but not subclinical myocardial disease or diastolic dysfunction may be due in part to difference in disease severity and potential differences in the role of monocyte and macrophage activation in atherosclerotic versus hypertensive cardiovascular pathologies [44]. For example, classical CD14high monocytes are progenitors of M1-type pro-inflammatory and phagocytic macrophages directly involved in atherosclerotic plaque formation, while non-classical CD14low monocytes are progenitors of M2-type macrophages, which may be pro-fibrotic in the context of chronic hemodynamic stress that can contribute to HF.

GDF-15 is a member of the transforming growth factor β superfamily involved in inflammation and cellular injury [45]. Elevated levels of GDF-15 have been strongly associated with all-cause mortality, incident HF, and poorer HF prognosis among the general population [46,47,48,49]. Higher GDF-15 has also been associated with subclinical cardiac structural and functional abnormalities among cART-treated PLWH—including higher LV mass index and lower LV ejection fraction—although conclusions on its association with diastolic dysfunction in particular have been inconsistent between study populations [38, 39]. To our knowledge, the association between GDF-15 and ECV fraction among PLWH has not previously been reported.

NT-proBNP is a powerful clinical biomarker used in diagnosis of HF, secreted in response to myocyte stretch due to increased ventricular preload, and elevated with both systolic and diastolic dysfunction [50]. Among PLWOH, elevated NT-proBNP is highly predictive of not only incident cardiovascular disease and mortality [51] but also subclinical cardiac remodeling, including myocardial fibrosis measured by ECV fraction [52, 53] and both cross-sectional and longitudinal increases in LAVI [54]. Among cART-treated PLWH, higher NT-proBNP has been cross-sectionally associated with higher left ventricular mass index, ECV fraction, and measures of both systolic and diastolic dysfunction [15, 39]. Results of the present study of LAVI are consistent with this literature but are not for ECV fraction, which may reflect lower absolute values and a narrower range of ECV in our study. The mild (11%) attenuation of the association between HIV infection and LAVI with addition of NT-proBNP as a model covariate suggests it may in part reflect HIV-associated differences in LAVI, but other HIV-related factors remain unmeasured and warrant further investigation.

We acknowledge limitations to this study. First, analyses were cross-sectional, so temporality and causation cannot be assessed. Second, sample size was relatively small, limiting our ability to detect small to moderate effect sizes or draw conclusions on effect measure modification by HIV serostatus. This is especially true for analyses of high LAVI, which was rare in our dataset. There are limitations inherent to the evaluated biomarkers, as well, specifically a lack of pathologic specificity. Even NT-proBNP, a well-established clinical biomarker used to rule out HF diagnosis, cannot reliably discriminate CVD pathologies [55] and may be elevated in extracardiac conditions such as renal insufficiency or systemic inflammation [56]. Interquartile ranges of some evaluated proteins were also relatively narrow, limiting prediction power. Additionally, while the distributions of demographic and key clinical characteristics among the participants in the current study are similar to that of the larger cohort previously published, we cannot rule out the possibility of selection bias due to analytic exclusions. Finally, multiple models were fit in this study, which may have led to type I errors.

In conclusion, our findings support prior literature establishing PLWH in the current cART era continue to have elevated levels of inflammation as well as markers of fibrosis and myocyte stress compared to PLWOH, specifically as measured by sCD14, GDF-15, and NT-proBNP. However, apart from an association between NT-proBNP and LAVI, the evaluated biomarkers do not well explain observed differences in LAVI and myocardial tissue abnormality by HIV serostatus. These results suggest mechanistic pathways other than those captured by the studied biomarkers should be investigated in future studies on excess risk of cardiovascular disease among contemporary cohorts of PLWH.

Availability of data and materials

The datasets analyzed in the current study are available from the corresponding author on reasonable request.

