1
|
Stephens VR, Ameli S, Major AS, Wanjalla CN. Mouse Models of HIV-Associated Atherosclerosis. Int J Mol Sci 2025; 26:3417. [PMID: 40244289 PMCID: PMC11989901 DOI: 10.3390/ijms26073417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide. Several factors are implicated in the pathogenesis of CVD, and efforts have been made to reduce traditional risks, yet CVD remains a complex burden. Notably, people living with HIV (PLWH) are twice as likely to develop CVD compared to persons without HIV (PWoH). Intensive statin therapy, the first-line treatment to prevent cardiovascular events, is effective at reducing morbidity and mortality. However, statin therapy has not reduced the overall prevalence of CVD. Despite antiretroviral therapy (ART), and new guidelines for statin use, PLWH have persistent elevation of inflammatory markers, which is suggested to be a bigger driver of future cardiovascular events than low-density lipoprotein. Herein, we have summarized the development of atherosclerosis and highlighted mouse models of atherosclerosis in the presence and absence of HIV. Since most mouse strains have several mechanisms that are atheroprotective, researchers have developed mouse models to study CVD using dietary and genetic manipulations. In evaluating the current methodologies for studying HIV-associated atherosclerosis, we have detailed the benefits of integrating multi-omics analyses, genetic manipulations, and immune cell profiling within mouse models. These advanced approaches significantly enhance our capacity to address critical gaps in understanding the immune mechanisms driving CVD, including in the context of HIV.
Collapse
Affiliation(s)
- Victoria R. Stephens
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sharareh Ameli
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Amy S. Major
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Tennessee Valley Health System, Department of Veterans Affairs, Nashville, TN 37212, USA
| | - Celestine N. Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (S.A.); (A.S.M.)
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
2
|
Muccini C, Bottanelli M, Castagna A, Spagnuolo V. Cardiometabolic complications in children and adolescents with HIV on antiretroviral therapy. Expert Opin Drug Metab Toxicol 2024; 20:893-905. [PMID: 39210779 DOI: 10.1080/17425255.2024.2395562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION The course of HIV infection has changed radically with the introduction of antiretroviral therapy (ART), which has significantly reduced mortality and improved quality of life. However, antiretroviral drugs can cause adverse effects, including cardiometabolic complications and diseases, which are among the most common. Compared to the adult population, there are fewer studies in the pediatric population on treatment-related complications. The purpose of this review is to provide an update on the literature regarding cardiometabolic complications and diseases in children and adolescents with HIV. AREAS COVERED A comprehensive literature review was conducted using PubMed and related bibliographies to provide an overview of the current knowledge of metabolic complications (dyslipidemia, insulin resistance, lipodystrophy, weight gain and liver complications) and diseases (prediabetes/diabetes and cardiovascular diseases) associated with ART in children and adolescents with HIV. EXPERT OPINION Metabolic complications are conditions that need to be closely monitored in children and adolescents with HIV, as they increase the risk of early development of non-communicable diseases, such as cardiovascular disease. Key areas for improvement include ensuring access to treatment, reducing side effects and improving diagnostic capabilities. Overcoming existing challenges will require collaborative efforts across disciplines, advances in technology, and targeted interventions to address socioeconomic disparities.
Collapse
Affiliation(s)
- Camilla Muccini
- Department of Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Antonella Castagna
- Department of Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Vincenzo Spagnuolo
- Department of Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
3
|
van der Post J, Guerra TEJ, van den Hof M, Vaz FM, Pajkrt D, van Genderen JG. Plasma Lipidomic Profiles in cART-Treated Adolescents with Perinatally Acquired HIV Compared to Matched Controls. Viruses 2024; 16:580. [PMID: 38675922 PMCID: PMC11053976 DOI: 10.3390/v16040580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Children with perinatally acquired human immunodeficiency virus (PHIV) are growing into adulthood with HIV and treatment-associated comorbidities, such as dyslipidemia and insulin resistance. HIV is identified as independent risk factor for cardiovascular disease (CVD). The hypothesis behind increased CVD risk associated with HIV includes vascular inflammation, dyslipidemia and combination antiretroviral therapy (cART) metabolomic toxicity. To investigate differences in lipid profiles and pathophysiological mechanisms of CVD risk in adolescents with PHIV, we compared the plasma lipidome of PHIV adolescents and HIV-negative controls. We additionally investigated the influence of current cART regimens and increased lipoprotein(a) (Lp(a)) levels on the plasma lipidome. We included 20 PHIV-infected adolescents and 20 HIV-negative controls matched for age, sex, ethnic origin and socio-economic status. Plasma lipidome was measured using Thermo Scientific Ultimate 3000 binary high-performance liquid chromatography (HPLC)-mass spectrometry. We evaluated the plasma lipidome in PHIV adolescents using different cART regimens (including those known to be associated with lipid alterations). The median age was 17.5 years (15.5-20.7) and 16.5 years (15.7-19.8) for PHIV adolescents and controls, respectively. Of PHIV adolescents, 45% used a non-nucleotide reverse transcriptase inhibitor (NNRTI)-based (25%) or protease inhibitor (PI)-based (20%) cART regimen. In this pilot study, we observed no significant differences between lipidomic profiles between PHIV adolescents and controls. We observed no differences in the plasma lipidome in participants with increased versus normal Lp(a) levels. Different cART regimens appear to influence chain length differences in the plasma lipidome of PHIV adolescents; however, the significance and causality of this observation remains undetermined. Further research on the influence of cART on lipid composition could further identify these alterations.
Collapse
Affiliation(s)
- Julie van der Post
- Department of Pediatric Infectious Diseases, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 DD Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Thiara E. J. Guerra
- Department of Pediatric Infectious Diseases, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 DD Amsterdam, The Netherlands
| | - Malon van den Hof
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health, Ageing & Later Life, Health Behaviors and Chronic Diseases, Amsterdam, The Netherlands
| | - Frédéric M. Vaz
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children’s Hospital, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
- Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of Metabolism, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC, Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Dasja Pajkrt
- Department of Pediatric Infectious Diseases, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 DD Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
- Amsterdam Infectious Diseases and Immunology Research Institute, Amsterdam, The Netherlands
| | - Jason G. van Genderen
- Department of Pediatric Infectious Diseases, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, 1100 DD Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Wan Z, Su J, Zhu X, Liu X, Guo Y, Xiang D, Zhou X, Peng X, Tao R, Cao Q, Lang G, Huang Y, Zhu B. Distinct Lipidomic Profiles between People Living with HIV Treated with E/C/F/TAF or B/F/TAF: An Open-Label Prospective Cohort Study. Infect Dis Ther 2024; 13:727-744. [PMID: 38489119 PMCID: PMC11058159 DOI: 10.1007/s40121-024-00943-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/15/2024] [Indexed: 03/17/2024] Open
Abstract
INTRODUCTION Elvitegravir/cobicistat/emtricitabine/tenofovir alafenamide (E/C/F/TAF) has been increasingly replaced by bictegravir/emtricitabine/tenofovir alafenamide (B/F/TAF) in the treatment of human immunodeficiency virus (HIV) owing to its more favorable pharmacokinetics and fewer drug-drug interactions. However, the effect of this switch on plasma lipids and lipidomic profiles remains poorly characterized. METHODS HIV infected patients on an E/C/F/TAF regimen were recruited into the study and followed up every 12 weeks. Participants were divided into E/C/F/TAF and B/F/TAF groups depending on whether they were switched to B/F/TAF during follow-up. Clinical information and blood samples were collected at 0, 12, and 24 weeks, and lipidomic analysis was performed using liquid chromatography mass spectrometry. RESULTS No significant differences were observed between the groups at baseline. At week 24, patients switched to B/F/TAF had lower triglyceride [mmol/L; 1.23 (0.62) versus 2.03 (0.75), P = 0.001] and very low-density lipoprotein cholesterol [mmol/L; 0.64 (0.26) versus 0.84 (0.32), P = 0.037) compared with patients who continued E/C/F/TAF therapy. Small decrease from baseline in Framingham general cardiovascular risk score (FRS) was observed in the B/F/TAF arm [week (W) 0: 2.59 (1.57) versus W24: 2.18 (1.01), P = 0.043]. Lipidomic analysis indicated that E/C/F/TAF treatment increased the levels of several diglycerides (DGs), triacylglycerols (TAGs), and lyso-phosphatidylcholines (LPCs), whereas switching to B/F/TAF led to increased sphingolipids and glycerophospholipids. After adjusting for demographic and clinical parameters, only DG (16:0/18:2), DG (18:2/22:6), DG (18:3/18:2), DG (20:5/18:2), TAG (18:3/18:2/21:5), TAG (20:5/18:2/22:6), and LPC (22:6) were found to be significantly associated with FRS (regression coefficient of 0.17-6.02, P < 0.05). Most of these FRS associate lipid species were significantly elevated in individuals treated with E/C/F/TAF instead of individuals treated with B/F/TAF. CONCLUSION E/C/F/TAF promotes the accumulation of lipid species closely associated with cardiovascular disease (CVD) risk among people living with HIV, whereas B/F/TAF has a decreased impact on CVD-related lipid profile and is associated with lower CVD risk. A graphical abstract is available with this article. TRIAL REGISTRATION ClinicalTrials.gov; identifier, NCT06019273.
