1
|
Cheng Y, Jung J, Guo L, Shuboni-Mulligan DD, Chen JF, Hu W, Guo ML. HIV-TAT dysregulates microglial lipid metabolism through SREBP2/miR-124 axis: Implication of lipid droplet accumulation microglia in NeuroHIV. Brain Behav Immun 2024; 123:108-122. [PMID: 39260763 DOI: 10.1016/j.bbi.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/26/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024] Open
Abstract
Chronic HIV infection can dysregulate lipid/cholesterol metabolism in the peripheral system, contributing to the higher incidences of diabetes and atherosclerosis in HIV (+) individuals. Recently, accumulating evidence indicate that HIV proteins can also dysregulate lipid/cholesterol metabolism in the brain and such dysregulation could be linked with the pathogenesis of HIV-associated neurological disorders (HAND)/NeuroHIV. To further characterize the association between lipid/cholesterol metabolism and HAND, we employed HIV-inducible transactivator of transcription (iTAT) and control mice to compare their brain lipid profiles. Our results reveal that HIV-iTAT mice possess dysregulated lipid profiles and have increased numbers of lipid droplets (LDs) accumulation microglia (LDAM) in the brains. HIV protein TAT can upregulate LDs formation through enhancing the lipid/cholesterol synthesis in vitro. Mechanistically, HIV-TAT increases the expression of sterol regulatory element-binding protein 2 (SREBP2) through microRNA-124 downregulation. Cholesterol synthesis inhibition can block HIV-TAT-mediated NLRP3 inflammasome activation and microglial activation in vitro as well as mitigate aging-related behavioral impairment and memory deficiency in HIV-iTAT mice. Taken together, our results indicate an inherent role of lipid metabolism and LDAM in the pathogenesis of NeuroHIV (immunometabolism). These findings suggest that LDAM reversal through modulating lipid/cholesterol metabolism could be a novel therapeutic target for ameliorating NeuroHIV symptoms in chronic HIV (+) individuals.
Collapse
Affiliation(s)
- Yan Cheng
- Department of Biomedical and Translational Sciences, Macro & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Jaekeun Jung
- Department of Biomedical and Translational Sciences, Macro & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Liyang Guo
- Department of Biomedical and Translational Sciences, Macro & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Dorela D Shuboni-Mulligan
- Department of Biomedical and Translational Sciences, Macro & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Jian-Fu Chen
- Center for Craniofacial Molecular Biology, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Wenhui Hu
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Ming-Lei Guo
- Department of Biomedical and Translational Sciences, Macro & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Macro & Joan Brock Virginia Health Science, Old Dominion University, Norfolk, VA 23507, USA.
| |
Collapse
|
2
|
Bailin SS, Gabriel CL, Gangula RD, Hannah L, Nair S, Carr JJ, Terry JG, Silver HJ, Simmons JD, Mashayekhi M, Kalams SA, Mallal S, Kropski JA, Wanjalla CN, Koethe JR. Single-cell Analysis of Subcutaneous Fat Reveals Pro-fibrotic Cells that Correlate with Visceral Adiposity in HIV. J Clin Endocrinol Metab 2024:dgae369. [PMID: 38820087 DOI: 10.1210/clinem/dgae369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 04/26/2024] [Accepted: 05/30/2024] [Indexed: 06/02/2024]
Abstract
CONTEXT Cardiometabolic diseases are common in persons with HIV (PWH) on antiretroviral therapy (ART), which has been attributed to preferential lipid storage in visceral adipose tissue (VAT) compared with subcutaneous adipose tissue (SAT). However, the relationship of SAT-specific cellular and molecular programs with VAT volume is poorly understood in PWH. OBJECTIVE We characterized SAT cell-type specific composition and transcriptional programs that are associated with greater VAT volume in PWH on contemporary ART. METHODS We enrolled PWH on long-term ART with a spectrum of metabolic health. Ninety-two participants underwent SAT biopsy for bulk RNA sequencing and 43 had single-cell RNA sequencing. Computed tomography quantified VAT volume and insulin resistance was calculated using HOMA2-IR. RESULTS VAT volume was associated with HOMA2-IR (p < 0.001). Higher proportions of SAT intermediate macrophages (IMs), myofibroblasts, and MYOC + fibroblasts were associated with greater VAT volume using partial Spearman's correlation adjusting for age, sex, and body mass index (ρ=0.34-0.49, p < 0.05 for all). Whole SAT transcriptomics showed PWH with greater VAT volume have increased expression of extracellular matrix (ECM)- and inflammation-associated genes, and reduced expression of lipolysis- and fatty acid metabolism-associated genes. CONCLUSIONS In PWH, greater VAT volume is associated with higher proportion of SAT IMs and fibroblasts, and a SAT ECM and inflammatory transcriptome, which is similar to findings in HIV-negative persons with obesity. These data identify SAT cell-type specific changes associated with VAT volume in PWH that could underlie the high rates of cardiometabolic diseases in PWH, though additional longitudinal studies are needed to define directionality and mechanisms.
Collapse
Affiliation(s)
- Samuel S Bailin
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Curtis L Gabriel
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, Tennessee, USA
| | - Rama D Gangula
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - LaToya Hannah
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sangeeta Nair
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John Jeffrey Carr
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James G Terry
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Heidi J Silver
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Joshua D Simmons
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mona Mashayekhi
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Spyros A Kalams
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Center for Translational Immunology and Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Simon Mallal
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Center for Translational Immunology and Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Insitute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia
- Vanderbilt Technologies for Advanced Genomics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jonathan A Kropski
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Department of Medicine, Division of Allergy and Pulmonology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Deparment of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Celestine N Wanjalla
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Center for Translational Immunology and Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John R Koethe
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Center for Translational Immunology and Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
3
|
Torigoe TH, Willcox DC, Shimabukuro M, Higa M, Gerschenson M, Andrukhiv A, Suzuki M, Morris BJ, Chen R, Gojanovich GS, Allsopp RC, Willcox BJ. Novel protective effect of the FOXO3 longevity genotype on mechanisms of cellular aging in Okinawans. NPJ AGING 2024; 10:18. [PMID: 38459055 PMCID: PMC10923797 DOI: 10.1038/s41514-024-00142-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/06/2024] [Indexed: 03/10/2024]
Abstract
The genetic association of FOXO3 genotypes with human longevity is well established, although the mechanism is not fully understood. We now report on the relationship of the FOXO3 longevity variant rs2802292 with telomere length, telomerase activity, FOXO3 expression, and inflammatory cytokine levels in men and women. In agreement with earlier work, the FOXO3 longevity variant conferred protection against telomere shortening of peripheral blood mononuclear cells from adults aged 55 years and older. This was accompanied by higher levels of telomerase activity in mononuclear cells for carriers of the longevity-associated FOXO3 G-allele of SNP rs2802292 (P = 0.015). FOXO3 mRNA expression increased slightly with age in both young (P = 0.02) and old (P = 0.08) G-allele carriers. Older female G-allele carriers displayed a modest decline in levels of pro-inflammatory cytokine IL-6 with age (P = 0.07). In contrast, older male G-allele carriers displayed an age-dependent increase in levels of anti-inflammatory cytokine IL-10 with age (P = 0.04). Thus, FOXO3 may act through several different pro-longevity mechanisms, which may differ by age and sex.
Collapse
Affiliation(s)
- Trevor H Torigoe
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
| | - D Craig Willcox
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
- Department of Human Welfare, Okinawa International University, Ginowan, Okinawa, Japan
- Okinawa Research Center for Longevity Science, Urasoe, Okinawa, Japan
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA
| | - Michio Shimabukuro
- Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
- Diabetes and Life-Style Related Disease Center, Tomishiro Central Hospital, Tomishiro, Okinawa, Japan
| | - Moritake Higa
- Diabetes and Life-Style Related Disease Center, Tomishiro Central Hospital, Tomishiro, Okinawa, Japan
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Anastasia Andrukhiv
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Makoto Suzuki
- Okinawa Research Center for Longevity Science, Urasoe, Okinawa, Japan
| | - Brian J Morris
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Randi Chen
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA
| | - Greg S Gojanovich
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Richard C Allsopp
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
- Okinawa Research Center for Longevity Science, Urasoe, Okinawa, Japan.
| | - Bradley J Willcox
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
- Okinawa Research Center for Longevity Science, Urasoe, Okinawa, Japan
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA
| |
Collapse
|
4
|
Bailin SS, Koethe JR, Rebeiro PF. The pathogenesis of obesity in people living with HIV. Curr Opin HIV AIDS 2024; 19:6-13. [PMID: 37934696 PMCID: PMC10842175 DOI: 10.1097/coh.0000000000000834] [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: 11/09/2023]
Abstract
PURPOSE OF REVIEW The public health challenge of overweight and obesity increasingly affects people living with HIV (PWH). These effects have also accelerated as the prevalence of antiretroviral therapy (ART) use has increased among PWH. It is therefore also critical that we examine and understand the pathogenesis of obesity among PWH.This review will aim to summarize relevant and recent literature related to the risks of weight gain and obesity associated with HIV disease progression, cardiometabolic disease, and multimorbidity among PWH. Further, we will discuss adipose tissue changes associated with weight gain and obesity and how these changes relate to metabolic complications. RECENT FINDINGS Several observational and experimental studies in recent years have evaluated the role of contemporary ART regimens, particularly integrase strand transfer inhibitors (INSTIs) and tenofovir alafenamide (TAF), as contributors to weight gain, obesity, and cardiometabolic disease, though the mechanisms remain unclear. Metabolic dysregulation has also been linked to ectopic fat deposition and alterations in innate and adaptive immune cell populations in adipose tissue that accompany HIV and obesity. These factors continue to contribute to an increasing burden of metabolic diseases in an aging HIV population. SUMMARY Obesity accompanies an increasing burden of metabolic disease among PWH, and understanding the role of fat partitioning and HIV and ART-related adipose tissue dysfunction may guide prevention and treatment strategies.
Collapse
Affiliation(s)
| | - John R Koethe
- Department of Medicine, Division of Infectious Diseases
| | - Peter F Rebeiro
- Department of Medicine, Division of Infectious Diseases
- Department of Medicine, Division of Epidemiology
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
5
|
Bailin SS, Kropski JA, Gangula RD, Hannah L, Simmons JD, Mashayekhi M, Ye F, Fan R, Mallal S, Warren CM, Kalams SA, Gabriel CL, Wanjalla CN, Koethe JR. Changes in subcutaneous white adipose tissue cellular composition and molecular programs underlie glucose intolerance in persons with HIV. Front Immunol 2023; 14:1152003. [PMID: 37711619 PMCID: PMC10499182 DOI: 10.3389/fimmu.2023.1152003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 08/07/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Subcutaneous adipose tissue (SAT) is a critical regulator of systemic metabolic homeostasis. Persons with HIV (PWH) have an increased risk of metabolic diseases and significant alterations in the SAT immune environment compared with the general population. Methods We generated a comprehensive single-cell multi-omic SAT atlas to characterize cellular compositional and transcriptional changes in 59 PWH across a spectrum of metabolic health. Results Glucose intolerance was associated with increased lipid-associated macrophages, CD4+ and CD8+ T effector memory cells, and decreased perivascular macrophages. We observed a coordinated intercellular regulatory program which enriched for genes related to inflammation and lipid-processing across multiple cell types as glucose intolerance increased. Increased CD4+ effector memory tissue-resident cells most strongly associated with altered expression of adipocyte genes critical for lipid metabolism and cellular regulation. Intercellular communication analysis demonstrated enhanced pro-inflammatory and pro-fibrotic signaling between immune cells and stromal cells in PWH with glucose intolerance compared with non-diabetic PWH. Lastly, while cell type-specific gene expression among PWH with diabetes was globally similar to HIV-negative individuals with diabetes, we observed substantially divergent intercellular communication pathways. Discussion These findings suggest a central role of tissue-resident immune cells in regulating SAT inflammation among PWH with metabolic disease, and underscore unique mechanisms that may converge to promote metabolic disease.