Abbreviations

AIDS:

Acquired immunodeficiency syndrome

ALIVE:

AIDS linked to the intra venous experience

AUDIT:

Alcohol use disorders identification test

BMI:

Body mass index

cART:

Combination antiretroviral therapy

CMR:

Cardiac magnetic resonance

CVD:

Cardiovascular disease

ECV:

Extracellular volume

eGFR:

Estimated glomerular filtration rate

GDF-15:

Growth differentiation factor-15

HCV:

Hepatitis C viral

HDL:

High-density lipoprotein

HF:

Heart failure

HFpEF:

Heart failure with preserved ejection fraction

HFrEF:

Heart failure with reduced ejection fraction

hsTnI:

High-sensitivity troponin I

IL-6:

Interleukin-6

IQR:

Interquartile range

LAVI:

Left atrial volume index

LDL:

Low-density lipoprotein

MACS:

Multicenter AIDS cohort study

MMP-2:

Matrix metalloproteinase-2

MOLLI:

Modified look-locker inversion sequence

NT-proBNP:

N-terminal prohormone of brain natriuretic peptide

OR:

Odds ratio

PLWH:

Persons living with HIV

PWOH:

Persons living without HIV

sCD14:

Soluble cluster of differentiation 14

TIMP-2:

Tissue inhibitor of metalloproteinase-2

WIHS:

Women’s interagency HIV study

References

  1. 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(2):214–20.

    Article  PubMed  Google Scholar 

  2. Shah ASV, Stelzle D, Lee KK, Beck EJ, Alam S, Clifford S, et al. Global burden of atherosclerotic cardiovascular disease in people living with HIV: systematic review and meta-analysis. Circulation. 2018;138(11):1100–12.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Feinstein MJ, Hsue PY, Benjamin LA, Bloomfield GS, Currier JS, Freiberg MS, et al. Characteristics, prevention, and management of cardiovascular disease in people living with HIV: a scientific statement from the American heart association. Circulation. 2019;140(2):e98–124.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Alonso A, Barnes AE, Guest JL, Shah A, Shao IY, Marconi V. HIV infection and incidence of cardiovascular diseases: an analysis of a large healthcare database. J Am Heart Assoc. 2019;8(14): e012241.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Freiberg MS, Chang CH, Skanderson M, Patterson OV, DuVall SL, Brandt CA, et al. Association between HIV infection and the risk of heart failure with reduced ejection fraction and preserved ejection fraction in the antiretroviral therapy era: results from the veterans aging cohort study. JAMA Cardiol. 2017;2(5):536–46.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Butt AA, Chang CC, Kuller L, Goetz MB, Leaf D, Rimland D, et al. Risk of heart failure with human immunodeficiency virus in the absence of prior diagnosis of coronary heart disease. Arch Intern Med. 2011;171(8):737–43.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Tseng ZH, Moffatt E, Kim A, Vittinghoff E, Ursell P, Connolly A, et al. Sudden cardiac death and myocardial fibrosis, determined by autopsy, in persons with HIV. N Engl J Med. 2021;384(24):2306–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Freiberg MS, Duncan MS, Alcorn C, Chang CH, Kundu S, Mumpuni A, et al. HIV infection and the risk of world health organization-defined sudden cardiac death. J Am Heart Assoc. 2021;10(18): e021268.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Erqou S, Lodebo BT, Masri A, Altibi AM, Echouffo-Tcheugui JB, Dzudie A, et al. Cardiac dysfunction among people living with HIV: a systematic review and meta-analysis. JACC Heart Fail. 2019;7(2):98–108.

    Article  PubMed  Google Scholar 

  10. deFilippi CR, Grinspoon SK. Myocardial dysfunction with contemporary management of HIV: prevalence, pathophysiology, and opportunities for prevention. JACC Heart Fail. 2019;7(2):109–11.

    Article  PubMed  Google Scholar 

  11. Wu KC, Haberlen SA, Plankey MW, Palella FJ, Piggott DA, Kirk GD, et al. Human immunodeficiency viral infection and differences in interstitial ventricular fibrosis and left atrial size. Eur Heart J Cardiovasc Imaging. 2021;22:888.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Ntusi N, O’Dwyer E, Dorrell L, Wainwright E, Piechnik S, Clutton G, et al. HIV-1-related cardiovascular disease is associated with chronic inflammation, frequent pericardial effusions, and probable myocardial edema. Circ Cardiovasc Imaging. 2016;9(3): e004430.

    Article  PubMed  Google Scholar 

  13. Luetkens JA, Doerner J, Schwarze-Zander C, Wasmuth JC, Boesecke C, Sprinkart AM, et al. Cardiac magnetic resonance reveals signs of subclinical myocardial inflammation in asymptomatic HIV-infected patients. Circ Cardiovasc Imaging. 2016;9(3): e004091.