Collapse
Affiliation(s)
- Zhikai Wan
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Junwei Su
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Xueling Zhu
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Xiang Liu
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Yongzheng Guo
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Dairong Xiang
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Xiaotang Zhou
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Xiaorong Peng
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Ran Tao
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Qing Cao
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Guanjing Lang
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Ying Huang
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Biao Zhu
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Bowman ER, Wilson M, Riedl KM, MaWhinney S, Jankowski CM, Funderburg NT, Erlandson KM. Lipidome Alterations with Exercise Among People With and Without HIV: An Exploratory Study. AIDS Res Hum Retroviruses 2022; 38:544-551. [PMID: 35302400 PMCID: PMC9297322 DOI: 10.1089/aid.2021.0154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Age-related comorbidities and physical function impairments in aging people with HIV (PWH) can be improved through exercise interventions. The mechanisms underlying these improvements, including lipidomic changes, are unknown. Sedentary adults (50-75 years old) with or without HIV participated in supervised endurance/resistance exercise for 24 weeks. Plasma lipid concentrations (∼1,200 lipid species from 13 lipid classes) at baseline and week 24 were measured by mass spectrometry. Given multiple comparisons, unadjusted and Benjamini-Hochberg corrected p values are reported. Analyses are considered exploratory. Twenty-five PWH and 24 controls had paired samples at baseline and week 24. The change in total triacylglycerol (TAG) concentrations after exercise intervention differed between groups (unadj-p = 0.006, adj-p = 0.078) with concentrations increasing among controls, but not among PWH. Changes in concentrations of TAG species composed of long-chain fatty acids differed between groups (unadj-p < 0.04) with increases among controls, but not among PWH. Changes in total diacylglycerol (DAG) concentration from baseline to week 24 differed between groups (unadj-p = 0.03, adj-p = 0.2) with an increase in PWH and a nonsignificant decrease in controls. Baseline to week 24 changes in DAGs composed of palmitic acid (16:0), palmitoleic acid (16:1), and stearic acid (18:0) differed by serostatus (unadj-p = 0.009-0.03; adj-p 0.10-0.12), with nonsignificant increases and decreases in concentrations in PWH and controls, respectively. Concentrations of individual lysophosphatidylcholine (LPC) and ceramide (CER) species also differed by HIV serostatus (unadj-p < = 0.05). Although exploratory, the effects of exercise on the lipidome may differ among people with and without HIV, potentially due to underlying alterations in lipid processing and fatty acid oxidation in PWH. Clinical Trials NCT02404792.
Collapse
Affiliation(s)
- Emily R. Bowman
- College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Melissa Wilson
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kenneth M. Riedl
- College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Samantha MaWhinney
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Catherine M. Jankowski
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - Kristine M. Erlandson
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
6
|
Dirajlal-Fargo S, McComsey GA. Cardiometabolic Complications in Youth With Perinatally Acquired HIV in the Era of Antiretroviral Therapy. Curr HIV/AIDS Rep 2021; 18:424-435. [PMID: 34652624 DOI: 10.1007/s11904-021-00574-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2021] [Indexed: 01/13/2023]
Abstract
PURPOSE OF REVIEW Antiretroviral therapy (ART) scale-up has dramatically reduced rates of pediatric HIV mortality and morbidity. Children living with perinatally acquired HIV (PHIV) are now expected to live through adolescence and well into adulthood, such that adolescents now represent the largest growing population living with HIV. This review aims to discuss the prevalence and mechanisms for cardiometabolic comorbidities in the setting of newer ART regimens and the research gaps that remain. RECENT FINDINGS Data highlight the continued risks of subclinical cardiometabolic complications in PHIV in the setting of newer ART. Novel techniques in imaging and omics may help identify early cardiometabolic abnormalities in this young population and potentially identify early changes in the mechanistic pathways related to these changes. Further studies to determine risk and management strategies of the cardiometabolic effects in PHIV adolescents, beyond ART, are warranted. Focus should be on prevention of these complications in youth to avoid new epidemic of diabetes and cardiovascular disease when these youths become aging adults.
Collapse
Affiliation(s)
- Sahera Dirajlal-Fargo
- Rainbow Babies and Children's Hospital, Cleveland, OH, USA. .,University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA. .,Case Western Reserve University, Cleveland, OH, USA. .,University Hospitals Cleveland Medical Center, Case School of Medicine, Cleveland, OH, 44106, USA.
| | - Grace A McComsey
- Rainbow Babies and Children's Hospital, Cleveland, OH, USA.,University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA.,Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
7
|
Dirajlal-Fargo S, Sattar A, Yu J, Albar Z, Chaves FC, Riedl K, Kityo C, Bowman E, McComsey GA, Funderburg N. Lipidome association with vascular disease and inflammation in HIV+ Ugandan children. AIDS 2021; 35:1615-1623. [PMID: 33878042 PMCID: PMC8286331 DOI: 10.1097/qad.0000000000002923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE HIV infection and antiretroviral therapy (ART) have both been linked to dyslipidemia and increased cardiovascular disease (CVD). The relationships among the lipidome, immune activation, and subclinical vascular disease in children with perinatally acquired HIV (PHIV) have not been investigated. METHODS Serum lipid composition, including 13 lipid classes constituting 850 different lipid species were measured by direct infusion-tandem mass spectrometry in samples from 20 ART-treated PHIV and 20 age-matched and sex-matched HIV- Ugandan children. All participants were between 10 and 18 years of age with no other known active infections. PHIVs had HIV-1 RNA level 50 copies/ml or less. In addition, common carotid artery intima--media thickness (IMT), as well as plasma marker of systemic inflammation (hsCRP, IL6, sTNFRa I), monocyte activation (soluble CD14 and CD163), and T-cell activation (expression of CD38 and HLA-DR on CD4+ and CD8+) were evaluated. RESULTS Median age (Q1, Q3) of study participants was 13 years (11, 15), 37% were boys, 75% were on an NNRTI-based ART regimen. The concentrations of cholesterol ester, LCER, phosphatidylcholines, and sphingomyelin lipid classes were significantly increased in serum of PHIV compared with HIV (P≤0.04). Biomarkers associated with CVD risk including hsCRP, sCD163, and T-cell activation were directly correlated with lipid species in PHIV (P ≤ 0.04). Contents of free fatty acids including palmitic (16 : 0), stearic (18 : 0), and arachidic acid (20 : 0) were positively correlated with IMT in PHIV. CONCLUSION Serum lipidome is altered in young virally suppressed PHIV on ART. A direct association between inflammation and lipid species known to be associated with CVD was observed.