Collapse
Affiliation(s)
- Samuel S. Bailin
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jonathan A. Kropski
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, United States
- Deparment of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
| | - Rama D. Gangula
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
| | - LaToya Hannah
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Joshua D. Simmons
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Mona Mashayekhi
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Fei Ye
- Department of Biostatics, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Run Fan
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Simon Mallal
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
- Insitute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia
- Vanderbilt Technologies for Advanced Genomics, Vanderbilt University Medical Center, Nashville, TN, United States
- Center for Translational Immunology and Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Christian M. Warren
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Spyros A. Kalams
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
- Center for Translational Immunology and Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Curtis L. Gabriel
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Nashville, TN, United States
| | - Celestine N. Wanjalla
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Center for Translational Immunology and Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - John R. Koethe
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, United States
- Center for Translational Immunology and Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
6
|
Guaraldi G, Bonfanti P, Di Biagio A, Gori A, Milić J, Saltini P, Segala FV, Squillace N, Taramasso L, Cingolani A. Evidence gaps on weight gain in people living with HIV: a scoping review to define a research agenda. BMC Infect Dis 2023; 23:230. [PMID: 37060030 PMCID: PMC10103467 DOI: 10.1186/s12879-023-08174-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 03/17/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Combined antiretroviral therapy (cART) dramatically improved survival in people living with HIV (PLWH) but is associated with weight gain (WG), raising concern for a possible obesity epidemic in PLWH. This scoping review aims to identify the gaps in the existing evidence on WG in PLWH and generate a future research agenda. METHODS This review was conducted according to the methodology for scoping studies and reported according to the PRISMA Extension for Scoping Review checklist. Articles published in English in the last 10 years indexed in Pubmed, WHO Global Index Medicus, or Embase were searched using specific queries focused on WG in PLWH. RESULTS Following the selection process, 175 included articles were reviewed to search for the available evidence on four specific topics: (I) definition of WG in PLWH, (II) pathogenesis of WG in PLWH, (III) impact of ART on WG, (IV) correlation of WG with clinical outcomes. A summary of the data enabled us to identify gaps and clearly define the following research agenda: (I) develop a data-driven definition of WG in PLWH and define noninvasive assessment methods for body weight and fat composition; (II) further investigate the interaction between HIV/cART and immunity, metabolism, and adipose tissue; (III) establish the specific role of individual drugs on WG; (IV) clarify the independent role of WG, cART, HIV, and metabolic factors on clinical events. CONCLUSIONS The proposed research agenda may help define future research and fill the knowledge gaps that have emerged from this review.
Collapse
Affiliation(s)
- Giovanni Guaraldi
- HIV Metabolic Clinic, University of Modena and Reggio Emilia, Modena, Italy
| | - Paolo Bonfanti
- Infectious Diseases Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- University of Milano-Bicocca, Milan, Italy
| | - Antonio Di Biagio
- Infectious Diseases Unit, Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Andrea Gori
- Infectious Diseases Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| | - Jovana Milić
- HIV Metabolic Clinic, University of Modena and Reggio Emilia, Modena, Italy
| | - Paola Saltini
- Infectious Diseases Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Francesco V Segala
- Infectious Diseases Unit, Fondazione Policlinico Universitario A. Gemelli-Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Nicola Squillace
- Infectious Diseases Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Lucia Taramasso
- Infectious Diseases Unit, Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Antonella Cingolani
- Infectious Diseases Unit, Fondazione Policlinico Universitario A. Gemelli-Università Cattolica Del Sacro Cuore, Rome, Italy
| |
Collapse
|
7
|
Ashuro AA, Zhang SC, Wang T, Chu QS, Fu YS, Fan YG, Ye DQ. The Effect of protease inhibitors-based antiretroviral therapy on serum/plasma interleukin-6 levels among PLHIV: a systematic review and meta-analysis. J Med Virol 2022; 94:4669-4676. [PMID: 35665943 DOI: 10.1002/jmv.27912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 05/15/2022] [Accepted: 05/31/2022] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Recommended treatment regimen for HIV infection includes protease inhibitors/ritonavir (PIs/r) combined with two nucleoside reverse transcriptase inhibitors (2NRTIs), that enable to achieve and maintain viral suppression, restore and preserve immune function. However, there were inconsistent findings on the levels of interleukin-6 levels (IL-6). METHODOLOGY Systematic review and meta-analysis were performed to quantify the pooled effects of PIs/r-based ART on serum/plasma IL-6 levels in PLHIV. PubMed, Web of Science, and Embase were searched from the earliest record to November 4, 2020. Data analysis was conducted on Stata version 16 and Review Manager 5.3. A random-effect model was used to compute a pooled effect size and weighted mean difference (WMD) was considered the summary effect size. Heterogeneity between studies was estimated by Cochrane's Q test (chi-square test) and I-square statistic and subgroup analysis were performed to explore the source of heterogeneity. RESULT Initial search identified 3098 records and five studies (seven trials) met inclusion criteria. The pooled mean difference in serum/plasma IL-6 levels from baseline to follow-up was 0.534 pg/ml (95% CI: -0.012, 1.08, P=0.05, I2 =76.4%). In subgroup analysis, there was a significant association between increased serum/plasma IL-6 levels, and age group ≥35 years old, baseline CD4+ counts <350 cell/mm3 , and mean viral load ≥ 4.5 log10 copies/ml. CONCLUSION We found that serum/plasma IL-6 levels increased after combined anti-retroviral therapy (cART) among treatment-naïve individuals who initiated a successful combination of PIs/r with 2NRTIs. This result also highlights the need to monitor serum/plasma IL-6 levels during antiviral therapy, which may aid in the effective future treatment of systemic inflammation and related disorders following elevated IL-6 levels. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Akililu Alemu Ashuro
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, China, 81 Meishan Road, Hefei, Anhui, 230032, PR China
| | - Si-Chen Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, China, 81 Meishan Road, Hefei, Anhui, 230032, PR China
| | - Ting Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, China, 81 Meishan Road, Hefei, Anhui, 230032, PR China
| | - Qin-Shu Chu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, China, 81 Meishan Road, Hefei, Anhui, 230032, PR China
| | - Yuan-Sheng Fu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, China, 81 Meishan Road, Hefei, Anhui, 230032, PR China
| | - Yin-Guang Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, China, 81 Meishan Road, Hefei, Anhui, 230032, PR China
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, China, 81 Meishan Road, Hefei, Anhui, 230032, PR China
| |
Collapse
|
8
|
Bailin SS, Gabriel CL, Fan R, Ye F, Nair S, Terry JG, Carr JJ, Silver H, Wanjalla CN, Mashayekhi M, Lima M, Woodward B, Hannah L, Fuseini H, Ferguson JF, Kropski JA, Koethe JR. Relationship of Subcutaneous Adipose Tissue Inflammation-Related Gene Expression With Ectopic Lipid Deposition in Persons With HIV. J Acquir Immune Defic Syndr 2022; 90:175-183. [PMID: 35125474 PMCID: PMC9203874 DOI: 10.1097/qai.0000000000002926] [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: 08/06/2021] [Accepted: 01/18/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Fat redistribution from subcutaneous adipose tissue (SAT) to the abdominal viscera, pericardium, liver, and skeletal muscle contributes to the rising burden of cardiometabolic disease among persons with HIV (PWH). Previous studies found SAT inflammation in PWH impairs lipid storage and persists despite plasma viral suppression on antiretroviral therapy (ART). In this study, we identified SAT immune-related genes associated with ectopic fat deposition in PWH on long-term ART. DESIGN AND METHODS A total of 92 PWH with well-controlled viremia underwent computed tomography imaging and abdominal SAT biopsy for gene expression analysis. SAT gene expression was measured using a NanoString panel of 255 immune-related genes. Associations between gene expression and computed tomography measurements of the volume and attenuation (radiodensity) of metabolically relevant ectopic fat depots were assessed using multivariable linear regression and network analysis. RESULTS Greater SAT volume was associated with higher visceral and pericardial adipose tissue volume, but lower skeletal muscle attenuation. Lower SAT attenuation, a measure of lipid content, was associated with lower visceral adipose tissue attenuation. Hierarchical clustering identified a subset of macrophage-related genes in SAT, including CCL2, CCL22, CCL13, CCR1, CD86, CD163, IL-6, IL-10, MRC1, and TREM2, which were associated with an increased lipid deposition in multiple ectopic depots. CONCLUSION Altered expression of macrophage-related genes in SAT is associated with differences in ectopic fat depot morphometrics among PWH on long-term ART, including in the pericardial and visceral compartments. These findings provide basis for future studies to assess host, virus, and treatment factors shaping the SAT immune environment and its effects on morphometric changes and metabolic comorbidities in PWH.
Collapse
Affiliation(s)
- Samuel S. Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Curtis L. Gabriel
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Run Fan
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fei Ye
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sangeeta Nair
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James G. Terry
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John Jeffrey Carr
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heidi Silver
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mona Mashayekhi
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Morgan Lima
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Beverly Woodward
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - LaToya Hannah
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hubaida Fuseini
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jane F. Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan A. Kropski
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
- Divison of Allergy, Pulmonary and Critical Care, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - John R. Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
9
|
Mashiko T, Tsukada K, Takada H, Wu SH, Kanayama K, Asahi R, Mori M, Kurisaki A, Oka S, Yoshimura K. Genetic and cytometric analyses of subcutaneous adipose tissue in patients with hemophilia and HIV-associated lipodystrophy. AIDS Res Ther 2022; 19:14. [PMID: 35246167 PMCID: PMC8895510 DOI: 10.1186/s12981-022-00432-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 01/31/2022] [Indexed: 11/15/2022] Open
Abstract
Background The authors recently performed plastic surgeries for a small number of patients with hemophilia, HIV infection, and morphologic evidence of lipodystrophy. Because the pathophysiological mechanism of HIV-associated lipodystrophy remains to be elucidated, we analyzed subcutaneous adipose tissues from the patients. Methods All six patients had previously been treated with older nucleoside analogue reverse-transcriptase inhibitors (NRTIs; stavudine, didanosine or zidovudine). Abdominal and inguinal subcutaneous fat samples were obtained from the HIV+ patients with hemophilia and HIV− healthy volunteers (n = 6 per group), and analyzed via DNA microarray, real-time PCR, flow cytometry and immunohistochemistry. Results The time from initial NRTI treatment to collecting samples were 21.7 years in average. Cytometric analysis revealed infiltration of inflammatory M1 macrophages into HIV-infected adipose tissue and depletion of adipose-derived stem cells, possibly due to exhaustion following sustained adipocyte death. Genetic analysis revealed that adipose tissue from HIV+ group had increased immune activation, mitochondrial toxicity, chronic inflammation, progressive fibrosis and adipocyte dysfunction (e.g. insulin resistance, inhibited adipocyte differentiation and accelerated apoptosis). Of note, both triglyceride synthesis and lipolysis were inhibited in adipose tissue from patients with HIV. Conclusions Our findings provide important insights into the pathogenesis of HIV-associated lipodystrophy, suggesting that fat redistribution may critically depend on adipocytes’ sensitivity to drug-induced mitochondrial toxicity, which may lead either to atrophy or metabolic complications. Supplementary Information The online version contains supplementary material available at 10.1186/s12981-022-00432-9.
