151
|
Chen X, Zheng Y, Tatsuoka C, Muzic RF, Okoye CC, O'Donnell JK, Zidar D, Avril N, Oliveira GH, Liu H, Bucher J, Machtay M, Yao M, Dorth JA. Chemoradiotherapy-related carotid artery inflammation in head and neck cancer patients quantified by [ 18F]FDG PET/CT. Oral Oncol 2019; 93:101-106. [PMID: 31109689 DOI: 10.1016/j.oraloncology.2019.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Radiotherapy (RT) is associated with an increased risk of cardiovascular disease (CVD), but little is known about the mechanism for vascular injury and methods for early detection. MATERIALS AND METHODS We conducted a prospective, pilot study of carotid artery inflammation using 18F-labeled 2-fluoro-2-deoxy-d-glucose ([18F]FDG) PET/CT imaging pre- and 3 months post-RT in head-and-neck cancer (HNC) patients. [18F]FDG uptake by the carotid arteries was measured by the maximum and mean target to background ratio (TBRMAX, TBRMEAN) and the mean partial volume corrected standardized uptake value (pvcSUVMEAN). RESULTS Of the 22 patients who completed both pre and post-RT scans, the majority (82%) had stage III or stage IV disease and received concurrent chemotherapy. TBRMAX, TBRMEAN, and pvcSUVMEAN were all significantly higher 3 months after RT versus before RT with mean difference values (95% CI; p-value) of 0.17 (0.1-0.25; 0.0001), 0.19 (0.12-0.25; 0.0001), and 0.31 g/ml (0.12-0.5; 0.002), respectively. Fifteen patients (68%) had HPV-positive tumors, which were associated with lower pre-RT [18F]FDG signal, but a greater increase in TBRMAX (19% vs 5%), TBRMEAN (21% vs 11%) and pvcSUVMEAN (20% increase vs 3% decrease), compared to HPV negativity. CONCLUSION There is a significant increase in carotid artery inflammation in HNC patients due to CRT that amounts to a degree that has previously been associated with higher risk for future CVD events. The subset of patients with HPV-positive tumors experienced the greatest increases in vascular inflammation due to CRT. Carotid [18F]FDG uptake may be an early biomarker of RT-related vascular injury.
Collapse
Affiliation(s)
- Xuguang Chen
- Department of Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yiran Zheng
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Curtis Tatsuoka
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Raymond F Muzic
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH, United States
| | - Christian C Okoye
- Department of Radiation Oncology, St. Bernards Medical Group, Jonesboro, AR, United States
| | - James K O'Donnell
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University Cleveland, OH, United States
| | - David Zidar
- Harrington Heart and Vascular Institute, Department of Medicine, University Hospitals, Case Western Reserve University, Cleveland, OH, United States
| | - Norbert Avril
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University Cleveland, OH, United States
| | - Guilherme H Oliveira
- Onco-Cardiology Program, and Advanced Heart Failure and Transplant Center, Harrington Heart and Vascular Institute, Department of Medicine, University Hospitals, Case Western Reserve University, Cleveland, OH, United States
| | - Hongyan Liu
- Department of Neurology, University Hospitals Cleveland Medical Center and Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Jessica Bucher
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Mitchell Machtay
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Min Yao
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Jennifer A Dorth
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, United States.