Collapse
Affiliation(s)
- Sahera Dirajlal-Fargo
- University Hospitals Cleveland Medical Center
- Rainbow Babies and Children's Hospital
- Case Western Reserve University, Cleveland
| | | | - Jiao Yu
- Case Western Reserve University, Cleveland
| | | | - Fabio C Chaves
- Department of Food Science and Technology, and the OSU Comprehensive Cancer Center, The Ohio State University, OH, USA
| | - Ken Riedl
- Department of Food Science and Technology, and the OSU Comprehensive Cancer Center, The Ohio State University, OH, USA
| | - Cissy Kityo
- Joint Clinical Research Centre, Kampala, Uganda
| | - Emily Bowman
- Ohio State University School of Health and Rehabilitation Sciences, Columbus, OH, USA
| | - Grace A McComsey
- University Hospitals Cleveland Medical Center
- Rainbow Babies and Children's Hospital
- Case Western Reserve University, Cleveland
| | - Nicholas Funderburg
- Ohio State University School of Health and Rehabilitation Sciences, Columbus, OH, USA
| |
Collapse
|
8
|
Chaudhary NS, Kind T, Willig AL, Saag MS, Shrestha S, Funderburg N, Wiener HW, Overton ET, Irvin MR. Changes in lipidomic profile by anti-retroviral treatment regimen: An ACTG 5257 ancillary study. Medicine (Baltimore) 2021; 100:e26588. [PMID: 34397689 PMCID: PMC8322553 DOI: 10.1097/md.0000000000026588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 06/21/2021] [Indexed: 02/07/2023] Open
Abstract
High cardiovascular disease risk in people living with HIV is partly attributed to antiretroviral therapy (ART). Lipid response to ART has been extensively studied, yet, little is known how small molecule lipids respond to Integrase inhibitor-based (INSTI-based) compared to Protease inhibitor-based (PI-based) ART regimens.Ancillary study to a phase 3, randomized, open-label trial [AIDS Clinical Trial Group A5257 Study] in treatment-naive HIV-infected patients randomized in a 1:1:1 ratio to receive ritonavir-boosted atazanavir (ATV/r), ritonavir-boosted darunavir (DRV/r) (both PI-based), or raltegravir with Tenofovir Disoproxil Fumarate-TDF plus emtricitabine (RAL, INSTI-based).We examined small molecule lipid response in a subcohort of 75 participants. Lipidomic assays of plasma samples collected pre- and post-ART treatment (48 weeks) were conducted using ultra-performance liquid chromatography coupled to time-of-flight mass spectrometry. The effect of ART regimens was regressed on lipid species response adjusting for the baseline covariates (lipids, age, sex, race, CD4 level, BMI, and smoking). Results were validated in the Centers for AIDS Research Network of Integrated Clinical Systems study (N = 16).Out of 417 annotated lipids, glycerophospholipids (P = .007) and sphingolipids (P = .028) had a higher response to ATV/r and DRV/r compared to RAL. The lysophosphatidylcholine (LPCs(16:1),(17:1),(20:3)) and phosphophatidylcholine species (PCs(40:7),(38:4)) had an opposite response to RAL versus ATV/r in the discovery and validation cohort. The INSTI-based regimen had an opposite response of ceramide species ((d38:1), (d42:2)), PCs((35:2), (38:4)), phosphatidylethanolamines (PEs(38:4), (38:6)), and sphingomyelin(SMd38:1) species compared with the PI-based regimens. There were no differences observed between 2 PI-based regimens.We observed differences in response of small molecule lipid species by ART regimens in treatment-naive people living with HIV.
Collapse
Affiliation(s)
- Ninad S. Chaudhary
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL
| | - Tobias Kind
- NIH West Coast Metabolomics Center, UC Davis Genome Center, University of California, Davis, CA
| | - Amanda L. Willig
- Division of Infectious Diseases, University of Alabama School of Medicine, Birmingham, AL
| | - Michael S. Saag
- Division of Infectious Diseases, University of Alabama School of Medicine, Birmingham, AL
| | - Sadeep Shrestha
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL
| | - Nicholas Funderburg
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, Columbus, OH
| | - Howard W. Wiener
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL
| | - E. Turner Overton
- Division of Infectious Diseases, University of Alabama School of Medicine, Birmingham, AL
| | - Marguerite R. Irvin
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
9
|
Bowman ER, Cameron CM, Richardson B, Kulkarni M, Gabriel J, Cichon MJ, Riedl KM, Mustafa Y, Cartwright M, Snyder B, Raman SV, Zidar DA, Koletar SL, Playford MP, Mehta NN, Sieg SF, Freeman ML, Lederman MM, Cameron MJ, Funderburg NT. Macrophage maturation from blood monocytes is altered in people with HIV, and is linked to serum lipid profiles and activation indices: A model for studying atherogenic mechanisms. PLoS Pathog 2020; 16:e1008869. [PMID: 33002093 PMCID: PMC7553323 DOI: 10.1371/journal.ppat.1008869] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/13/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022] Open
Abstract
People with HIV (PWH) are at increased risk for atherosclerotic cardiovascular disease (ASCVD). Proportions of vascular homing monocytes are enriched in PWH; however, little is known regarding monocyte-derived macrophages (MDMs) that may drive atherosclerosis in this population. We isolated PBMCs from people with and without HIV, and cultured these cells for 5 days in medium containing autologous serum to generate MDMs. Differential gene expression (DGE) analysis of MDMs from PWH identified broad alterations in innate immune signaling (IL-1β, TLR expression, PPAR βδ) and lipid processing (LXR/RXR, ACPP, SREBP1). Transcriptional changes aligned with the functional capabilities of these cells. Expression of activation markers and innate immune receptors (CD163, TLR4, and CD300e) was altered on MDMs from PWH, and these cells produced more TNFα, reactive oxygen species (ROS), and matrix metalloproteinases (MMPs) than did cells from people without HIV. MDMs from PWH also had greater lipid accumulation and uptake of oxidized LDL. PWH had increased serum levels of free fatty acids (FFAs) and ceramides, with enrichment of saturated FAs and a reduction in polyunsaturated FAs. Levels of lipid classes and species that are associated with CVD correlated with unique DGE signatures and altered metabolic pathway activation in MDMs from PWH. Here, we show that MDMs from PWH display a pro-atherogenic phenotype; they readily form foam cells, have altered transcriptional profiles, and produce mediators that likely contribute to accelerated ASCVD. People with HIV (PWH) are at greater risk for developing cardiovascular disease (CVD) than the general public, but the mechanisms underlying this increased risk are poorly understood. Macrophages play key roles in the pathogenesis of atherosclerosis, and are potential targets for therapeutic intervention. Here, we investigate phenotypic and functional abnormalities in monocyte-derived macrophages (MDMs) isolated from PWH that may drive CVD risk in this population. MDMs were differentiated in the presence of autologous serum, enabling us to explore the contributions of serum components (lipids, inflammatory cytokines, microbial products) as drivers of altered MDM function. We link serum levels of inflammatory biomarkers and CVD-associated lipid species to MDM activation. Our study provides new insight into drivers of pro-atherogenic MDM phenotype in PWH, and identifies directions for future study and potential intervention strategies to mitigate CVD risk.