Collapse
|
10
|
Yeregui E, Masip J, Viladés C, Domingo P, Pacheco YM, Blanco J, Mallolas J, Alba V, Vargas M, García-Pardo G, Negredo E, Olona M, Vidal-González J, Peraire M, Martí A, Reverté L, Gómez-Bertomeu F, Leal M, Vidal F, Peraire J, Rull A. Adipokines as New Biomarkers of Immune Recovery: Apelin Receptor, RBP4 and ZAG Are Related to CD4+ T-Cell Reconstitution in PLHIV on Suppressive Antiretroviral Therapy. Int J Mol Sci 2022; 23:ijms23042202. [PMID: 35216318 PMCID: PMC8874604 DOI: 10.3390/ijms23042202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 11/26/2022] Open
Abstract
A significant proportion of people living with HIV (PLHIV) who successfully achieve virological suppression fail to recover CD4+ T-cell counts. Since adipose tissue has been discovered as a key immune organ, this study aimed to assess the role of adipokines in the HIV immunodiscordant response. This is a multicenter prospective study including 221 PLHIV starting the first antiretroviral therapy (ART) and classified according to baseline CD4+ T-cell counts/µL (controls > 200 cells/µL and cases ≤ 200 cells/µL). Immune failure recovery was considered when cases did not reach more than 250 CD4+ T cells/µL at 144 weeks (immunological nonresponders, INR). Circulating adipokine concentrations were longitudinally measured using enzyme-linked immunosorbent assays. At baseline, apelin receptor (APLNR) and zinc-alpha-2-glycoprotein (ZAG) concentrations were significantly lower in INRs than in immunological responders (p = 0.043 and p = 0.034), and they remained lower during all ART follow-up visits (p = 0.044 and p = 0.028 for APLNR, p = 0.038 and p = 0.010 for ZAG, at 48 and 144 weeks, respectively). ZAG levels positively correlated with retinol-binding protein 4 (RBP4) levels (p < 0.01), and low circulating RBP4 concentrations were related to a low CD4+ T-cell gain (p = 0.018 and p = 0.039 at 48 and 144 weeks, respectively). Multiple regression adjusted for clinical variables and adipokine concentrations confirmed both low APLNR and RBP4 as independent predictors for CD4+ T cells at 144 weeks (p < 0.001). In conclusion, low APLNR and RBP4 concentrations were associated with poor immune recovery in treated PLHIV and could be considered predictive biomarkers of a discordant immunological response.
Collapse
Affiliation(s)
- Elena Yeregui
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Institut Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
| | - Jenifer Masip
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
| | - Consuelo Viladés
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Institut Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
| | - Pere Domingo
- Infectious Diseases Unit, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
| | - Yolanda M. Pacheco
- Laboratory of Immunology, Institute of Biomedicine of Seville, IBiS, 41013 Seville, Spain;
- UGC Clinical Laboratories, Virgen del Rocío University Hospital/CSIC/University of Seville, 41013 Seville, Spain
| | - Julià Blanco
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
- IrsiCaixa AIDS Research Institute, 08916 Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Spain
- Infectious Diseases and Immunity, Faculty of Medicine, Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), 08500 Vic, Spain
| | - Josep Mallolas
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
- HIV Unit and Infectious Diseases Service, Hospital Clinic-IDIBAPS, 08036 Barcelona, Spain
| | - Verónica Alba
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
| | - Montserrat Vargas
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
| | - Graciano García-Pardo
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Institut Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
| | - Eugènia Negredo
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
- Infectious Diseases and Immunity, Faculty of Medicine, Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), 08500 Vic, Spain
- Fundació de la Lluita contra les Infeccions, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Montserrat Olona
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
| | | | - Maria Peraire
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
| | - Anna Martí
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Institut Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
| | - Laia Reverté
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Institut Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
| | - Fréderic Gómez-Bertomeu
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
| | - Manuel Leal
- Internal Medicine Service, Hospital Viamed Santa Ángela de la Cruz, 41014 Seville, Spain;
| | - Francesc Vidal
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Institut Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
- Correspondence: (F.V.); (A.R.)
| | - Joaquim Peraire
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Institut Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
| | - Anna Rull
- Infection and Immunity Research Group (INIM), Hospital Universitari de Tarragona Joan XXIII (HJ23), 43005 Tarragona, Spain; (E.Y.); (J.M.); (C.V.); (V.A.); (M.V.); (G.G.-P.); (M.O.); (A.M.); (L.R.); (F.G.-B.); (J.P.)
- Institut Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
- Infection and Immunity Research Group (INIM), Universitat Rovira i Virgili (URV), 43003 Tarragona, Spain;
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.B.); (J.M.); (E.N.)
- Correspondence: (F.V.); (A.R.)
| |
Collapse
|
11
|
Poret JM, Guidry JJ, Simon L, Molina PE. Chronic binge alcohol and ovariectomy dysregulate omental adipose tissue metaboproteome in simian immunodeficiency virus-infected female macaques. Physiol Genomics 2021; 53:358-371. [PMID: 34252326 DOI: 10.1152/physiolgenomics.00001.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Effective antiretroviral therapy (ART) has significantly reduced mortality of people living with HIV (PLWH), and the prevalence of at-risk alcohol use is higher among PLWH. Increased survival and aging of PLWH is associated with increased prevalence of metabolic comorbidities especially among menopausal women, and adipose tissue metabolic dysregulation may be a significant contributing factor. We examined the differential effects of chronic binge alcohol (CBA) administration and ovariectomy (OVX) on the omental adipose tissue (OmAT) proteome in a subset of simian immunodeficiency virus (SIV)-infected macaques of a longitudinal parent study. Quantitative discovery-based proteomics identified 1,429 differentially expressed proteins. Ingenuity Pathway Analysis (IPA) was used to calculate z-scores, or activation predictions, for functional pathways and diseases. Results revealed that protein changes associated with functional pathways centered around the "OmAT metaboproteome profile." Based on z-scores, CBA did not affect functional pathways of metabolic disease but dysregulated proteins involved in adenosine monophosphate-activated protein kinase (AMPK) signaling and lipid metabolism. OVX-mediated proteome changes were predicted to promote pathways involved in glucose- and lipid-associated metabolic disease. Proteins involved in apoptosis, necrosis, and reactive oxygen species (ROS) pathways were also predicted to be activated by OVX and these were predicted to be inhibited by CBA. These results provide evidence for the role of ovarian hormone loss in mediating OmAT metaboproteome dysregulation in SIV and suggest that CBA modifies OVX-associated changes. In the context of OVX, CBA administration produced larger metabolic and cellular effects, which we speculate may reflect a protective role of estrogen against CBA-mediated adipose tissue injury in female SIV-infected macaques.
Collapse
Affiliation(s)
- Jonquil M Poret
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Jessie J Guidry
- Department of Biochemistry and The Proteomic Core Facility, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Liz Simon
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Patricia E Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
12
|
Bourgeois C, Gorwood J, Olivo A, Le Pelletier L, Capeau J, Lambotte O, Béréziat V, Lagathu C. Contribution of Adipose Tissue to the Chronic Immune Activation and Inflammation Associated With HIV Infection and Its Treatment. Front Immunol 2021; 12:670566. [PMID: 34220817 PMCID: PMC8250865 DOI: 10.3389/fimmu.2021.670566] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
White adipose tissue (AT) contributes significantly to inflammation – especially in the context of obesity. Several of AT’s intrinsic features favor its key role in local and systemic inflammation: (i) large distribution throughout the body, (ii) major endocrine activity, and (iii) presence of metabolic and immune cells in close proximity. In obesity, the concomitant pro-inflammatory signals produced by immune cells, adipocytes and adipose stem cells help to drive local inflammation in a vicious circle. Although the secretion of adipokines by AT is a prime contributor to systemic inflammation, the lipotoxicity associated with AT dysfunction might also be involved and could affect distant organs. In HIV-infected patients, the AT is targeted by both HIV infection and antiretroviral therapy (ART). During the primary phase of infection, the virus targets AT directly (by infecting AT CD4 T cells) and indirectly (via viral protein release, inflammatory signals, and gut disruption). The initiation of ART drastically changes the picture: ART reduces viral load, restores (at least partially) the CD4 T cell count, and dampens inflammatory processes on the whole-body level but also within the AT. However, ART induces AT dysfunction and metabolic side effects, which are highly dependent on the individual molecules and the combination used. First generation thymidine reverse transcriptase inhibitors predominantly target mitochondrial DNA and induce oxidative stress and adipocyte death. Protease inhibitors predominantly affect metabolic pathways (affecting adipogenesis and adipocyte homeostasis) resulting in insulin resistance. Recently marketed integrase strand transfer inhibitors induce both adipocyte adipogenesis, hypertrophy and fibrosis. It is challenging to distinguish between the respective effects of viral persistence, persistent immune defects and ART toxicity on the inflammatory profile present in ART-controlled HIV-infected patients. The host metabolic status, the size of the pre-established viral reservoir, the quality of the immune restoration, and the natural ageing with associated comorbidities may mitigate and/or reinforce the contribution of antiretrovirals (ARVs) toxicity to the development of low-grade inflammation in HIV-infected patients. Protecting AT functions appears highly relevant in ART-controlled HIV-infected patients. It requires lifestyle habits improvement in the absence of effective anti-inflammatory treatment. Besides, reducing ART toxicities remains a crucial therapeutic goal.