    Article  PubMed  Google Scholar 

  14. Thiara DK, Liu CY, Raman F, Mangat S, Purdy JB, Duarte HA, et al. Abnormal myocardial function is related to myocardial steatosis and diffuse myocardial fibrosis in HIV-infected adults. J Infect Dis. 2015;212(10):1544–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Shuldiner SR, Wong LY, Peterson TE, Wolfson J, Jermy S, Saad H, et al. Myocardial fibrosis among antiretroviral therapy-treated persons with human immunodeficiency virus in South Africa. Open Forum Infect Dis. 2021;8(1):ofaa600.

    Article  PubMed  CAS  Google Scholar 

  16. Zanni MV, Awadalla M, Toribio M, Robinson J, Stone LA, Cagliero D, et al. Immune correlates of diffuse myocardial fibrosis and diastolic dysfunction among aging women with human immunodeficiency virus. J Infect Dis. 2020;221(8):1315–20.

    CAS  PubMed  Google Scholar 

  17. Hsue PY, Hunt PW, Ho JE, Farah HH, Schnell A, Hoh R, et al. Impact of HIV infection on diastolic function and left ventricular mass. Circ Heart Fail. 2010;3(1):132–9.

    Article  PubMed  Google Scholar 

  18. Doria de Vasconcellos H, Post WS, Ervin AM, Haberlen SA, Budoff M, Malvestutto C, et al. Associations between HIV Serostatus and cardiac structure and function evaluated by 2-dimensional echocardiography in the multicenter AIDS cohort study. J Am Heart Assoc. 2021;10(7):e019709.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Thomas TP, Grisanti LA. The dynamic interplay between cardiac inflammation and fibrosis. Front Physiol. 2020;11: 529075.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Marques MD, Nauffal V, Ambale-Venkatesh B, Vasconcellos HD, Wu C, Bahrami H, et al. Association between inflammatory markers and myocardial fibrosis. Hypertension. 2018;72(4):902–8.

    Article  CAS  PubMed  Google Scholar 

  21. Kuusisto J, Kärjä V, Sipola P, Kholová I, Peuhkurinen K, Jääskeläinen P, et al. Low-grade inflammation and the phenotypic expression of myocardial fibrosis in hypertrophic cardiomyopathy. Heart. 2012;98(13):1007–13.

    Article  PubMed  Google Scholar 

  22. Tao S, Ashikaga H, Ciuffo LA, Yoneyama K, Lima JAC, Frank TF, et al. Impaired left atrial function predicts inappropriate shocks in primary prevention implantable cardioverter-defibrillator candidates. J Cardiovasc Electrophysiol. 2017;28(7):796–805.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Eng J, McClelland RL, Gomes AS, Hundley WG, Cheng S, Wu CO, et al. Adverse left ventricular remodeling and age assessed with cardiac MR imaging: the multi-ethnic study of atherosclerosis. Radiology. 2016;278(3):714–22.

    Article  PubMed  Google Scholar 

  24. Moon JC, Messroghli DR, Kellman P, Piechnik SK, Robson MD, Ugander M, et al. Myocardial T1 mapping and extracellular volume quantification: a society for cardiovascular magnetic resonance (SCMR) and CMR working group of the European society of cardiology consensus statement. J Cardiovasc Magn Reson. 2013;15:92.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Kawel-Boehm N, Hetzel SJ, Ambale-Venkatesh B, Captur G, Francois CJ, Jerosch-Herold M, et al. Reference ranges (“normal values”) for cardiovascular magnetic resonance (CMR) in adults and children: 2020 update. J Cardiovasc Magn Reson. 2020;22(1):87.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Liu CY, Liu YC, Wu C, Armstrong A, Volpe GJ, van der Geest RJ, et al. Evaluation of age-related interstitial myocardial fibrosis with cardiac magnetic resonance contrast-enhanced T1 mapping: MESA (multi-ethnic study of atherosclerosis). J Am Coll Cardiol. 2013;62(14):1280–7.