Collapse
Affiliation(s)
- Emily R. Bowman
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| | - Cheryl M. Cameron
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Brian Richardson
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Manjusha Kulkarni
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, Columbus, Ohio, United States of America
| | - Janelle Gabriel
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, Columbus, Ohio, United States of America
| | - Morgan J. Cichon
- Department of Food Science & Technology and the Nutrient & Phytochemical Shared Resource, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | - Kenneth M. Riedl
- Department of Food Science & Technology and the Nutrient & Phytochemical Shared Resource, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | - Yousef Mustafa
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Michael Cartwright
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Brandon Snyder
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, Columbus, Ohio, United States of America
| | - Subha V. Raman
- Department of Internal Medicine, Division of Cardiovascular Medicine, Ohio State University, Columbus, Ohio, United States of America
| | - David A. Zidar
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
| | - Susan L. Koletar
- Department of Medicine, Division of Infectious Diseases, Ohio State University, Columbus, Ohio, United States of America
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Scott F. Sieg
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, Ohio, United States of America
| | - Michael L. Freeman
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, Ohio, United States of America
| | - Michael M. Lederman
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, Ohio, United States of America
| | - Mark J. Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Nicholas T. Funderburg
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
10
|
Lipidomics Reveals Reduced Inflammatory Lipid Species and Storage Lipids after Switching from EFV/FTC/TDF to RPV/FTC/TDF: A Randomized Open-Label Trial. J Clin Med 2020; 9:jcm9051246. [PMID: 32344934 PMCID: PMC7288166 DOI: 10.3390/jcm9051246] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/14/2020] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
HIV and antiretroviral therapy affect lipid metabolism. Lipidomics quantifies several individual species that are overlooked using conventional biochemical analyses, outperforming traditional risk equations. We aimed to compare the plasma lipidomic profile of HIV patients taking efavirenz (EFV) or rilpivirine (RPV). Patients ≥ 18 years old on EFV co-formulated with emtricitabine and tenofovir disoproxil fumarate (FTC/TDF) with HIV-RNA < 50 copies/mL for ≥6 months were randomized to continue EFV/FTC/TDF (n = 14) or switch to RPV/FTC/TDF (n =15). Lipidomic analyses conducted by mass spectrometry (MS) were performed at baseline and after 12 and 24 weeks. OWLiver® Care and OWLiver® tests were performed to estimate the presence of fatty liver disease (NAFLD). No significant differences (83% male, median age 44 years, 6 years receiving EFV/FTC/TDF, CD4+ count 740 cells/mm3, TC 207 [57 HDL-C/133 LDL-C] mg/dL, TG 117 mg/dL) were observed between the groups at baseline. Significant reductions in plasma lipids and lipoproteins but increased circulating bilirubin concentrations were observed in patients who switched to RPV/FTC/TDF. Patients on RPV/FTC/TDF showed a decrease in the global amount of storage lipids (-0.137 log2 [fold-change] EFV vs. 0.059 log2 [fold-change] RPV) but an increase in lysophosphatidylcholines (LPCs) and total steroids. Compared with EFV, RPV increased metabolites with anti-inflammatory properties and reduced the repository of specific lipotoxic lipids.
Collapse
|
11
|
Bowman E, Funderburg NT. Lipidome Abnormalities and Cardiovascular Disease Risk in HIV Infection. Curr HIV/AIDS Rep 2020; 16:214-223. [PMID: 30993515 DOI: 10.1007/s11904-019-00442-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Human immunodeficiency virus (HIV) infection and its treatment with antiretroviral therapy (ART) are associated with lipid abnormalities that may enhance cardiovascular disease risk (CVD). RECENT FINDINGS Chronic inflammation persists in HIV+ individuals, and complex relationships exist among lipids and inflammation, as immune activation may be both a cause and a consequence of lipid abnormalities in HIV infection. Advances in mass spectrometry-based techniques now allow for detailed measurements of individual lipid species; improved lipid measurement might better evaluate CVD risk compared with the prognostic value of traditional assessments. Lipidomic analyses have begun to characterize dynamic changes in lipid composition during HIV infection and following treatment with ART, and further investigation may identify novel lipid biomarkers predictive of adverse outcomes. Developing strategies to improve management of comorbidities in the HIV+ population is important, and statin therapy and lifestyle modifications, including diet and exercise, may help to improve lipid levels and mitigate CVD risk.
Collapse
Affiliation(s)
- Emily Bowman
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University College of Medicine, 453 W. 10th Ave. 535A Atwell Hall, Columbus, OH, 43210, USA
| | - Nicholas T Funderburg
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University College of Medicine, 453 W. 10th Ave. 535A Atwell Hall, Columbus, OH, 43210, USA.
| |
Collapse
|
12
|
Chai JC, Deik AA, Hua S, Wang T, Hanna DB, Xue X, Haberlen SA, Shah SJ, Suh Y, Lazar JM, Gustafson D, Hodis HN, Landay AL, Anastos K, Post WS, Kaplan RC, Clish CB, Qi Q. Association of Lipidomic Profiles With Progression of Carotid Artery Atherosclerosis in HIV Infection. JAMA Cardiol 2019; 4:1239-1249. [PMID: 31642867 PMCID: PMC6813587 DOI: 10.1001/jamacardio.2019.4025] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/18/2019] [Indexed: 01/02/2023]
Abstract
Importance Lipid metabolism disruption and excess risk of cardiovascular disease (CVD) have been observed in HIV-infected individuals, but the associations among HIV infection, plasma lipidome, and CVD risk have not been well understood. Objective To evaluate plasma lipidomic profiles and their associations with carotid artery atherosclerosis in individuals with HIV and individuals without HIV. Design, Setting, and Participants Prospective analysis in the Women's Interagency HIV Study and Multicenter AIDS Cohort Study during a 7-year follow-up (from 2004-2006 to 2011-2013) at multicenter HIV cohorts in the United States. The study included 737 participants aged 35 to 55 years (520 with HIV and 217 without HIV) without CVD or carotid artery plaque at baseline. Data were analyzed between April 2017 and July 2019. Exposures Two hundred eleven plasma lipid species. Main Outcomes and Measures Poisson regression was used to examine the associations of baseline lipid species with risk of plaque measured by repeated B-mode carotid artery ultrasonography imaging. Results Of the 737 included participants, 398 (54%) were women, 351 (48%) were African American (non-Hispanic), 156 of 737 (21%) were nonwhite Hispanic, and the mean (SD) age was 45 (6) years. After adjusting for demographic and behavioral factors, we identified 12 lipid species, representing independent signals for 10 lipid classes, associated with risk of plaque. Nine lipid species remained significant after further adjusting for conventional CVD risk factors, although many of them showed moderate to high association with conventional blood lipids (eg, total and low-density lipoprotein cholesterols and triglycerides). Cholesteryl ester (16:1) (risk ratio [RR] per standard deviation, 1.28; 95% CI, 1.08-1.52), ceramide (16:0) (RR, 1.29; 95% CI, 1.02-1.63), lysophosphatidylcholine (20:4) (RR, 1.28; 95% CI, 1.05-1.58), lysophosphatidylethanolamine (16:0) (RR, 1.28; 95% CI, 1.05-1.57), phosphatidylethanolamine (38:6) (RR, 1.33; 95% CI, 1.08-1.64), phosphatidylethanolamine-plasmalogen (36:2) (RR, 1.25; 95% CI, 1.04-1.52), phosphatidylserine-plasmalogen (36:3) (RR, 1.19; 95% CI, 1.00-1.43), and triacylglycerol (54:6) (RR, 1.26; 95% CI, 1.04-1.54) were associated with increased risk of plaque, while phosphatidylcholine (36:4) (RR, 0.65; 95% CI, 0.54-0.77) was associated with decreased risk of plaque. Most of these plaque-increased lipid species showed higher levels in individuals with HIV, particularly among individuals with HIV using antiretroviral therapy compared with individuals without HIV. Network analysis identified 9 lipid modules, and 2 modules composed of triacylglycerols and phosphatidylcholines with long and unsaturated acyl chains, respectively, showed the strongest associations with increased risk of plaque. Conclusions and Relevance This study identified multiple plasma lipid species associated with carotid artery atherosclerosis, and alterations in these lipid species might be associated with HIV infection and antiretroviral therapy. Our data suggest unfavorable associations of long-chain and unsaturated triacylglycerols and phosphatidylcholines with carotid artery plaque formation.