| |
Collapse
|
152
|
Ruscica M, Watts GF, Sirtori CR. PCSK9 in HIV infection: New opportunity or red herring? Atherosclerosis 2019; 284:216-217. [DOI: 10.1016/j.atherosclerosis.2019.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/24/2022]
|
153
|
Whelton SP, Deal JA, Zikusoka M, Jacobson LP, Sarkar S, Palella FJ, Kingsley L, Budoff M, Witt MD, Brown TT, Post WS. Associations between lipids and subclinical coronary atherosclerosis. AIDS 2019; 33:1053-1061. [PMID: 30946159 PMCID: PMC6457132 DOI: 10.1097/qad.0000000000002151] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Whether HIV modifies the relationship of serum lipids with coronary atherosclerosis and coronary plaque subtypes is uncertain. We examined the associations between traditional lipids and coronary atherosclerosis among HIV-infected (HIV+) and HIV-uninfected (HIV-) men. DESIGN The Multicenter AIDS Cohort Study is an observational cohort with a total of 429 HIV+ and 303 HIV- men who had non-contrast cardiac computed tomography performed to measure coronary artery calcium and coronary computed tomography angiography to measure coronary stenosis, coronary plaque presence, and composition. METHODS We used multivariable adjusted prevalence ratios to examine the relationship between the SD difference in each lipid parameter and coronary atherosclerosis. RESULTS Total cholesterol (TC)/HDL-cholesterol had the strongest associations with coronary atherosclerosis regardless of HIV status. Overall, lipid parameters were most strongly associated with the presence of mixed plaque, stenosis more than 50%, and coronary artery calcium for both HIV+ and HIV- men. HIV+ men had similar, but weaker associations, between lipid parameters and coronary atherosclerosis compared with HIV- men. The strongest association was between the TC/HDL-cholesterol and stenosis more than 50% for both HIV+ [prevalence ratios 1.25 per SD (95% confidence interval 1.07-1.43)] and HIV- men [prevalence ratios 1.46 per SD (95% confidence interval 1.08-1.85)]. CONCLUSION The associations between lipids and coronary atherosclerosis tended to be weaker for HIV+ compared with HIV- men, although TC/HDL had the strongest association for both HIV+ and HIV- men. A weaker association between lipid levels and coronary atherosclerosis for HIV+ men may contribute to the decreased discrimination of cardiovascular disease risk observed in HIV+ individuals.
Collapse
Affiliation(s)
- Seamus P. Whelton
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine. 600 North Wolfe Street, Baltimore, MD 21287
| | - Jennifer A. Deal
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health 2024 E. Monument St, Suite 2-700, Baltimore, MD 21205
| | - Michelle Zikusoka
- MedStar Health System, 5601 Lock Raven Boulevard, Baltimore, MD 21239
| | - Lisa P. Jacobson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health 2024 E. Monument St, Suite 2-700, Baltimore, MD 21205
| | - Sudipa Sarkar
- Division of Endocrinology, Department of Medicine, Johns Hopkins University School of Medicine. 600 North Wolfe Street, Baltimore, MD 21287
| | - Frank J. Palella
- Division of Infectious Diseases, Feinberg School of Medicine of Northwestern University. 645 N. Michigan Ave, Chicago, IL 60611
| | - Lawrence Kingsley
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health. 130 De Soto Street, Pittsburgh, PA 15261
| | - Matthew Budoff
- Division of HIV Medicine, Los Angeles Biomedical Research Institute at Harbor UCLA Medical Center. 1124 West Carson St. Torrance, CA 90502
| | - Mallory D. Witt
- Division of HIV Medicine, Los Angeles Biomedical Research Institute at Harbor UCLA Medical Center. 1124 West Carson St. Torrance, CA 90502
| | - Todd T. Brown
- Division of Endocrinology, Department of Medicine, Johns Hopkins University School of Medicine. 600 North Wolfe Street, Baltimore, MD 21287
| | - Wendy S. Post
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine. 600 North Wolfe Street, Baltimore, MD 21287
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health 2024 E. Monument St, Suite 2-700, Baltimore, MD 21205
| |
Collapse
|
154
|
Bertrand L, Méroth F, Tournebize M, Leda AR, Sun E, Toborek M. Targeting the HIV-infected brain to improve ischemic stroke outcome. Nat Commun 2019; 10:2009. [PMID: 31043599 PMCID: PMC6494822 DOI: 10.1038/s41467-019-10046-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 04/12/2019] [Indexed: 12/13/2022] Open
Abstract
HIV-associated cerebrovascular events remain highly prevalent even in the current era of antiretroviral therapy (ART). We hypothesize that low-level HIV replication and associated inflammation endure despite antiretroviral treatment and affect ischemic stroke severity and outcomes. Using the EcoHIV infection model and the middle cerebral artery occlusion as the ischemic stroke model in mice, we present in vivo analysis of the relationship between HIV and stroke outcome. EcoHIV infection increases infarct size and negatively impacts tissue and functional recovery. Ischemic stroke also results in an increase in EcoHIV presence in the affected regions, suggesting post-stroke reactivation that magnifies pro-inflammatory status. Importantly, ART with a high CNS penetration effectiveness (CPE) is more beneficial than low CPE treatment in limiting tissue injury and accelerating post-stroke recovery. These results provide potential insight for treatment of HIV-infected patients that are at risk of developing cerebrovascular disease, such as ischemic stroke.