Collapse
Affiliation(s)
- Christine Bourgeois
- CEA - Université Paris Saclay - INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Jennifer Gorwood
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Anaelle Olivo
- CEA - Université Paris Saclay - INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Laura Le Pelletier
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Jacqueline Capeau
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Olivier Lambotte
- CEA - Université Paris Saclay - INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,AP-HP, Groupe Hospitalier Universitaire Paris Saclay, Hôpital Bicêtre, Service de Médecine Interne et Immunologie Clinique, Le Kremlin-Bicêtre, France
| | - Véronique Béréziat
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Claire Lagathu
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| |
Collapse
|
13
|
Su J, Shiau S, Arpadi SM, Strehlau R, Burke M, Patel F, Kuhn L, Coovadia A, Yin MT. Switch to Efavirenz Attenuates Lipoatrophy in Girls With Perinatal HIV. J Pediatr Gastroenterol Nutr 2021; 72:e15-e20. [PMID: 32804904 PMCID: PMC8832869 DOI: 10.1097/mpg.0000000000002907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Children with HIV (CHIV) have lifetime exposure to antiretrovirals (ART); therefore, optimizing their regimens to have the least impact on fat redistribution is a priority. METHODS This is a cross-sectional study of 219 perinatally infected CHIV and 219 HIV-uninfected controls from similar socioeconomic backgrounds in Johannesburg, South Africa. We compared total body and regional fat distribution in CHIV on suppressive ART regimens with controls and, among CHIV, between ritonavir-boosted lopinavir (LPV/r)-based and efavirenz (EFV)-based regimens. RESULTS The mean age of the 219 uninfected children (45% girls) and the 219 CHIV (48% girls) was 7.0 and 6.4 years, respectively. CHIV had lower adjusted total body fat (P = 0.005) and lower percentage fat at the trunk (P = 0.020), arms (P = 0.001), and legs (P < 0.001) than uninfected children. CHIV on LPV/r had similar body composition as those on EFV, except for arm fat mass (P = 0.030). When stratified by sex, girls with HIV on LPV/r had lower adjusted total (P = 0.007), trunk (P = 0.002), arms (P = 0.008), legs (P = 0.048) fat mass; trunk-to-total body fat (P = 0.044); and higher legs-to-total body fat (P = 0.011) than those on EFV. CONCLUSIONS South African CHIV receiving ART had lower global and partial fat mass and percentage fat than healthy controls. In girls with HIV with sustained virologic suppression on ART, switching from LPV/r to EFV could attenuate fat mass loss, indicating that EFV-based regimen may be a better option in this group of individuals.
Collapse
Affiliation(s)
- Junwei Su
- The Department of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Stephanie Shiau
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ
| | - Stephen M Arpadi
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Renate Strehlau
- Empilweni Services and Research Unit, Department of Pediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital, University of the Witwatersrand, Johannesburg, South Africa
| | - Megan Burke
- Empilweni Services and Research Unit, Department of Pediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital, University of the Witwatersrand, Johannesburg, South Africa
| | - Faeezah Patel
- Empilweni Services and Research Unit, Department of Pediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital, University of the Witwatersrand, Johannesburg, South Africa
| | - Louise Kuhn
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Ashraf Coovadia
- Empilweni Services and Research Unit, Department of Pediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital, University of the Witwatersrand, Johannesburg, South Africa
| | - Michael T Yin
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
14
|
Dysfunctional Immunometabolism in HIV Infection: Contributing Factors and Implications for Age-Related Comorbid Diseases. Curr HIV/AIDS Rep 2020; 17:125-137. [PMID: 32140979 DOI: 10.1007/s11904-020-00484-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW An increasing body of evidence indicates that persons living with HIV (PLWH) display dysfunctional immunometabolism. Here, we provide an updated review of this topic and its relationship to HIV-associated immune stimuli and age-related disease. RECENT FINDINGS HIV infection alters immunometabolism by increasing reliance on aerobic glycolysis for energy and productive infection and repurposing oxidative phosphorylation machinery for immune cell proliferation and survival. Recent studies in PLWH with diabetes mellitus or cardiovascular disease have identified an association with elevated T cell and monocyte glucose metabolism, respectively. Immunometabolic dysfunction has also been observed in PLWH in frailty and additional studies suggest a role for immunometabolism in non-AIDS defining cancers and neurocognitive disease. There is a plethora of HIV-associated immune stimuli that could drive immunometabolic dysfunction and age-related disease in PLWH, but studies directly examining their relationship are lacking. Immunometabolic dysfunction is characteristic of HIV infection and is a potential link between HIV-associated stimuli and age-related comorbidities.
Collapse
|
15
|
Gelpi M, Ueland PM, Trøseid M, Mocroft A, Lebech AM, Ullum H, Midttun Ø, Lundgren J, Nielsen SD. Abdominal Adipose Tissue Is Associated With Alterations in Tryptophan-Kynurenine Metabolism and Markers of Systemic Inflammation in People With Human Immunodeficiency Virus. J Infect Dis 2020; 221:419-427. [PMID: 31538186 DOI: 10.1093/infdis/jiz465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/11/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND While both adipose tissue accumulation and tryptophan metabolism alterations are features of human immunodeficiency virus (HIV) infection, their interplay is unclear. We investigated associations between abdominal adipose tissue, alterations in kynurenine pathway of tryptophan metabolism, and systemic inflammation in people with HIV (PWH). METHODS Eight hundred sixty-four PWH and 75 uninfected controls were included. Plasma samples were collected and analyzed for kynurenine metabolites, neopterin, high-sensitivity C-reactive protein (hs-CRP), and lipids. Regression models were used to test associations in PWH. RESULTS PWH had higher kynurenine-to-tryptophan ratio than uninfected individuals (P < .001). In PWH, increase in waist-to-hip ratio was associated with higher kynurenine-to-tryptophan ratio (P = .009) and quinolinic-to-kynurenic acid ratio (P = .006) and lower kynurenic acid concentration (P = .019). Quinolinic-to-kynurenic acid ratio was associated with higher hs-CRP (P < .001) and neopterin concentrations (P < .001), while kynurenic acid was associated with lower hs-CRP (P = .025) and neopterin concentrations (P = .034). CONCLUSIONS In PWH, increase in abdominal adipose tissue was associated with increased quinolinic-to-kynurenic acid ratio, suggesting activation of proinflammatory pathway of kynurenine metabolism, with reduction of anti-inflammatory molecules and increase in systemic inflammation. Our results suggest dysregulation of kynurenine metabolism associated with abdominal fat accumulation to be a potential source of inflammation in HIV infection.
Collapse
Affiliation(s)
- Marco Gelpi
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Per Magne Ueland
- Section for Pharmacology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marius Trøseid
- Section of Clinical Immunology and Infectious Diseases, University Hospital Rikshospitalet, Oslo, Norway
| | - Amanda Mocroft
- HIV Epidemiology and Biostatistics Unit, Department of Infection and Population Health, University College London, London, United Kingdom
| | - Anne-Mette Lebech
- Department of Infectious Diseases, Hvidovre Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Henrik Ullum
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Øivind Midttun
- Section for Pharmacology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jens Lundgren
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Centre of Excellence for Health, Immunity and Infections, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Susanne D Nielsen
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Abstract
Early in the HIV epidemic, lipodystrophy, characterized by subcutaneous fat loss (lipoatrophy), with or without central fat accumulation (lipohypertrophy), was recognized as a frequent condition among people living with HIV (PLWH) receiving combination antiretroviral therapy. The subsequent identification of thymidine analogue nucleoside reverse transcriptase inhibitors as the cause of lipoatrophy led to the development of newer antiretroviral agents; however, studies have demonstrated continued abnormalities in fat and/or lipid storage in PLWH treated with newer drugs (including integrase inhibitor-based regimens), with fat gain due to restoration to health in antiretroviral therapy-naive PLWH, which is compounded by the rising rates of obesity. The mechanisms of fat alterations in PLWH are complex, multifactorial and not fully understood, although they are known to result in part from the direct effects of HIV proteins and antiretroviral agents on adipocyte health, genetic factors, increased microbial translocation, changes in the adaptive immune milieu after infection, increased tissue inflammation and accelerated fibrosis. Management includes classical lifestyle alterations with a role for pharmacological therapies and surgery in some patients. Continued fat alterations in PLWH will have an important effect on lifespan, healthspan and quality of life as patients age worldwide, highlighting the need to investigate the critical uncertainties regarding pathophysiology, risk factors and management.
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW The proportion of overweight and obese persons with HIV (PWH) has increased since the introduction of antiretroviral therapy (ART). We aim to summarize recent literature on risks of weight gain, discuss adipose tissue changes in HIV and obesity, and synthesize current understanding of how excess adiposity and HIV contribute to metabolic complications. RECENT FINDINGS Recent studies have implicated contemporary ART regimens, including use of integrase strand transfer inhibitors and tenofovir alafenamide, as a contributor to weight gain, though the mechanisms are unclear. Metabolic dysregulation is linked to ectopic fat and alterations in adipose immune cell populations that accompany HIV and obesity. These factors contribute to an increasing burden of metabolic diseases in the aging HIV population. Obesity compounds an increasing burden of metabolic disease among PWH, and understanding the role of fat partitioning and HIV- and ART-related adipose tissue dysfunction may guide prevention and treatment strategies.
Collapse
Affiliation(s)
- Samuel S Bailin
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN, 37232-2582, USA
| | - Curtis L Gabriel
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Celestine N Wanjalla
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN, 37232-2582, USA
| | - John R Koethe
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN, 37232-2582, USA.
| |
Collapse
|
18
|
Gojanovich GS, Shikuma CM, Milne C, Libutti DE, Chow DC, Gerschenson M. Subcutaneous Adipocyte Adenosine Triphosphate Levels in HIV Infected Patients. AIDS Res Hum Retroviruses 2020; 36:75-82. [PMID: 31407586 DOI: 10.1089/aid.2019.0121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lipoatrophy, or fat wasting, remains a syndrome plaguing HIV+ patients receiving antiretroviral (ARV) therapy. Both HIV infection per se and certain ARV are associated with lowered adipose tissue mitochondrial deoxyribonucleic acid (mtDNA) and mitochondrial ribonucleic acid (mtRNA) levels, but effects on adenosine triphosphate (ATP) production are unclear. We hypothesized that such alterations would accompany lowering of ATP levels in fat of HIV+ patients and would be worse in those displaying lipoatrophy. Gluteal-fold, subcutaneous adipose tissue was obtained from HIV seronegative control patients, from HIV+ ARV-naive patients, and those on ARV with or without lipoatrophy. Cellular ATP was measured in isolated adipocytes and preadipocyte fraction cells by bioluminescence. mtDNA copies/cell and oxidative phosphorylation (OXPHOS) mtRNA transcripts were evaluated by quantitative polymerase chain reactions. ATP levels were consistently higher in preadipocyte fraction cells than adipocytes, but values strongly correlated with each other (r = 0.66, p < .001). ATP levels in adipocytes were higher in both ARV-naive and nonlipoatrophic HIV+ patients compared to seronegative controls, but significantly lower in adipocytes and preadipocytes of lipoatrophic versus other HIV+ patients. Fat mtDNA copies/cell and OXPHOS mtRNA transcripts were lower in lipoatrophic patient samples compared to HIV seronegative. The ratio of specific OXPHOS transcripts to each other was significantly higher in nonlipoatrophic patients versus all groups, and this ratio correlated significantly with ATP levels in adipocytes. Thus, HIV infection is associated with an increase in adipose tissue ATP stores. Decreases in adipose mtDNA and OXPHOS mtRNA are found in those with HIV on ARV; however, ATP level is effected only in patients displaying lipoatrophy.