Collapse
Affiliation(s)
- Jin Choul Chai
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Amy A. Deik
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Simin Hua
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - David B. Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Sabina A. Haberlen
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland
| | - Sanjiv J. Shah
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Jason M. Lazar
- Department of Neurology, State University of New York-Downstate Medical Center, Brooklyn, New York
| | - Deborah Gustafson
- Department of Neurology, State University of New York-Downstate Medical Center, Brooklyn, New York
| | - Howard N. Hodis
- Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles
| | - Alan L. Landay
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, Illinois
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Wendy S. Post
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Clary B. Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
13
|
Mena A, Clavero E, Díaz-Díaz JL, Castro A. Similar plasma lipidomic profile in people living with HIV treated with a darunavir-based or an integrase inhibitor-based antiretroviral therapy. Sci Rep 2019; 9:17184. [PMID: 31748628 PMCID: PMC6868233 DOI: 10.1038/s41598-019-53761-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 11/05/2019] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular disease is an important cause of morbidity and mortality in people living with HIV (PLWH), who commonly experience lipid disturbances. The aim of this study was to determine whether the plasma lipidomic profile differs between PLWH receiving a darunavir-based ART and those receiving integrase inhibitor-based ART. This was a cross-sectional study of unselected patients for whom metabolomic analysis was performed using ultra-high-performance liquid chromatography coupled to mass spectrometry. Data for the two subgroups were compared by calculating the log2 of the fold change for each metabolite and then grouping these into the main lipid families. Sixty-two PLWH aged 49.3 ± 8.6 years (82% men) were included: 12 patients (19.4%) had hypertension, 8 (12.9%) had type 2 diabetes, 25 (41.0%) had dyslipidaemia and 9 (14.5%) were taking statins, without significant differences in all these variables between the two groups. Twenty-five (40.3%) received darunavir-based ART and 37 (59.7%) integrase inhibitor-based ART. Although the differences were not statistically significant, patients treated with darunavir-based ART had higher concentrations of total cholesterol (211 mg/dL vs 194 mg/dL), LDL-cholesterol (132 mg/dL vs 117 mg/dL) and triglycerides (155 mg/dL vs 122 mg/dL), and lower HDL-cholesterol concentration (50 mg/dL vs 52 mg/dL). The main lipid families and metabolites differed slightly between groups (log2-fold change; P-value): ceramides (-0.07; 0.49), phosphatidylinositols (-0.05; 0.63), diacylglycerols (0.10; 0.64), phosphatidylethanolamines (0.03; 0.78), triacylglycerols (0.27; 0.18) and lysophosphatidylethanolamines (0.03; 0.83). In the integrase inhibitor-based group, the use of tenofovir alafenamide fumarate significantly increases the majority of lipid fractions, when compared with tenofovir disoproxil fumarate. The lipidomic profile did not differ between PLWH treated with darunavir-based or integrase inhibitor-based ART. This was especially true for ceramides, which are involved in cardiovascular disease. Further studies are needed to study the impact of ART in lipidomic profile.
Collapse
Affiliation(s)
- Alvaro Mena
- Grupo de Virología Clínica, Instituto de Investigación Biomédica de A Coruña (INIBIC)-Complejo Hospitalario Universitario de A Coruña (CHUAC), Sergas. Universidad de A Coruña (UDC), A Coruña, Spain. .,Unidad de Enfermedades Infecciosas, Servicio de Medicina Interna, Complejo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain.
| | - Elvira Clavero
- Grupo de Virología Clínica, Instituto de Investigación Biomédica de A Coruña (INIBIC)-Complejo Hospitalario Universitario de A Coruña (CHUAC), Sergas. Universidad de A Coruña (UDC), A Coruña, Spain.,Servicio de Medicina Interna, Complejo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| | - José Luis Díaz-Díaz
- Servicio de Medicina Interna, Complejo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| | - Angeles Castro
- Grupo de Virología Clínica, Instituto de Investigación Biomédica de A Coruña (INIBIC)-Complejo Hospitalario Universitario de A Coruña (CHUAC), Sergas. Universidad de A Coruña (UDC), A Coruña, Spain.,Unidad de Enfermedades Infecciosas, Servicio de Medicina Interna, Complejo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, Spain
| |
Collapse
|
14
|
Bowman ER, Kulkarni M, Gabriel J, Cichon MJ, Riedl K, Belury MA, Lake JE, Richardson B, Cameron C, Cameron M, Koletar SL, Lederman MM, Sieg SF, Funderburg NT. Altered Lipidome Composition Is Related to Markers of Monocyte and Immune Activation in Antiretroviral Therapy Treated Human Immunodeficiency Virus (HIV) Infection and in Uninfected Persons. Front Immunol 2019; 10:785. [PMID: 31040846 PMCID: PMC6477036 DOI: 10.3389/fimmu.2019.00785] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/25/2019] [Indexed: 12/21/2022] Open
Abstract
Background: HIV infection and antiretroviral therapy (ART) have both been linked to dyslipidemia and increased cardiovascular disease (CVD) risk. Alterations in the composition of saturated (SaFA), monounsaturated (MUFA), and polyunsaturated (PUFA) fatty acids are related to inflammation and CVD progression in HIV-uninfected (HIV-) populations. The relationships among the lipidome and markers of monocyte and immune activation in HIV-infected (HIV+) individuals are not well understood. Methods: Concentrations of serum lipids and their fatty acid composition were measured by direct infusion-tandem mass spectrometry in samples from 20 ART-treated HIV+ individuals and 20 HIV- individuals. Results: HIV+ individuals had increased levels of free fatty acids (FFAs) with enrichment of SaFAs, including palmitic acid (16:0) and stearic acid (18:0), and these levels were directly associated with markers of monocyte (CD40, HLA-DR, TLR4, CD36) and serum inflammation (LBP, CRP). PUFA levels were reduced significantly in HIV+ individuals, and many individual PUFA species levels were inversely related to markers of monocyte activation, such as tissue factor, TLR4, CD69, and SR-A. Also in HIV+ individuals, the composition of lysophosphatidylcholine (LPC) was enriched for SaFAs; LPC species containing SaFAs were directly associated with IL-6 levels and monocyte activation. We similarly observed direct relationships between levels of SaFAs and inflammation in HIV uninfected individuals. Further, SaFA exposure altered monocyte subset phenotypes and inflammatory cytokine production in vitro. Conclusions: The lipidome is altered in ART-treated HIV infection, and may contribute to inflammation and CVD progression. Detailed lipidomic analyses may better assess CVD risk in both HIV+ and HIV- individuals than does traditional lipid profiling.