Collapse
Affiliation(s)
- Luc Bertrand
- University of Miami Miller School of Medicine, Department of Biochemistry and Molecular Biology, Miami, FL, 33136, USA.
| | - Fannie Méroth
- University of Miami Miller School of Medicine, Department of Biochemistry and Molecular Biology, Miami, FL, 33136, USA
| | - Marie Tournebize
- University of Miami Miller School of Medicine, Department of Biochemistry and Molecular Biology, Miami, FL, 33136, USA
| | - Ana Rachel Leda
- University of Miami Miller School of Medicine, Department of Biochemistry and Molecular Biology, Miami, FL, 33136, USA
| | - Enze Sun
- University of Miami Miller School of Medicine, Department of Biochemistry and Molecular Biology, Miami, FL, 33136, USA
| | - Michal Toborek
- University of Miami Miller School of Medicine, Department of Biochemistry and Molecular Biology, Miami, FL, 33136, USA.
| |
Collapse
|
155
|
Bowman ER, Kulkarni M, Gabriel J, Cichon MJ, Riedl K, Belury MA, Lake JE, Richardson B, Cameron C, Cameron M, Koletar SL, Lederman MM, Sieg SF, Funderburg NT. Altered Lipidome Composition Is Related to Markers of Monocyte and Immune Activation in Antiretroviral Therapy Treated Human Immunodeficiency Virus (HIV) Infection and in Uninfected Persons. Front Immunol 2019; 10:785. [PMID: 31040846 PMCID: PMC6477036 DOI: 10.3389/fimmu.2019.00785] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/25/2019] [Indexed: 12/21/2022] Open
Abstract
Background: HIV infection and antiretroviral therapy (ART) have both been linked to dyslipidemia and increased cardiovascular disease (CVD) risk. Alterations in the composition of saturated (SaFA), monounsaturated (MUFA), and polyunsaturated (PUFA) fatty acids are related to inflammation and CVD progression in HIV-uninfected (HIV-) populations. The relationships among the lipidome and markers of monocyte and immune activation in HIV-infected (HIV+) individuals are not well understood. Methods: Concentrations of serum lipids and their fatty acid composition were measured by direct infusion-tandem mass spectrometry in samples from 20 ART-treated HIV+ individuals and 20 HIV- individuals. Results: HIV+ individuals had increased levels of free fatty acids (FFAs) with enrichment of SaFAs, including palmitic acid (16:0) and stearic acid (18:0), and these levels were directly associated with markers of monocyte (CD40, HLA-DR, TLR4, CD36) and serum inflammation (LBP, CRP). PUFA levels were reduced significantly in HIV+ individuals, and many individual PUFA species levels were inversely related to markers of monocyte activation, such as tissue factor, TLR4, CD69, and SR-A. Also in HIV+ individuals, the composition of lysophosphatidylcholine (LPC) was enriched for SaFAs; LPC species containing SaFAs were directly associated with IL-6 levels and monocyte activation. We similarly observed direct relationships between levels of SaFAs and inflammation in HIV uninfected individuals. Further, SaFA exposure altered monocyte subset phenotypes and inflammatory cytokine production in vitro. Conclusions: The lipidome is altered in ART-treated HIV infection, and may contribute to inflammation and CVD progression. Detailed lipidomic analyses may better assess CVD risk in both HIV+ and HIV- individuals than does traditional lipid profiling.