    Article  PubMed  Google Scholar 

  27. Sinha A, Feinstein M. Epidemiology, pathophysiology, and prevention of heart failure in people with HIV. Prog Cardiovasc Dis. 2020;63(2):134–41.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Grund B, Baker JV, Deeks SG, Wolfson J, Wentworth D, Cozzi-Lepri A, et al. Relevance of interleukin-6 and D-dimer for serious non-AIDS morbidity and death among HIV-positive adults on suppressive antiretroviral therapy. PLoS ONE. 2016;11(5): e0155100.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Nordell AD, McKenna M, Borges Á, Duprez D, Neuhaus J, Neaton JD, et al. Severity of cardiovascular disease outcomes among patients with HIV is related to markers of inflammation and coagulation. J Am Heart Assoc. 2014;3(3): e000844.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Hsue PY, Scherzer R, Hunt PW, Schnell A, Bolger AF, Kalapus SC, et al. Carotid intima-media thickness progression in HIV-infected adults occurs preferentially at the carotid bifurcation and is predicted by inflammation. J Am Heart Assoc. 2012. https://doi.org/10.1161/JAHA.111.000422.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Bahrami H, Budoff M, Haberlen SA, Rezaeian P, Ketlogetswe K, Tracy R, et al. Inflammatory markers associated with subclinical coronary artery disease: the multicenter AIDS cohort study. J Am Heart Assoc. 2016. https://doi.org/10.1161/JAHA.116.003371.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Hanna DB, Lin J, Post WS, Hodis HN, Xue X, Anastos K, et al. Association of macrophage inflammation biomarkers with progression of subclinical carotid artery atherosclerosis in HIV-infected women and men. J Infect Dis. 2017;215(9):1352–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. 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 

  34. 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 

  35. Suthahar N, Meijers WC, Silljé HHW, de Boer RA. From inflammation to fibrosis-molecular and cellular mechanisms of myocardial tissue remodelling and perspectives on differential treatment opportunities. Curr Heart Fail Rep. 2017;14(4):235–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Wada NI, Jacobson LP, Margolick JB, Breen EC, Macatangay B, Penugonda S, et al. The effect of HAART-induced HIV suppression on circulating markers of inflammation and immune activation. AIDS. 2015;29(4):463–71.

    Article  CAS  PubMed  Google Scholar 

  37. Méndez-Lagares G, Romero-Sánchez MC, Ruiz-Mateos E, Genebat M, Ferrando-Martínez S, Muñoz-Fernández M, et al. Long-term suppressive combined antiretroviral treatment does not normalize the serum level of soluble CD14. J Infect Dis. 2013;207(8):1221–5.

    Article  PubMed  CAS  Google Scholar 

  38. Secemsky EA, Scherzer R, Nitta E, Wu AH, Lange DC, Deeks SG, et al. Novel biomarkers of cardiac stress, cardiovascular dysfunction, and outcomes in HIV-infected individuals. JACC Heart Fail. 2015;3(8):591–9.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Kipke J, Margevicius S, Kityo C, Mirembe G, Buggey J, Yun CH, et al. Sex, HIV status, and measures of cardiac stress and fibrosis in Uganda. J Am Heart Assoc. 2021;10(11): e018767.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wu Z, Zhang Z, Lei Z, Lei P. CD14: Biology and role in the pathogenesis of disease. Cytokine Growth Factor Rev. 2019;48:24–31.

    Article  CAS  PubMed  Google Scholar 

  41. 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 

  42. 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 

  43. Hunt PW, Sinclair E, Rodriguez B, Shive C, Clagett B, Funderburg N, et al. Gut epithelial barrier dysfunction and innate immune activation predict mortality in treated HIV infection. J Infect Dis. 2014;210(8):1228–38.

  44. van der Vorst EPC, Weber C. Novel features of monocytes and macrophages in cardiovascular biology and disease. Arterioscler Thromb Vasc Biol. 2019;39(2):e30–7.

    PubMed  Google Scholar 

  45. Wischhusen J, Melero I, Fridman WH. Growth/differentiation factor-15 (GDF-15): from biomarker to novel targetable immune checkpoint. Front Immunol. 2020;11:951.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wang TJ, Wollert KC, Larson MG, Coglianese E, McCabe EL, Cheng S, et al. Prognostic utility of novel biomarkers of cardiovascular stress: the Framingham heart study. Circulation. 2012;126(13):1596–604.

    Article  CAS  PubMed  Google Scholar 

  47. Kempf T, von Haehling S, Peter T, Allhoff T, Cicoira M, Doehner W, et al. Prognostic utility of growth differentiation factor-15 in patients with chronic heart failure. J Am Coll Cardiol. 2007;50(11):1054–60.

    Article  CAS  PubMed  Google Scholar 

  48. Anand IS, Kempf T, Rector TS, Tapken H, Allhoff T, Jantzen F, et al. Serial measurement of growth-differentiation factor-15 in heart failure: relation to disease severity and prognosis in the valsartan heart failure trial. Circulation. 2010;122(14):1387–95.