Collapse
Affiliation(s)
- Greg S. Gojanovich
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Cecilia M. Shikuma
- Department of Medicine, Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Cris Milne
- Department of Medicine, Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Daniel E. Libutti
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Dominic C. Chow
- Department of Medicine, Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
- Department of Pediatrics, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| |
Collapse
|
19
|
Yadav A, Kossenkov AV, Knecht VR, Showe LC, Ratcliffe SJ, Montaner LJ, Tebas P, Collman RG. Evidence for Persistent Monocyte and Immune Dysregulation After Prolonged Viral Suppression Despite Normalization of Monocyte Subsets, sCD14 and sCD163 in HIV-Infected Individuals. Pathog Immun 2019; 4:324-362. [PMID: 31893252 PMCID: PMC6930814 DOI: 10.20411/pai.v4i2.336] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 11/21/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND People living with HIV on antiretroviral therapy (HIV/ART) experience excess non-AIDS comorbidities, and also remain at increased risk for certain infections and viral malignancies. Monocytes/macrophages are central to many of these comorbidities, and elevated plasma cytokines and immune activation during untreated infection are often incompletely reversed by ART and are also associated with comorbidities. METHODS We investigated monocyte surface markers, gene expression, and plasma cytokines in 11 HIV-infected older individuals (median 53 years) who started therapy with low CD4 counts (median 129 cells/µl), with elevated hsCRP (≥ 2mg/L) despite long-term ART (median 7.4 years), along with matched controls. RESULTS Frequency of monocyte subsets (based on CD14/CD16/CD163), were not different from controls, but surface expression of CD163 was increased (P = 0.021) while PD1 was decreased (P = 0.013) along with a trend for higher tissue factor (P = 0.096). As a group, HIV/ART participants had elevated plasma CCL2 (MCP-1; P = 0.0001), CXCL9 (MIG; P = 0.04), and sIL2R (P = 0.015), which were correlated, while sCD14 was not elevated. Principal component analysis of soluble markers revealed that 6/11 HIV/ART participants clustered with controls, while 5 formed a distinct group, driven by IL-10, CCL11, CXCL10, CCL2, CXCL9, and sIL2R. These individuals were significantly older than those clustering with controls. Transcriptomic analysis revealed multiple genes linked to immune functions including inflammation, immune cell development, and cell-cell signaling that were downregulated in HIV/ART monocytes and distinct from patterns in untreated subjects. CONCLUSIONS Long-term ART-treated individuals normalize monocyte subsets but exhibit immune dysregulation involving both aberrant inflammation and monocyte dysfunction, as well as inter-individual heterogeneity, suggesting complex mechanisms linking monocytes and HIV/ART comorbidities.
Collapse
Affiliation(s)
- Anjana Yadav
- Department of Medicine; University of Pennsylvania Perelman School of Medicine; Philadelphia, Pennsylvania
| | | | - Vincent R Knecht
- Department of Medicine; University of Pennsylvania Perelman School of Medicine; Philadelphia, Pennsylvania
| | | | - Sarah J Ratcliffe
- Department of Biostatistics and Epidemiology; University of Pennsylvania Perelman School of Medicine; Philadelphia, Pennsylvania
| | | | - Pablo Tebas
- Department of Medicine; University of Pennsylvania Perelman School of Medicine; Philadelphia, Pennsylvania
| | - Ronald G Collman
- Department of Medicine; University of Pennsylvania Perelman School of Medicine; Philadelphia, Pennsylvania
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW We aim to provide an in-depth review of recent literature highlighting the role of inflammation involving the adipose tissue, liver, skeletal muscles, and gastrointestinal tract in the development of metabolic complications among persons living with HIV (PLWH). RECENT FINDINGS Recent studies in PLWH have demonstrated a significant association between circulating inflammatory markers and development of insulin resistance and metabolic complications. In adipose tissue, pro-inflammatory cytokine expression inhibits adipocyte insulin signaling, which alters lipid and glucose homeostasis. Increased lipolysis and lipogenesis elevate levels of circulating free fatty acids and promote ectopic fat deposition in liver and skeletal muscles. This leads to lipotoxicity characterized by a pro-inflammatory response with worsening insulin resistance. Finally, HIV is associated with gastrointestinal tract inflammation and changes in the gut microbiome resulting in reduced diversity, which is an additional risk factor for diabetes. Metabolic complications in PLWH are in part due to chronic, multisite tissue inflammation resulting in dysregulation of glucose and lipid trafficking, utilization, and storage.
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to examine the evidence describing adipose tissue as a reservoir for HIV-1 and how this often expansive anatomic compartment contributes to HIV persistence. RECENT FINDINGS Memory CD4 T cells and macrophages, the major host cells for HIV, accumulate in adipose tissue during HIV/SIV infection of humans and rhesus macaques. Whereas HIV and SIV proviral DNA is detectable in CD4 T cells of multiple fat depots in virtually all infected humans and monkeys examined, viral RNA is less frequently detected, and infected macrophages may be less prevalent in adipose tissue. However, based on viral outgrowth assays, adipose-resident CD4 T cells are latently infected with virus that is replication-competent and infectious. Additionally, adipocytes interact with CD4 T cells and macrophages to promote immune cell activation and inflammation which may be supportive for HIV persistence. Antiviral effector cells, such as CD8 T cells and NK/NKT cells, are abundant in adipose tissue during HIV/SIV infection and typically exceed CD4 T cells, whereas B cells are largely absent from adipose tissue of humans and monkeys. Additionally, CD8 T cells in adipose tissue of HIV patients are activated and have a late differentiated phenotype, with unique TCR clonotypes of less diversity relative to blood CD8 T cells. With respect to the distribution of antiretroviral drugs in adipose tissue, data is limited, but there may be class-specific penetration of fat depots. The trafficking of infected immune cells within adipose tissues is a common event during HIV/SIV infection of humans and monkeys, but the virus may be mostly transcriptionally dormant. Viral replication may occur less in adipose tissue compared to other major reservoirs, such as lymphoid tissue, but replication competence and infectiousness of adipose latent virus are comparable to other tissues. Due to the ubiquitous nature of adipose tissue, inflammatory interactions among adipocytes and CD4 T cells and macrophages, and selective distribution of antiretroviral drugs, the sequestration of infected immune cells within fat depots likely represents a major challenge for cure efforts.
Collapse
Affiliation(s)
- Jacob Couturier
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St., MSB 2.112, Houston, TX, 77030, USA
| | - Dorothy E Lewis
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St., MSB 2.112, Houston, TX, 77030, USA.
| |
Collapse
|
22
|
Nicu C, Hardman JA, Pople J, Paus R. Do human dermal adipocytes switch from lipogenesis in anagen to lipophagy and lipolysis during catagen in the human hair cycle? Exp Dermatol 2019; 28:432-435. [PMID: 30776154 DOI: 10.1111/exd.13904] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/14/2019] [Accepted: 01/31/2019] [Indexed: 12/13/2022]
Abstract
In murine skin, dermal white adipose tissue (DWAT) undergoes fluctuations in size across the hair cycle, whereas changes in size, function and metabolism of dermal adipocytes (DAs) during the human scalp hair cycle remain unexplored. Transmission electron microscopy results suggest that during anagen-catagen transition, human DAs co-opt the autophagy machinery to undergo lipophagy within their lipid droplets. Whole-mount staining of hair follicles (HFs) and surrounding DWAT for the autophagy marker LC3B confirms the increased presence of LC3B+ lipid droplets adjacent to catagen HFs; moreover, DWAT around catagen HFs engages in greater glycerol release compared to DWAT surrounding anagen HFs. Thus, we hypothesize that human DAs switch from lipogenesis during anagen to lipophagy together with lipolysis during catagen. We propose various experiments to further prove this hypothesis, whose systematic exploration should help to better characterize the functions of human DWAT and its communication with the HF.
Collapse
Affiliation(s)
- Carina Nicu
- Centre for Dermatology Research, The University of Manchester, Manchester, UK.,NIHR Manchester Biomedical Research Centre and Manchester Academic Health Science Centre, Manchester, UK
| | - Jonathan Alan Hardman
- Centre for Dermatology Research, The University of Manchester, Manchester, UK.,NIHR Manchester Biomedical Research Centre and Manchester Academic Health Science Centre, Manchester, UK
| | | | - Ralf Paus
- Centre for Dermatology Research, The University of Manchester, Manchester, UK.,NIHR Manchester Biomedical Research Centre and Manchester Academic Health Science Centre, Manchester, UK.,Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
23
|
Relationship of visceral and subcutaneous adipose depots to markers of arterial injury and inflammation among individuals with HIV. AIDS 2019; 33:229-236. [PMID: 30325779 DOI: 10.1097/qad.0000000000002060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Persons living with HIV (PLWH) well treated on antiretroviral therapies remain at risk for ensuing arterial disease. We investigated the relationship between adipose depots and biomarkers of arterial injury and inflammation to gain insight into the link between body composition and CVD risk. DESIGNS/METHODS One hundred and fifty-five HIV-infected and 70 non-HIV infected individuals were well phenotyped for body composition. Adipose depots were assessed via single-slice abdominal computed tomography (CT). Circulating markers of arterial disease and generalized inflammation [lipoprotein-associated phospholipase A2 (LpPLA2), oxidized low-density lipoprotein (oxLDL), high-sensitivity cardiac troponin T (hs-cTnT), high-sensitivity C-reactive protein (hsCRP)] were evaluated. RESULTS Despite similar BMI and visceral adipose tissue (VAT), HIV-infected individuals had significantly lower subcutaneous adipose tissue [SAT, 199 (126-288) vs. 239 (148-358) cm(2), P = 0.04] than non-HIV infected individuals. Among HIV-infected individuals, reduced SAT inversely correlated with LpPLA2 (ρ = -0.19, P = 0.02) and hs-cTnT (ρ = -0.24, P = 0.004), whereas increased VAT significantly and positively related to LpPLA2 (ρ = 0.25, P = 0.003), oxLDL (ρ = 0.28, P = 0.0005), hs-cTnT (ρ = 0.28, P = 0.0007) and hsCRP (ρ = 0.32, P = < 0.0001). Similar analyses among the non-HIV infected individuals revealed significant relationships between SAT and LpPLA2 (ρ = -0.24, P = 0.05), as well as VAT and LpPLA2 (ρ = 0.37, P = 0.002), oxLDL (ρ = 0.24, P = 0.05) and hsCRP (ρ = 0.29, P = .02). In modelling performed among the HIV group, simultaneously controlling for VAT, SAT, age and relevant HIV-related parameters, reduced SAT was an independent predictor of LpPLA2 (P = 0.04) and hs-cTnT (P = 0.005) and increased VAT was an independent predictor of LpPLA2 (P = 0.001), oxLDL (P = 0.02), hs-cTnT (P = 0.04) and hsCRP (P = 0.04). CONCLUSION Fat redistribution phenotypes, characterized by SAT loss and/or VAT accumulation, may be linked to arterial injury and inflammation in HIV.