Collapse
Affiliation(s)
- Emily R Bowman
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH, United States
| | - Manjusha Kulkarni
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH, United States
| | - Janelle Gabriel
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH, United States
| | - Morgan J Cichon
- Personalized Food and Nutritional Metabolomics for Health Discovery Theme, The Ohio State University, Columbus, OH, United States
| | - Kenneth Riedl
- Personalized Food and Nutritional Metabolomics for Health Discovery Theme, The Ohio State University, Columbus, OH, United States
| | - Martha A Belury
- Department of Human Sciences, Ohio State University, Columbus, OH, United States
| | - Jordan E Lake
- Division of Infectious Diseases, Department of Medicine, The University of Texas Health Science Center, Houston, TX, United States
| | - Brian Richardson
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Cheryl Cameron
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
| | - Mark Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Susan L Koletar
- Division of Infectious Diseases, Department of Medicine, Ohio State University, Columbus, OH, United States
| | - Michael M Lederman
- Division of Infectious Diseases, Department of Internal Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH, United States
| | - Scott F Sieg
- Division of Infectious Diseases, Department of Internal Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH, United States
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH, United States
| |
Collapse
|
15
|
Tarancon-Diez L, Rodríguez-Gallego E, Rull A, Peraire J, Viladés C, Portilla I, Jimenez-Leon MR, Alba V, Herrero P, Leal M, Ruiz-Mateos E, Vidal F. Immunometabolism is a key factor for the persistent spontaneous elite control of HIV-1 infection. EBioMedicine 2019; 42:86-96. [PMID: 30879922 PMCID: PMC6491381 DOI: 10.1016/j.ebiom.2019.03.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Approximately 25% of elite controllers (ECs) lose their virological control by mechanisms that are only partially known. Recently, immunovirological and proteomic factors have been associated to the loss of spontaneous control. Our aim was to perform a metabolomic approach to identify the underlying mechanistic pathways and potential biomarkers associated with this loss of control. METHODS Plasma samples from EC who spontaneously lost virological control (Transient Controllers, TC, n = 8), at two and one year before the loss of control, were compared with a control group of EC who persistently maintained virological control during the same follow-up period (Persistent Controllers, PC, n = 8). The determination of metabolites and plasma lipids was performed by GC-qTOF and LC-qTOF using targeted and untargeted approaches. Metabolite levels were associated with the polyfunctionality of HIV-specific CD8+T-cell response. FINDINGS Our data suggest that, before the loss of control, TCs showed a specific circulating metabolomic profile characterized by aerobic glycolytic metabolism, deregulated mitochondrial function, oxidative stress and increased immunological activation. In addition, CD8+ T-cell polyfunctionality was strongly associated with metabolite levels. Finally, valine was the main differentiating factor between TCs and PCs. INTERPRETATION All these metabolomic differences should be considered not only as potential biomarkers but also as therapeutic targets in HIV infection. FUND: This work was supported by grants from Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Fondos FEDER; Red de Investigación en Sida, Gilead Fellowship program, Spanish Ministry of Education and Spanish Ministry of Economy and Competitiveness.
Collapse
Affiliation(s)
- Laura Tarancon-Diez
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| | - Esther Rodríguez-Gallego
- Hospital Universitari de Tarragona Joan XXIII, IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Anna Rull
- Hospital Universitari de Tarragona Joan XXIII, IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Joaquim Peraire
- Hospital Universitari de Tarragona Joan XXIII, IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Consuelo Viladés
- Hospital Universitari de Tarragona Joan XXIII, IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Irene Portilla
- Infectious Diseases, Instituto de Investigación Sanitaria y Biomédica de Alicante, ISABIAL - FISABIO, Hospital General Universitario de Alicante, Alicante, Spain
| | - María Reyes Jimenez-Leon
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| | - Verónica Alba
- Hospital Universitari de Tarragona Joan XXIII, IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Pol Herrero
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Reus, Spain
| | - Manuel Leal
- Servicio de Medicina Interna, Hospital Viamed Santa Ángela de la Cruz, Sevilla, Spain; Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío, Sevilla
| | - Ezequiel Ruiz-Mateos
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain.
| | - Francesc Vidal
- Hospital Universitari de Tarragona Joan XXIII, IISPV, Universitat Rovira i Virgili, Tarragona, Spain.
| |
Collapse
|
16
|
Persistent metabolic changes in HIV-infected patients during the first year of combination antiretroviral therapy. Sci Rep 2018; 8:16947. [PMID: 30446683 PMCID: PMC6240055 DOI: 10.1038/s41598-018-35271-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 11/01/2018] [Indexed: 12/26/2022] Open
Abstract
The HIV-human metabolic relationship is a complex interaction convoluted even more by antiretroviral therapy (cART) and comorbidities. The ability of cART to undo the HIV induced metabolic dysregulation is unclear and under-investigated. Using targeted metabolomics and multiplex immune biomarker analysis, we characterized plasma samples obtained from 18 untreated HIV-1-infected adult patients and compared these to a non-HIV infected (n = 23) control population. The biogenic amine perturbations during an untreated HIV infection implicated altered tryptophan- nitrogen- and muscle metabolism. Furthermore, the lipid profiles of untreated patients were also significantly altered compared to controls. In untreated HIV infection, the sphingomyelins and phospholipids correlated negatively to markers of infection IP-10 and sIL-2R whereas a strong association was found between triglycerides and MCP-1. In a second cohort, we characterized plasma samples obtained from 28 HIV-1-infected adult patients before and 12 months after the start of cART, to investigate the immune-metabolic changes associated with cART. The identified altered immune-metabolic pathways of an untreated HIV infection showed minimal change after 12 months of cART. In conclusion, 12 months of cART impacts only mildly on the metabolic dysregulation underlying an untreated HIV infection and provide insights into the comorbidities present in virally suppressed HIV patients.
Collapse
|
17
|
Trevillyan JM, Wong G, Puls R, Petoumenos K, Emery S, Mellett NA, Mundra PA, Meikle PJ, Hoy JF, for the ALTAIR Study Group. Changes in plasma lipidome following initiation of antiretroviral therapy. PLoS One 2018; 13:e0202944. [PMID: 30157268 PMCID: PMC6114786 DOI: 10.1371/journal.pone.0202944] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 08/13/2018] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION HIV and antiretroviral therapy (ART) have been associated with increased cardiovascular disease and important changes in lipid metabolism. Advances in mass-spectrometry technology allow for the detailed assessment of individual lipid species which may illuminate the mechanisms underlying increased cardiovascular risk. We describe the change in plasma lipidome with initiation of antiretroviral therapy and compare these by regimen. METHODS Plasma lipid profiling (by electrospray isonisation-tandem mass spectrometry) was performed on ARV-naive HIV positive participants randomised to one of three regimens; tenofovir/emtricitabine with efavirenz, ritonavir-boosted atazanavir (atazanavir/r) or zidovudine/abacavir. Participants (n = 115) who remained on their randomised regimen with complete samples available at baseline, week 12 and 48 were included. 306 lipid species from 22 lipid classes were analysed. RESULTS Initiation of ART led to significant changes in lipidome which were partly dependent on the randomised regimen received. This led to significant differences in 72 lipid species and 7 classes (cholesterol ester, free cholesterol, phosphatidylcholine, GM3 ganglioside, trihexosylceramide, monohexosylceramide, and ceramides) by arm at week 48. Consistently higher lipid concentrations were seen with efavirenz compared with atazanavir/r or zidovudine/abacavir. Twelve of the lipid species and two lipid classes (cholesterol esters and ceramides) that were significantly increased in the efavirenz arm compared with the atazanavir/r or zidovudine/abacavir arms have previously been associated with future cardiovascular events in HIV positive patients. Change in HIV viral load was predictive of change in 3 lipid species. CONCLUSIONS Initiation of ART lead to significant changes in the plasma lipidome that were greatest in those receiving efavirenz.