    Article  CAS  PubMed  Google Scholar 

  49. Xanthakis V, Larson MG, Wollert KC, Aragam J, Cheng S, Ho J, et al. Association of novel biomarkers of cardiovascular stress with left ventricular hypertrophy and dysfunction: implications for screening. J Am Heart Assoc. 2013;2(6): e000399.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Goetze JP, Bruneau BG, Ramos HR, Ogawa T, de Bold MK, de Bold AJ. Cardiac natriuretic peptides. Nat Rev Cardiol. 2020;17(11):698–717.

    Article  CAS  PubMed  Google Scholar 

  51. Alpert MA. Natriuretic peptides as predictors of cardiovascular events and all-cause mortality. J Am Coll Cardiol. 2021;77(5):572–4.

    Article  PubMed  Google Scholar 

  52. Liu CY, Heckbert SR, Lai S, Ambale-Venkatesh B, Ostovaneh MR, McClelland RL, et al. Association of elevated NT-proBNP with myocardial fibrosis in the multi-ethnic study of atherosclerosis (MESA). J Am Coll Cardiol. 2017;70(25):3102–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Yang EY, Khan MA, Graviss EA, Nguyen DT, Bhimaraj A, Nambi V, et al. Relationship of extracellular volume assessed on cardiac magnetic resonance and serum cardiac troponins and natriuretic peptides with heart failure outcomes. Sci Rep. 2019;9(1):20168.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Varadarajan V, Ambale-Venkatesh B, Hong SY, Habibi M, Ashikaga H, Wu CO, et al. Association of longitudinal changes in NT-proBNP with changes in left atrial volume and function: MESA. Am J Hypertens. 2021;34(6):626–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JGF, Coats AJS, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: the task force for the diagnosis and treatment of acute and chronic heart failure of the European society of cardiology (ESC) developed with the special contribution of the heart failure association (HFA) of the ESC. Eur Heart J. 2016;37(27):2129–200.

    Article  PubMed  Google Scholar 

  56. Tsai SH, Lin YY, Chu SJ, Hsu CW, Cheng SM. Interpretation and use of natriuretic peptides in non-congestive heart failure settings. Yonsei Med J. 2010;51(2):151–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all the study participants for their commitment and support of this research.

Funding

This study was funded by the United States National Institutes of Health (NIH)/National Heart, Lung, and Blood Institute (NHLBI): R01 HL126552 (KCW and WSP) and U01-DA036297 (ALIVE data collection). Data in this manuscript collected by MACS and WIHS, now the MACS/WIHS Combined Cohort Study (MWCCS), was supported by the NIH: U01-HL146201, U01-HL146193, U01-HL146240, and U01-HL146205 (MACS, WIHS data collection). Full MWCCS acknowledgement may be found at https://statepi.jhsph.edu/mwccs/acknowledgements/. TEP is supported by NIH/NHLBI Award Number T32 HL007227. TTB is supported in part by K24 AI120834. Architect i2000sR assays for galectin-3, high sensitivity troponin I, and N-terminal prohormone of brain natriuretic peptide were provided by Abbott under a research agreement. This work was also supported in part by a generous grant from Dr. Nancy Grasmick.

Author information

Authors and Affiliations

Authors

Contributions

Study conception and/or design: KCW, WSP. Data acquisition: KCW, WSP, MWP, FJP, DAP, JBM, TTB. Data analysis and presentation: CL, SAH, TEP, FB. Interpretation: SAH, TEP, KCW, WSP, CL. Manuscript preparation and revision: TEP (major), KCW, WSP, SAH, CL, JBM, TTB, MWP, FJP, DAP, FB. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Tess E. Peterson.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the Institutional Review Board of the Johns Hopkins University School of Medicine and was conducted in accordance with the declaration of Helsinki. Written informed consent was obtained from all subjects.

Consent for publication

Not Applicable.

Competing interests

TTB has served as a consultant to Janssen, ViiV Healthcare, Gilead Sciences, Merck, and Theratechnologies. Other 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

: Fig. S1. Study participant flow diagram. Table S1. Adjusted associations between HIV clinical characteristics and biomarkers of inflammation, fibrosis, and myocyte stretch among persons living with HIV (n = 235).

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

Peterson, T.E., Landon, C., Haberlen, S.A. et al. Circulating biomarker correlates of left atrial size and myocardial extracellular volume fraction among persons living with and without HIV. BMC Cardiovasc Disord 22, 393 (2022). https://doi.org/10.1186/s12872-022-02835-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12872-022-02835-y

Keywords