Collapse
|
24
|
Adipose Tissue is Enriched for Activated and Late-Differentiated CD8+ T Cells and Shows Distinct CD8+ Receptor Usage, Compared With Blood in HIV-Infected Persons. J Acquir Immune Defic Syndr 2018; 77:e14-e21. [PMID: 29040163 DOI: 10.1097/qai.0000000000001573] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Adverse viral and medication effects on adipose tissue contribute to the development of metabolic disease in HIV-infected persons, but T cells also have a central role modulating local inflammation and adipocyte function. We sought to characterize potentially proinflammatory T-cell populations in adipose tissue among persons on long-term antiretroviral therapy and assess whether adipose tissue CD8 T cells represent an expanded, oligoclonal population. METHODS We recruited 10 HIV-infected, non-diabetic, overweight or obese adults on efavirenz, tenofovir, and emtricitabine for >4 years with consistent viral suppression. We collected fasting blood and subcutaneous abdominal adipose tissue to measure the percentage of CD4 and CD8 T cells expressing activation, exhaustion, late differentiation/senescence, and memory surface markers. We performed T-cell receptor (TCR) sequencing on sorted CD8 cells. We compared the proportion of each T-cell subset and the TCR repertoire diversity, in blood versus adipose tissue. RESULTS Adipose tissue had a higher percentage of CD3CD8 T cells compared with blood (61.0% vs. 51.7%, P < 0.01) and was enriched for both activated CD8HLA-DR T cells (5.5% vs. 0.9%, P < 0.01) and late-differentiated CD8CD57 T cells (37.4% vs. 22.7%, P < 0.01). Adipose tissue CD8 T cells displayed distinct TCRβ V and J gene usage, and the Shannon Entropy index, a measure of overall TCRβ repertoire diversity, was lower compared with blood (4.39 vs. 4.46; P = 0.05). CONCLUSIONS Adipose tissue is enriched for activated and late-differentiated CD8 T cells with distinct TCR usage. These cells may contribute to tissue inflammation and impaired adipocyte fitness in HIV-infected persons.
Collapse
|
25
|
Wanjalla CN, McDonnell WJ, Koethe JR. Adipose Tissue T Cells in HIV/SIV Infection. Front Immunol 2018; 9:2730. [PMID: 30559739 PMCID: PMC6286992 DOI: 10.3389/fimmu.2018.02730] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue comprises one of the largest organs in the body and performs diverse functions including energy storage and release, regulation of appetite and other neuroendocrine signaling, and modulation of immuity, among others. Adipocytes reside in a complex compartment where antigen, antigen presenting cells, innate immune cells, and adaptive immune cells interact locally and exert systemic effects on inflammation, circulating immune cell profiles, and metabolic homeostasis. T lymphocytes are a major component of the adipose tissue milieu which are altered in disease states such as obesity and human immunodeficiency virus (HIV) infection. While obesity, HIV infection, and simian immunodeficiency virus (SIV; a non-human primate virus similar to HIV) infection are accompanied by enrichment of CD8+ T cells in the adipose tissue, major phenotypic differences in CD4+ T cells and other immune cell populations distinguish HIV/SIV infection from obesity. Furthermore, DNA and RNA species of HIV and SIV can be detected in the stromal vascular fraction of visceral and subcutaneous adipose tissue, and replication-competent HIV resides in local CD4+ T cells. Here, we review studies of adipose tissue CD4+ and CD8+ T cell populations in HIV and SIV, and contrast the findings with those reported in obesity.
Collapse
Affiliation(s)
- Celestine N Wanjalla
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States.,Center for Translational Immunology and Infectious Disease, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Wyatt J McDonnell
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States.,Center for Translational Immunology and Infectious Disease, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, United States
| | - John R Koethe
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States.,Center for Translational Immunology and Infectious Disease, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
26
|
Zanetti HR, Roever L, Gonçalves A, Resende ES. Human Immunodeficiency Virus Infection, Antiretroviral Therapy, and Statin: a Clinical Update. Curr Atheroscler Rep 2018; 20:9. [PMID: 29423787 DOI: 10.1007/s11883-018-0708-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW This clinical update is intended to focus in relationship between HIV infection and use of antiretroviral therapy (ART) and statin. RECENT FINDINGS Though ART significantly changed the course of HIV infection, it is related to numerous side effects principally to the lipid profile. In this way, statins became one of the most used lipid-lowering therapies in this population. In our clinical update, we evaluated studies that demonstrate the relationship and molecular mechanisms that HIV infection and ART use trigger dyslipidemia and also the use of statin to reduce this condition. We have demonstrated that use of statin can be used in dyslipidemic HIV-infected people as long as there is no drug interaction with ART. Recently, studies using rosuvastatin have shown greater effects when compared to the other statins.
Collapse
Affiliation(s)
- Hugo Ribeiro Zanetti
- Master Institute of Education President Antônio Carlos, Avenida Minas Gerais, 1889 - Centro, Araguari, MG, 38.440-046, Brazil. .,Department of Clinical Research, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - Leonardo Roever
- Department of Clinical Research, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Alexandre Gonçalves
- Master Institute of Education President Antônio Carlos, Avenida Minas Gerais, 1889 - Centro, Araguari, MG, 38.440-046, Brazil.,Department of Clinical Research, Federal University of Uberlândia, Uberlândia, MG, Brazil.,Paulista University, Brasília, DF, Brazil.,Atenas College, Paracatu, MG, Brazil
| | - Elmiro Santos Resende
- Department of Clinical Research, Federal University of Uberlândia, Uberlândia, MG, Brazil
| |
Collapse
|
27
|
Merino KM, Allers C, Didier ES, Kuroda MJ. Role of Monocyte/Macrophages during HIV/SIV Infection in Adult and Pediatric Acquired Immune Deficiency Syndrome. Front Immunol 2017; 8:1693. [PMID: 29259605 PMCID: PMC5723290 DOI: 10.3389/fimmu.2017.01693] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/16/2017] [Indexed: 12/17/2022] Open
Abstract
Monocytes/macrophages are a diverse group of cells that act as first responders in innate immunity and then as mediators for adaptive immunity to help clear infections. In performing these functions, however, the macrophage inflammatory responses can also contribute to pathogenesis. Various monocyte and tissue macrophage subsets have been associated with inflammatory disorders and tissue pathogeneses such as occur during HIV infection. Non-human primate research of simian immunodeficiency virus (SIV) has been invaluable in better understanding the pathogenesis of HIV infection. The question of HIV/SIV-infected macrophages serving as a viral reservoir has become significant for achieving a cure. In the rhesus macaque model, SIV-infected macrophages have been shown to promote pathogenesis in several tissues resulting in cardiovascular, metabolic, and neurological diseases. Results from human studies illustrated that alveolar macrophages could be an important HIV reservoir and humanized myeloid-only mice supported productive HIV infection and viral persistence in macrophages during ART treatment. Depletion of CD4+ T cells is considered the primary cause for terminal progression, but it was reported that increasing monocyte turnover was a significantly better predictor in SIV-infected adult macaques. Notably, pediatric cases of HIV/SIV exhibit faster and more severe disease progression than adults, yet neonates have fewer target T cells and generally lack the hallmark CD4+ T cell depletion typical of adult infections. Current data show that the baseline blood monocyte turnover rate was significantly higher in neonatal macaques compared to adults and this remained high with disease progression. In this review, we discuss recent data exploring the contribution of monocytes and macrophages to HIV/SIV infection and progression. Furthermore, we highlight the need to further investigate their role in pediatric cases of infection.
Collapse
Affiliation(s)
- Kristen M. Merino
- Division of Immunology, Tulane National Primate Research Center, Covington LA, United States
| | - Carolina Allers
- Division of Immunology, Tulane National Primate Research Center, Covington LA, United States
| | - Elizabeth S. Didier
- Division of Microbiology, Tulane National Primate Research Center, Covington LA, United States
| | - Marcelo J. Kuroda
- Division of Immunology, Tulane National Primate Research Center, Covington LA, United States
| |
Collapse
|
28
|
Abstract
HIV infection and antiretroviral therapy (ART) treatment exert diverse effects on adipocytes and stromal-vascular fraction cells, leading to changes in adipose tissue quantity, distribution, and energy storage. A HIV-associated lipodystrophic condition was recognized early in the epidemic, characterized by clinically apparent changes in subcutaneous, visceral, and dorsocervical adipose depots. Underlying these changes is altered adipose tissue morphology and expression of genes central to adipocyte maturation, regulation, metabolism, and cytokine signaling. HIV viral proteins persist in circulation and locally within adipose tissue despite suppression of plasma viremia on ART, and exposure to these proteins impairs preadipocyte maturation and reduces adipocyte expression of peroxisome proliferator-activated receptor gamma (PPAR-γ) and other genes involved in cell regulation. Several early nucleoside reverse transcriptase inhibitor and protease inhibitor antiretroviral drugs demonstrated substantial adipocyte toxicity, including reduced mitochondrial DNA content and respiratory chain enzymes, reduced PPAR-γ and other regulatory gene expression, and increased proinflammatory cytokine production. Newer-generation agents, such as integrase inhibitors, appear to have fewer adverse effects. HIV infection also alters the balance of CD4+ and CD8+ T cells in adipose tissue, with effects on macrophage activation and local inflammation, while the presence of latently infected CD4+ T cells in adipose tissue may constitute a protected viral reservoir. This review provides a synthesis of the literature on how HIV virus, ART treatment, and host characteristics interact to affect adipose tissue distribution, immunology, and contribution to metabolic health, and adipocyte maturation, cellular regulation, and energy storage. © 2017 American Physiological Society. Compr Physiol 7:1339-1357, 2017.
Collapse
Affiliation(s)
- John R Koethe
- Division of Infectious Diseases, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
29
|
Mitchell BI, Byron MM, Ng RC, Chow DC, Ndhlovu LC, Shikuma CM. Elevation of Non-Classical (CD14+/lowCD16++) Monocytes Is Associated with Increased Albuminuria and Urine TGF-β1 in HIV-Infected Individuals on Stable Antiretroviral Therapy. PLoS One 2016; 11:e0153758. [PMID: 27097224 PMCID: PMC4838224 DOI: 10.1371/journal.pone.0153758] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 04/04/2016] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE High rates of albuminuria are observed among HIV-infected individuals on stable antiretroviral therapy (ART). Though pro-inflammatory and pro-fibrotic responses are described as components of albuminuria in the general population, it is unclear how these responses are associated to albuminuria in ART-treated chronic HIV. We investigated the relationship of monocyte subsets and urine inflammatory and fibrotic biomarkers to albuminuria in ART-treated HIV-infected participants. DESIGN AND METHODS Cross-sectional analyses were performed on Hawaii Aging with HIV-cardiovascular disease study cohort participants who were required at entry to be ≥40 years old and on ART ≥3 months. Monocyte subpopulations were determined in banked peripheral blood mononuclear cells (PBMC) using multi-parametric flow-cytometry. Entry random urine samples were assessed for albumin-to-creatinine ratios (UACR). Urine samples were measured for inflammatory and fibrotic biomarkers using Luminex technology. RESULTS Among 96 HIV-infected subjects with measured UACR (87% male, 59% Caucasian, and 89% undetectable HIV RNA with median CD4 of 495.5 cells/μL), 18 patients (19%) had albuminuria. Non-classical (CD14low/+CD16++) monocytes were significantly elevated in subjects with albuminuria (p = 0.034) and were correlated to UACR (r = 0.238, p = 0.019). Elevated non-classical monocyte counts were significant predictors of worsening albuminuria, independent of traditional- and ART-associated risk factors (β = 0.539, p = 0.007). Urine TGF-β1 and collagen-IV were significantly higher in albuminuric compared to non-albuminuric participants (TGF-β1; p = 0.039 and collagen-IV; p = 0.042). Urine TGF-β1 was significantly correlated with non-classical monocyte counts (r = 0.464, p = 0.017). CONCLUSION Alterations in monocyte subpopulations and urine pro-fibrotic factors may play a role in kidney dysfunction during chronic HIV infection and warrants further study.