Collapse
Affiliation(s)
- Janine M. Trevillyan
- Department of Infectious Diseases, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Australia
- * E-mail:
| | - Gerard Wong
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Rebekah Puls
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | | | - Sean Emery
- The Kirby Institute, UNSW Sydney, Sydney, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Australia
| | | | | | | | - Jennifer F. Hoy
- Department of Infectious Diseases, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Australia
- Department of Infectious Diseases, Alfred Hospital, Melbourne, Australia
| | | |
Collapse
|
18
|
Pinto DSM, da Silva MJLV. Cardiovascular Disease in the Setting of Human Immunodeficiency Virus Infection. Curr Cardiol Rev 2018; 14:25-41. [PMID: 29189172 PMCID: PMC5872259 DOI: 10.2174/1573403x13666171129170046] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/10/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022] Open
Abstract
Background: Since the introduction of Antiretroviral Therapy (ART), the life expectancy and health quality for patients infected with Human Immunodeficiency Virus (HIV) have significant-ly improved. Nevertheless, as a result of not only the deleterious effects of the virus itself and pro-longed ART, but also the effects of aging, cardiovascular diseases have emerged as one of the most common causes of death among these patients. Objective: The purpose of this review is to explore the new insights on the spectrum of Cardiovascu-lar Disease (CVD) in HIV infection, with emphasis on the factors that contribute to the atherosclerot-ic process and its role in the development of acute coronary syndrome in the setting of infection. Methods: A literature search using PubMed, ScienceDirect and Web of Science was performed. Ar-ticles up to Mar, 2017, were selected for inclusion. The search was conducted using MeSH terms, with the following key terms: [human immunodeficiency virus AND (cardiovascular disease OR coronary heart disease) AND (antiretroviral therapy AND (cardiovascular disease OR coronary heart disease))]. Results: Clinical cardiovascular disease tends to appear approximately 10 years before in infected in-dividuals, when compared to the general population. The pathogenesis behind the cardiovascular, HIV-associated complications is complex and multifactorial, involving traditional CVD risk factors, as well as factors associated with the virus itself - immune activation and chronic inflammation – and the metabolic disorders related to ART regimens. Conclusion: Determining the cardiovascular risk among HIV-infected patients, as well as targeting and treating conditions that predispose to CVD, are now emerging concerns among physicians.
Collapse
Affiliation(s)
- Daniela Sofia Martins Pinto
- Department of Medicine, Faculty of Medicine, Porto University, Al. Prof. Hernâni Monteiro 4200-319, Porto, Portugal
| | | |
Collapse
|
19
|
Abstract
Infection with the human immunodeficiency virus (HIV), and subsequent treatment with antiretroviral therapy (ART), is often associated with perturbations in lipid profiles. Furthermore, persistent inflammation, in spite of suppression of viral replication by ART, likely contributes to modifications in lipid composition and function, exacerbating risk for development of cardiovascular disease (CVD). Increased levels of several pro-inflammatory lipid species, including oxidized low-density lipoprotein (LDL) and high-density lipoprotein (HDL), have been measured in HIV-infected persons and are associated with markers of immune activation. The mechanisms linked to this bidirectional relationship in which inflammation increases lipid levels and promotes their modification, and these modified lipid species perpetuate inflammatory processes, require further investigation. Treatment with statins and other lifestyle modifications, including improvement in dietary intake and exercise, are critical to reducing CVD risk. Well-designed clinical trials that take into account the complex relationships among lipids and inflammation within persons infected with HIV need to be considered.
Collapse
Affiliation(s)
- Nicholas T Funderburg
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University, 453 W. 10th Ave., 535A Atwell Hall, Columbus, OH, 43210, USA
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, Bethesda, MD, 20892, USA.
| |
Collapse
|
20
|
Gilotra TS, Geraci SA. C-Reactive Protein as an Independent Cardiovascular Risk Predictor in HIV+ Patients: A Focused Review of Published Studies. J Clin Med Res 2017; 9:891-899. [PMID: 29038665 PMCID: PMC5633088 DOI: 10.14740/jocmr3154w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 09/08/2017] [Indexed: 12/23/2022] Open
Abstract
Patients infected with the human immunodeficiency virus (HIV+) are living longer and at heightened risk for developing cardiovascular events (CVEs). Commonly used prediction tools appear to misrepresent their CVE risk to varying degrees and in varying directions. Inclusion of markers of cellular infection, chronic immune activation and/or systemic inflammation into risk models might provide better predictive accuracy. Observational studies assessing the relationship of high-sensitivity C-reactive protein (hs-CRP) to CVE in HIV+ patients have reported inconsistent findings. This review of published studies attempted to determine if the available evidence supports its potential use in new models for stable, treated HIV+ patients. We searched the PubMed database using keywords and combinations of “HIV” AND “cardiovascular risk” AND “CRP”. Papers presenting original analyses, associating hs-CRP concentration as an independent variable to hard cardiovascular outcomes (myocardial infarction and cardiovascular death), or to hard CVE as part of a composite endpoint, were included. Five observational studies met inclusion/exclusion criteria for review. Three papers identified an association between elevated hs-CRP and CVE, while two others failed to find any significant association. All reports were heterogeneous in terms of independent variables, controls, and designs. The larger and more rigorous studies, employing higher rates of confounder controls and more objective endpoints in their composites, showed positive associations. Though not conclusive, the preponderance of the evidence at this time supports CRP as a potentially valuable factor to be studied in prospective cardiovascular risk prediction investigations in HIV+ patients.
Collapse
Affiliation(s)
- Tarvinder S Gilotra
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Stephen A Geraci
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Division of Cardiology, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| |
Collapse
|
21
|
Yeoh HL, Cheng AC, Cherry CL, Weir JM, Meikle PJ, Hoy JF, Crowe SM, Palmer CS. Immunometabolic and Lipidomic Markers Associated With the Frailty Index and Quality of Life in Aging HIV+ Men on Antiretroviral Therapy. EBioMedicine 2017; 22:112-121. [PMID: 28754302 PMCID: PMC5552224 DOI: 10.1016/j.ebiom.2017.07.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/15/2017] [Accepted: 07/17/2017] [Indexed: 01/09/2023] Open
Abstract
Chronic immune activation persists despite antiretroviral therapy (ART) in HIV+ individuals and underpins an increased risk of age-related co-morbidities. We assessed the Frailty Index in older HIV+ Australian men on ART. Immunometabolic markers on monocytes and T cells were analyzed using flow cytometry, plasma innate immune activation markers by ELISA, and lipidomic profiling by mass spectrometry. The study population consisted of 80 HIV+ men with a median age of 59 (IQR, 56-65), and most had an undetectable viral load (92%). 24% were frail, and 76% were non-frail. Frailty was associated with elevated Glucose transporter-1 (Glut1) expression on the total monocytes (p=0.04), increased plasma levels of innate immune activation marker sCD163 (OR, 4.8; CI 1.4-15.9, p=0.01), phosphatidylethanolamine PE(36:3) (OR, 5.1; CI 1.7-15.5, p=0.004) and triacylglycerol TG(16:1_18:1_18:1) (OR, 3.4; CI 1.3-9.2, p=0.02), but decreased expression of GM3 ganglioside, GM3(d18:1/18:0) (OR, 0.1; CI 0.0-0.6, p=0.01) and monohexosylceramide HexCerd(d18:1/22:0) (OR, 0.1; CI 0.0-0.5, p=0.004). There is a strong inverse correlation between quality of life and the concentration of PE(36:3) (ρ=-0.33, p=0.004) and PE(36:4) (ρ=-0.37, p=0.001). These data suggest that frailty is associated with increased innate immune activation and abnormal lipidomic profile. These markers should be investigated in larger, longitudinal studies to determine their potential as biomarkers for frailty.
Collapse
Affiliation(s)
- Hui-Ling Yeoh
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Allen C Cheng
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Department of Epidemiology and Preventive Medicine, Monash University, Level 6, The Alfred Centre (Alfred Hospital), 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Catherine L Cherry
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; School of Physiology, University of the Witwatersrand, 1 Jan Smuts Avenue, Braamfontein, 2000 Johannesburg, South Africa
| | - Jacquelyn M Weir
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Jennifer F Hoy
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Suzanne M Crowe
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Clovis S Palmer
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
22
|
Meikle P, Low H, Churchill MJ, Bukrinsky M, Sviridov D. Lipidomic dataset of plasma from patients infected with wild type and nef-deficient HIV-1 strain. Data Brief 2015; 6:168-75. [PMID: 26862555 PMCID: PMC4706608 DOI: 10.1016/j.dib.2015.11.067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 11/15/2022] Open
Abstract
Previous in vitro and in vivo studies demonstrated that HIV protein nef plays a key role in impairing cellular and systemic cholesterol metabolism in HIV disease, but clinical support for these findings is lacking. Here we present the data of comparative lipidomic analysis (330 lipid species) of plasma samples from HIV-negative subjects, patients infected with WT HIV-1 strain and patients infected with nef-deficient strain of HIV-1. We determine which effects of HIV on plasma lipidome are explained by the presence of nef. The data can be used to evaluate cardiovascular risk in HIV disease and to assess the role of nef in HIV-induced disturbances in systemic lipid metabolism. The full impact of nef deficiency on lipid and lipoprotein metabolism in HIV-infected patients is presented in the accompanying study "Lipid Metabolism in Patients Infected with Nef-deficient HIV-1 Strain" [1].