Collapse
Affiliation(s)
- Brooks I. Mitchell
- Department of Tropical Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Hawaii Center for AIDS, University of Hawaii, Honolulu, Hawaii, United States of America
- * E-mail:
| | - Mary Margaret Byron
- Department of Tropical Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Roland C. Ng
- Department of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Dominic C. Chow
- Hawaii Center for AIDS, University of Hawaii, Honolulu, Hawaii, United States of America
- Department of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Lishomwa C. Ndhlovu
- Department of Tropical Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Hawaii Center for AIDS, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Cecilia M. Shikuma
- Department of Tropical Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Hawaii Center for AIDS, University of Hawaii, Honolulu, Hawaii, United States of America
- Department of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| |
Collapse
|
30
|
|
31
|
The meningeal lymphatic system: a route for HIV brain migration? J Neurovirol 2015; 22:275-81. [PMID: 26572785 DOI: 10.1007/s13365-015-0399-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/15/2015] [Accepted: 10/23/2015] [Indexed: 01/08/2023]
Abstract
Two innovative studies recently identified functional lymphatic structures in the meninges that may influence the development of HIV-associated neurological disorders (HAND). Until now, blood vessels were assumed to be the sole transport system by which HIV-infected monocytes entered the brain by bypassing a potentially hostile blood-brain barrier through inflammatory-mediated semi-permeability. A cascade of specific chemokine signals promote monocyte migration from blood vessels to surrounding brain tissues via a well-supported endothelium, where the cells differentiate into tissue macrophages capable of productive HIV infection. Lymphatic vessels on the other hand are more loosely organized than blood vessels. They absorb interstitial fluid from bodily tissues where HIV may persist and exchange a variety of immune cells (CD4(+) T cells, monocytes, macrophages, and dendritic cells) with surrounding tissues through discontinuous endothelial junctions. We propose that the newly discovered meningeal lymphatics are key to HIV migration among viral reservoirs and brain tissue during periods of undetectable plasma viral loads due to suppressive combinational antiretroviral therapy, thus redefining the migration process in terms of a blood-lymphatic transport system.
Collapse
|
32
|
Wright EJ, Grund B, Cysique LA, Robertson KR, Brew BJ, Collins G, Shlay JC, Winston A, Read TRH, Price RW. Factors associated with neurocognitive test performance at baseline: a substudy of the INSIGHT Strategic Timing of AntiRetroviral Treatment (START) trial. HIV Med 2015; 16 Suppl 1:97-108. [PMID: 25711328 DOI: 10.1111/hiv.12238] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2014] [Indexed: 11/29/2022]
Abstract
OBJECTIVES We describe neuropsychological test performance (NP) in antiretroviral treatment (ART)-naïve HIV-positive individuals with CD4 cell counts above 500 cells/μL. METHODS In a neurology substudy of the International Network for Strategic Initiatives in Global HIV Trials (INSIGHT) Strategic Timing of AntiRetroviral Treatment (START) study, eight neurocognitive tests were administered. The primary measure of NP was the quantitative NP z-score (QNPZ-8), the average of the z-scores for the eight tests. Associations of baseline factors with QNPZ-8 scores were assessed by multiple regression. Mild neurocognitive impairment (NCI) was defined as z-scores < -1 in at least two of six cognitive domains. RESULTS A total of 608 participants had a median age of 34 years; 11% were women and 15% were black; the median time since HIV diagnosis was 0.9 years; the median CD4 cell count was 633 cells/μL; 19.9% had mild NCI. Better NP was independently associated with younger age, being white, higher body mass index (0.10 per 10 kg/m(2) higher), and higher haematocrit percentage (0.19 per 10% higher). Worse NP was associated with longer time since HIV diagnosis (-0.17 per 10 years), diabetes (-0.29) and higher Framingham risk score (-0.15 per 10 points higher). QNPZ-8 scores differed significantly between geographical locations, with the lowest scores in Brazil and Argentina/Chile. CONCLUSIONS This is the largest study of NP in ART-naïve HIV-positive adults with CD4 counts > 500 cells/μL. Demographic factors and diabetes were most strongly associated with NP. Unmeasured educational/sociocultural factors may explain geographical differences. Poorer NP was independently associated with longer time since HIV diagnosis, suggesting that untreated HIV infection might deleteriously affect NP, but the effect was small.
Collapse
Affiliation(s)
- E J Wright
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria, Australia; Burnet Institute, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Damouche A, Lazure T, Avettand-Fènoël V, Huot N, Dejucq-Rainsford N, Satie AP, Mélard A, David L, Gommet C, Ghosn J, Noel N, Pourcher G, Martinez V, Benoist S, Béréziat V, Cosma A, Favier B, Vaslin B, Rouzioux C, Capeau J, Müller-Trutwin M, Dereuddre-Bosquet N, Le Grand R, Lambotte O, Bourgeois C. Adipose Tissue Is a Neglected Viral Reservoir and an Inflammatory Site during Chronic HIV and SIV Infection. PLoS Pathog 2015; 11:e1005153. [PMID: 26402858 PMCID: PMC4581628 DOI: 10.1371/journal.ppat.1005153] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 08/14/2015] [Indexed: 12/14/2022] Open
Abstract
Two of the crucial aspects of human immunodeficiency virus (HIV) infection are (i) viral persistence in reservoirs (precluding viral eradication) and (ii) chronic inflammation (directly associated with all-cause morbidities in antiretroviral therapy (ART)-controlled HIV-infected patients). The objective of the present study was to assess the potential involvement of adipose tissue in these two aspects. Adipose tissue is composed of adipocytes and the stromal vascular fraction (SVF); the latter comprises immune cells such as CD4+ T cells and macrophages (both of which are important target cells for HIV). The inflammatory potential of adipose tissue has been extensively described in the context of obesity. During HIV infection, the inflammatory profile of adipose tissue has been revealed by the occurrence of lipodystrophies (primarily related to ART). Data on the impact of HIV on the SVF (especially in individuals not receiving ART) are scarce. We first analyzed the impact of simian immunodeficiency virus (SIV) infection on abdominal subcutaneous and visceral adipose tissues in SIVmac251 infected macaques and found that both adipocytes and adipose tissue immune cells were affected. The adipocyte density was elevated, and adipose tissue immune cells presented enhanced immune activation and/or inflammatory profiles. We detected cell-associated SIV DNA and RNA in the SVF and in sorted CD4+ T cells and macrophages from adipose tissue. We demonstrated that SVF cells (including CD4+ T cells) are infected in ART-controlled HIV-infected patients. Importantly, the production of HIV RNA was detected by in situ hybridization, and after the in vitro reactivation of sorted CD4+ T cells from adipose tissue. We thus identified adipose tissue as a crucial cofactor in both viral persistence and chronic immune activation/inflammation during HIV infection. These observations open up new therapeutic strategies for limiting the size of the viral reservoir and decreasing low-grade chronic inflammation via the modulation of adipose tissue-related pathways.
Collapse
Affiliation(s)
- Abderaouf Damouche
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Thierry Lazure
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’anatomo-pathologie, Le Kremlin-Bicêtre, France
| | - Véronique Avettand-Fènoël
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, EA 7327, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Laboratoire de Virologie, Paris, France
| | - Nicolas Huot
- Institut Pasteur, Unité HIV, Inflammation et Persistance, Paris, France
| | | | - Anne-Pascale Satie
- INSERM, U1085-IRSET, Université de Rennes 1, Campus de Beaulieu, Rennes, France
| | - Adeline Mélard
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, EA 7327, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Laboratoire de Virologie, Paris, France
| | - Ludivine David
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, EA 7327, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Laboratoire de Virologie, Paris, France
| | | | - Jade Ghosn
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, EA 7327, Paris, France
| | - Nicolas Noel
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Bicêtre, Service de Médecine Interne et Immunologie clinique, Le Kremlin-Bicêtre, France
| | - Guillaume Pourcher
- Assistance Publique—Hôpitaux de Paris, Hôpital Béclère, Service de Chirurgie Viscérale Minimale invasive, Clamart, France
- INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Valérie Martinez
- Assistance Publique—Hôpitaux de Paris, Hôpital Antoine Béclère, Service de Médecine Interne et Immunologie clinique, Clamart, France
| | - Stéphane Benoist
- Assistance Publique—Hôpitaux de Paris, Hôpital Bicêtre, Service de Chirurgie générale et digestive, Le Kremlin-Bicêtre, France
| | - Véronique Béréziat
- INSERM UMR S938, CDR Saint-Antoine; Sorbonne Universités, UPMC Univ Paris 6, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Tenon, Service de Biochimie et Hormonologie; ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Antonio Cosma
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Benoit Favier
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Bruno Vaslin
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Christine Rouzioux
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, EA 7327, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Laboratoire de Virologie, Paris, France
| | - Jacqueline Capeau
- INSERM UMR S938, CDR Saint-Antoine; Sorbonne Universités, UPMC Univ Paris 6, Paris, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Tenon, Service de Biochimie et Hormonologie; ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
| | | | - Nathalie Dereuddre-Bosquet
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Roger Le Grand
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
| | - Olivier Lambotte
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
- Assistance Publique—Hôpitaux de Paris, Hôpital Bicêtre, Service de Médecine Interne et Immunologie clinique, Le Kremlin-Bicêtre, France
| | - Christine Bourgeois
- Université Paris Sud, UMR 1184, Le Kremlin-Bicêtre, France
- CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
- INSERM, U1184, Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre, France
- * E-mail:
| |
Collapse
|
34
|
The Molecular Signature of HIV-1-Associated Lipomatosis Reveals Differential Involvement of Brown and Beige/Brite Adipocyte Cell Lineages. PLoS One 2015; 10:e0136571. [PMID: 26305325 PMCID: PMC4549259 DOI: 10.1371/journal.pone.0136571] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 08/04/2015] [Indexed: 11/19/2022] Open
Abstract
Highly active antiretroviral therapy has remarkably improved quality of life of HIV-1-infected patients. However, this treatment has been associated with the so-called lipodystrophic syndrome, which conveys a number of adverse metabolic effects and morphological alterations. Among them, lipoatrophy of subcutaneous fat in certain anatomical areas and hypertrophy of visceral depots are the most common. Less frequently, lipomatous enlargements of subcutaneous fat at distinct anatomic areas occur. Lipomatous adipose tissue in the dorso-cervical area ("buffalo hump") has been associated with a partial white-to-brown phenotype transition and with increased cell proliferation, but, to date, lipomatous enlargements arising in other parts of the body have not been characterized. In order to establish the main molecular events associated with the appearance of lipomatosis in HIV-1 patients, we analyzed biopsies of lipomatous tissue from "buffalo hump" and from other anatomical areas in patients, in comparison with healthy subcutaneous adipose tissue, using a marker gene expression approach. Both buffalo-hump and non-buffalo-hump lipomatous adipose tissues exhibited similar patterns of non-compromised adipogenesis, unaltered inflammation, non-fibrotic phenotype and proliferative activity. Shorter telomere length, prelamin A accumulation and SA-β-Gal induction, reminiscent of adipocyte senescence, were also common to both types of lipomatous tissues. Buffalo hump biopsies showed expression of marker genes of brown adipose tissue (e.g. UCP1) and, specifically, of "classical" brown adipocytes (e.g. ZIC1) but not of beige/brite adipocytes. No such brown fat-related gene expression occurred in lipomatous tissues at other anatomical sites. In conclusion, buffalo hump and other subcutaneous adipose tissue enlargements from HIV-1-infected patients share a similar lipomatous character. However, a distorted induction of white-to-"classical brown adipocyte" phenotype appears unique of dorso-cervical lipomatosis. Thus, the insults caused by HIV-1 viral infection and/or antiretroviral therapy leading to lipomatosis are acting in a location- and adipocyte lineage-dependent manner.
Collapse
|
35
|
Kuan EL, Ivanov S, Bridenbaugh EA, Victora G, Wang W, Childs EW, Platt AM, Jakubzick CV, Mason RJ, Gashev AA, Nussenzweig M, Swartz MA, Dustin ML, Zawieja DC, Randolph GJ. Collecting lymphatic vessel permeability facilitates adipose tissue inflammation and distribution of antigen to lymph node-homing adipose tissue dendritic cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:5200-10. [PMID: 25917096 DOI: 10.4049/jimmunol.1500221] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/22/2015] [Indexed: 12/31/2022]
Abstract
Collecting lymphatic vessels (CLVs), surrounded by fat and endowed with contractile muscle and valves, transport lymph from tissues after it is absorbed into lymphatic capillaries. CLVs are not known to participate in immune responses. In this study, we observed that the inherent permeability of CLVs allowed broad distribution of lymph components within surrounding fat for uptake by adjacent macrophages and dendritic cells (DCs) that actively interacted with CLVs. Endocytosis of lymph-derived Ags by these cells supported recall T cell responses in the fat and also generated Ag-bearing DCs for emigration into adjacent lymph nodes (LNs). Enhanced recruitment of DCs to inflammation-reactive LNs significantly relied on adipose tissue DCs to maintain sufficient numbers of Ag-bearing DCs as the LN expanded. Thus, CLVs coordinate inflammation and immunity within adipose depots and foster the generation of an unexpected pool of APCs for Ag transport into the adjacent LN.
Collapse
Affiliation(s)
- Emma L Kuan
- Department of Gene and Cell Medicine, Graduate Program in Immunology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Stoyan Ivanov
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Eric A Bridenbaugh
- Division of Lymphatic Biology, Department of Systems Biology and Translational Medicine, Cardiovascular Research Institute, Texas A&M Health Science Center College of Medicine, Temple, TX 76504
| | - Gabriel Victora
- Program in Molecular Pathogenesis, Skirball Institute for Biomolecular Medicine, The Helen L. and Martin S. Kimmel Center for Biology and Medicine, New York University School of Medicine, New York, NY 10016
| | - Wei Wang
- Division of Lymphatic Biology, Department of Systems Biology and Translational Medicine, Cardiovascular Research Institute, Texas A&M Health Science Center College of Medicine, Temple, TX 76504
| | - Ed W Childs
- Department of Surgery, Cardiovascular Research Institute, Texas A&M Health Science Center College of Medicine, Temple, TX 76504
| | - Andrew M Platt
- Department of Gene and Cell Medicine, Graduate Program in Immunology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | | | - Robert J Mason
- Department of Medicine, National Jewish Health, Denver, CO 80206
| | - Anatoliy A Gashev
- Division of Lymphatic Biology, Department of Systems Biology and Translational Medicine, Cardiovascular Research Institute, Texas A&M Health Science Center College of Medicine, Temple, TX 76504
| | - Michel Nussenzweig
- Laboratory of Molecular Immunology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065; and
| | - Melody A Swartz
- Institute of Bioengineering, Swiss Federal Institute of Technology, Lausanne 1015, Switzerland
| | - Michael L Dustin
- Program in Molecular Pathogenesis, Skirball Institute for Biomolecular Medicine, The Helen L. and Martin S. Kimmel Center for Biology and Medicine, New York University School of Medicine, New York, NY 10016
| | - David C Zawieja
- Division of Lymphatic Biology, Department of Systems Biology and Translational Medicine, Cardiovascular Research Institute, Texas A&M Health Science Center College of Medicine, Temple, TX 76504
| | - Gwendalyn J Randolph
- Department of Gene and Cell Medicine, Graduate Program in Immunology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110;
| |
Collapse
|
36
|
Abstract
OBJECTIVE The objective of this study is to determine whether adipose tissue functions as a reservoir for HIV-1. DESIGN We examined memory CD4(+) T cells and HIV DNA in adipose tissue-stromal vascular fraction (AT-SVF) of five patients [four antiretroviral therapy (ART)-treated and one untreated]. To determine whether adipocytes stimulate CD4(+) T cells and regulate HIV production, primary human adipose cells were cocultured with HIV-infected CD4(+) T cells. METHODS AT-SVF T cells were studied by flow cytometry, and AT-SVF HIV DNA (Gag and Env) was examined by nested PCR and sequence analyses. CD4(+) T-cell activation and HIV production were measured by flow cytometry and ELISA. RESULTS AT-SVF CD3(+) T cells were activated (>60% CD69(+)) memory CD4(+) and CD8(+) T cells in uninfected and HIV-infected persons, but the AT-SVF CD4(+)/CD8(+) ratio was lower in HIV patients. HIV DNA (Gag and Env) was detected in AT-SVF of all five patients examined by nested PCR, comparably to other tissues [peripheral blood mononuclear cell (PBMC), lymph node or thymus]. In coculture experiments, adipocytes increased CD4(+) T-cell activation and HIV production approximately two to three-fold in synergy with gamma-chain cytokines interleukin (IL)-2, IL7 or IL15. These effects were mitigated by neutralizing antibodies against IL6 and integrin-α1β1. Adipocytes also enhanced T-cell viability. CONCLUSION Adipose tissues of ART-treated patients harbour activated memory CD4(+) T cells and HIV DNA. Adipocytes promote CD4(+) T-cell activation and HIV production in concert with intrinsic adipose factors. Adipose tissue may be an important reservoir for HIV.
Collapse
|
37
|
Egaña-Gorroño L, Martínez E, Pérez I, Escribà T, Domingo P, Gatell JM, Arnedo M. Contribution of genetic background and antiretroviral therapy to body fat changes in antiretroviral-naive HIV-infected adults. J Antimicrob Chemother 2014; 69:3076-84. [PMID: 25185137 DOI: 10.1093/jac/dku266] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES To evaluate the association of host genetics with changes in limb or trunk fat in a group of antiretroviral therapy (ART)-naive HIV-infected patients prospectively followed up according to the initiation and the type of ART. METHODS Fifty single nucleotide polymorphisms (SNPs) in 26 genes, associated with obesity, insulin resistance, lipid metabolism or lipodystrophy in previously published genetic studies, were assessed in ART-naive HIV-infected Caucasian patients divided into three groups: 24 (27%) did not start ART, 29 (32.6%) received zidovudine or stavudine and 36 (40.4%) received neither zidovudine nor stavudine in their initial regimen. Patients underwent body fat measurements (using dual-energy X-ray absorptiometry) at baseline and Month 12. A multivariate model using backward stepwise elimination was used to assess the influence of SNPs and baseline levels of non-genetic covariates on changes in limb or trunk fat. RESULTS The baseline characteristics were: 73% men, 17% coinfected with hepatitis C virus and/or hepatitis B virus, median age 37 years, median CD4+ T cell count 228/mm(3), median HIV-RNA 5.2 log copies/mL, median plasma glucose 85 mg/dL, median plasma insulin 9.1 IU/mL, median limb fat 5.6 kg and median trunk fat 7.0 kg. There were no baseline differences among the three groups except for the CD4+ T cell count. The decrease in limb fat was greater in the no-ART group relative to the other two groups (P < 0.05). The multivariate model showed associations of rs1801278 in IRS1 (P = 0.029, OR = 0.13), baseline viral load (P = 0.006; OR = 4.453) and baseline glucose levels (P = 0.008, OR = 0.926) with loss of limb fat, and rs2228671 in LDLR (P = 0.012, OR = 0.108), rs405509 in APOE (P = 0.048, OR = 0.205), baseline viral load (P = 0.005, OR = 0.186) and baseline CD4+ T cell count (P = 0.01, OR = 1.008) with gain of trunk fat. CONCLUSIONS Specific polymorphisms in IRS1 (limb fat loss) and LDLR and APOE (trunk fat gain) were identified as independent markers of fat changes irrespective of the initiation of ART and the type of ART and deserve further validation.
Collapse
Affiliation(s)
- L Egaña-Gorroño
- Group of Genomics and Pharmacogenomics, Retrovirology and Viral Immunopathology Laboratory, IDIBAPS, Barcelona, Spain
| | - E Martínez
- Department of Infectious Diseases, Hospital Clinic de Barcelona, University of Barcelona, Barcelona, Spain
| | - I Pérez
- Department of Infectious Diseases, Hospital Clinic de Barcelona, University of Barcelona, Barcelona, Spain
| | - T Escribà
- Group of Genomics and Pharmacogenomics, Retrovirology and Viral Immunopathology Laboratory, IDIBAPS, Barcelona, Spain
| | - P Domingo
- Department of Infectious Diseases, Hospital de Sant Pau, Barcelona, Spain
| | - J M Gatell
- Group of Genomics and Pharmacogenomics, Retrovirology and Viral Immunopathology Laboratory, IDIBAPS, Barcelona, Spain Department of Infectious Diseases, Hospital Clinic de Barcelona, University of Barcelona, Barcelona, Spain
| | - M Arnedo
- Group of Genomics and Pharmacogenomics, Retrovirology and Viral Immunopathology Laboratory, IDIBAPS, Barcelona, Spain
| |
Collapse
|
38
|
|