Collapse
Affiliation(s)
- Peter Meikle
- Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC 3004, Australia
| | - Hann Low
- Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC 3004, Australia
| | - Melissa J Churchill
- Macfarlane Burnett Institute for Medical Research and Public Health, 85 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Michael Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, 2300 I St. NW, Ross Hall, Washington DC 20037, USA
| | - Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC 3004, Australia
| |
Collapse
|
23
|
Low H, Cheng L, Di Yacovo MS, Churchill MJ, Meikle P, Bukrinsky M, Hill AF, Sviridov D. Lipid metabolism in patients infected with Nef-deficient HIV-1 strain. Atherosclerosis 2015; 244:22-8. [PMID: 26581048 DOI: 10.1016/j.atherosclerosis.2015.10.103] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/26/2015] [Accepted: 10/26/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND HIV protein Nef plays a key role in impairing cholesterol metabolism in both HIV infected and bystander cells. The existence of a small cohort of patients infected with Nef-deficient strain of HIV presented a unique opportunity to test the effect of Nef on lipid metabolism in a clinical setting. METHODS Here we report the results of a study comparing six patients infected with Nef-deficient strain of HIV (ΔNefHIV) with six treatment-naïve patients infected with wild-type HIV (WT HIV). Lipoprotein profile, size and functionality of high density lipoprotein (HDL) particles as well as lipidomic and microRNA profiles of patient plasma were analyzed. RESULTS We found that patients infected with ΔNefHIV had lower proportion of subjects with plasma HDL-C levels <1 mmol/l compared to patients infected with WT HIV. Furthermore, compared to a reference group of HIV-negative subjects, there was higher abundance of smaller under-lipidated HDL particles in plasma of patients infected with WT HIV, but not in those infected with ΔNefHIV. Lipidomic analysis of plasma revealed differences in abundance of phosphatidylserine and sphingolipids between patients infected with ΔNefHIV and WT HIV. MicroRNA profiling revealed that plasma abundance of 24 miRNAs, many of those involved in regulation of lipid metabolism, was differentially regulated by WT HIV and ΔNefHIV. CONCLUSION Our findings are consistent with HIV protein Nef playing a significant role in pathogenesis of lipid-related metabolic complications of HIV disease.
Collapse
Affiliation(s)
- Hann Low
- Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Lesley Cheng
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Maria-Silvana Di Yacovo
- Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia; Institut de Resercha Biomedica Bellvitge, University of Barcelona, Gran Via de l'Hospitalet, 199, 08908 Hospitalet de Llobregat, Barcelona, Spain
| | - Melissa J Churchill
- Macfarlane Burnett Institute for Medical Research and Public Health, 85 Commercial Rd, Melbourne, VIC, 3004, Australia
| | - Peter Meikle
- Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia; Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Michael Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, 2300 I St. NW, Ross Hall, Washington DC, 20037, USA
| | - Andrew F Hill
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
24
|
Ramezani A, Dubrovsky L, Pushkarsky T, Sviridov D, Karandish S, Raj DS, Fitzgerald ML, Bukrinsky M. Stimulation of Liver X Receptor Has Potent Anti-HIV Effects in a Humanized Mouse Model of HIV Infection. J Pharmacol Exp Ther 2015; 354:376-83. [PMID: 26126533 PMCID: PMC4538872 DOI: 10.1124/jpet.115.224485] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 06/29/2015] [Indexed: 01/24/2023] Open
Abstract
Previous studies demonstrated that liver X receptor (LXR) agonists inhibit human immunodeficiency virus (HIV) replication by upregulating cholesterol transporter ATP-binding cassette A1 (ABCA1), suppressing HIV production, and reducing infectivity of produced virions. In this study, we extended these observations by analyzing the effect of the LXR agonist T0901317 [N-[4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl]-N-(2,2,2-trifluoroethyl)benzenesulfonamide] on the ongoing HIV infection and investigating the possibility of using LXR agonist for pre-exposure prophylaxis of HIV infection in a humanized mouse model. Pre-exposure of monocyte-derived macrophages to T0901317 reduced susceptibility of these cells to HIV infection in vitro. This protective effect lasted for up to 4 days after treatment termination and correlated with upregulated expression of ABCA1, reduced abundance of lipid rafts, and reduced fusion of the cells with HIV. Pre-exposure of peripheral blood leukocytes to T0901317 provided only a short-term protection against HIV infection. Treatment of HIV-exposed humanized mice with LXR agonist starting 2 weeks postinfection substantially reduced viral load. When eight humanized mice were pretreated with LXR agonist prior to HIV infection, five animals were protected from infection, two had viral load at the limit of detection, and one had viral load significantly reduced relative to mock-treated controls. T0901317 pretreatment also reduced HIV-induced dyslipidemia in infected mice. In conclusion, these results reveal a novel link between LXR stimulation and cell resistance to HIV infection and suggest that LXR agonists may be good candidates for development as anti-HIV agents, in particular for pre-exposure prophylaxis of HIV infection.
Collapse
Affiliation(s)
- Ali Ramezani
- George Washington University School of Medicine and Health Sciences, Washington, DC (A.R., L.D., T.P., S.K., D.S.R., M.B.); Baker International Diabetes Institute, Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); and Harvard Medical School, Lipid Metabolism Unit, Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts (M.L.F.)
| | - Larisa Dubrovsky
- George Washington University School of Medicine and Health Sciences, Washington, DC (A.R., L.D., T.P., S.K., D.S.R., M.B.); Baker International Diabetes Institute, Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); and Harvard Medical School, Lipid Metabolism Unit, Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts (M.L.F.)
| | - Tatiana Pushkarsky
- George Washington University School of Medicine and Health Sciences, Washington, DC (A.R., L.D., T.P., S.K., D.S.R., M.B.); Baker International Diabetes Institute, Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); and Harvard Medical School, Lipid Metabolism Unit, Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts (M.L.F.)
| | - Dmitri Sviridov
- George Washington University School of Medicine and Health Sciences, Washington, DC (A.R., L.D., T.P., S.K., D.S.R., M.B.); Baker International Diabetes Institute, Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); and Harvard Medical School, Lipid Metabolism Unit, Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts (M.L.F.)
| | - Sara Karandish
- George Washington University School of Medicine and Health Sciences, Washington, DC (A.R., L.D., T.P., S.K., D.S.R., M.B.); Baker International Diabetes Institute, Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); and Harvard Medical School, Lipid Metabolism Unit, Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts (M.L.F.)
| | - Dominic S Raj
- George Washington University School of Medicine and Health Sciences, Washington, DC (A.R., L.D., T.P., S.K., D.S.R., M.B.); Baker International Diabetes Institute, Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); and Harvard Medical School, Lipid Metabolism Unit, Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts (M.L.F.)
| | - Michael L Fitzgerald
- George Washington University School of Medicine and Health Sciences, Washington, DC (A.R., L.D., T.P., S.K., D.S.R., M.B.); Baker International Diabetes Institute, Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); and Harvard Medical School, Lipid Metabolism Unit, Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts (M.L.F.)
| | - Michael Bukrinsky
- George Washington University School of Medicine and Health Sciences, Washington, DC (A.R., L.D., T.P., S.K., D.S.R., M.B.); Baker International Diabetes Institute, Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); and Harvard Medical School, Lipid Metabolism Unit, Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts (M.L.F.)
| |
Collapse
|