Collapse
Affiliation(s)
- Emily R Bowman
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH, United States
| | - Manjusha Kulkarni
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH, United States
| | - Janelle Gabriel
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH, United States
| | - Morgan J Cichon
- Personalized Food and Nutritional Metabolomics for Health Discovery Theme, The Ohio State University, Columbus, OH, United States
| | - Kenneth Riedl
- Personalized Food and Nutritional Metabolomics for Health Discovery Theme, The Ohio State University, Columbus, OH, United States
| | - Martha A Belury
- Department of Human Sciences, Ohio State University, Columbus, OH, United States
| | - Jordan E Lake
- Division of Infectious Diseases, Department of Medicine, The University of Texas Health Science Center, Houston, TX, United States
| | - Brian Richardson
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Cheryl Cameron
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
| | - Mark Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Susan L Koletar
- Division of Infectious Diseases, Department of Medicine, Ohio State University, Columbus, OH, United States
| | - Michael M Lederman
- Division of Infectious Diseases, Department of Internal Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH, United States
| | - Scott F Sieg
- Division of Infectious Diseases, Department of Internal Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH, United States
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH, United States
| |
Collapse
|
156
|
Mesquita EC, Coelho LE, Amancio RT, Veloso V, Grinsztejn B, Luz P, Bozza FA. Severe infection increases cardiovascular risk among HIV-infected individuals. BMC Infect Dis 2019; 19:319. [PMID: 30975092 PMCID: PMC6460818 DOI: 10.1186/s12879-019-3894-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 03/11/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The identification and management of cardiovascular risk factors became a major clinical issue among HIV-infected individuals in the post-cART era. As in the past decades the link between acute infections and cardiovascular diseases became clear in the general population, we sorted to investigate the role of severe infections on incident cardiovascular diseases (CVDs) among HIV-infected individuals. METHODS HIV-infected individuals aged ≥18 years, with no history of CVD were followed from January 2000 to December 2013 until the occurrence of the first CVD event, death or end of study, whichever occurred first. To explore the effect of severe infections on the incidence of CVD we used extended Cox regression models and stratified post-hospitalization follow-up time into three periods: < 3 months, 3-12 months and > 12 months post discharge. RESULTS One hundred-eighty four persons from 3384 HIV-infected individuals developed incident CVD events during the follow-up (incidence rate = 11.10/1000 PY (95%CI: 9.60-12.82)). Risk of an incident CVD was 4-fold higher at < 3 months post-hospitalization for severe infections (adjusted hazard ratio [aHR], 4.52; 95% confidence interval [CI] 2.46-8.30), after adjusting for sociodemographic and clinical factors as well as comorbidities. This risk remained significant up to one year (3-12 months post hospital discharge aHR 2.39, 95% CI 1.30-4.38). Additionally, non-white race/ethnicity (aHR 1.49, 95% CI 1.10-2.02), age ≥ 60 years (aHR 2.01, 95% CI 1.01-3.97) and hypertension (aHR 1.90, 95% CI 1.38-2.60) were associated with an increased risk of CVD events. High CD4 (≥ 500 cells/mm3: aHR 0.41, 95% CI 0.27-0.62) and cART use (aHR 0.21, 95% CI 0.14-0.31) reduced the risk of CVD events. CONCLUSIONS We provide evidence for a time-dependent association between severe infection and incident cardiovascular disease in HIV-infected individuals. cART use, and high CD4 count were significantly associated with reduced hazards of CVD.
Collapse
Affiliation(s)
- Emersom Cicilini Mesquita
- Laboratório de Pesquisa Clínica em Medicina Intensiva, Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Lara Esteves Coelho
- Laboratório de HIV, Instituto de Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Rodrigo Teixeira Amancio
- Laboratório de Pesquisa Clínica em Medicina Intensiva, Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Valdilea Veloso
- Laboratório de HIV, Instituto de Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Beatriz Grinsztejn
- Laboratório de HIV, Instituto de Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Paula Luz
- Laboratório de HIV, Instituto de Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Fernando Augusto Bozza
- Laboratório de Pesquisa Clínica em Medicina Intensiva, Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
- Instituto D’Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, Brazil
| |
Collapse
|
157
|
Lawal IO, Ankrah AO, Stoltz AC, Sathekge MM. Radionuclide imaging of inflammation in atherosclerotic vascular disease among people living with HIV infection: current practice and future perspective. Eur J Hybrid Imaging 2019; 3:5. [PMID: 34191183 PMCID: PMC8218042 DOI: 10.1186/s41824-019-0053-7] [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: 01/08/2019] [Accepted: 03/11/2019] [Indexed: 01/03/2023] Open
Abstract
People living with human immunodeficiency virus (HIV) infection have twice the risk of atherosclerotic vascular disease compared with non-infected individuals. Inflammation plays a critical role in the development and progression of atherosclerotic vascular disease. Therapies targeting inflammation irrespective of serum lipid levels have been shown to be effective in preventing the occurrence of CVD. Radionuclide imaging is a viable method for evaluating arterial inflammation. This evaluation is useful in quantifying CVD risk and for assessing the effectiveness of anti-inflammatory treatment. The most tested radionuclide method for quantifying arterial inflammation among people living with HIV infection has been with F-18 FDG PET/CT. The level of arterial uptake of F-18 FDG correlates with vascular inflammation and with the risk of development and progression of atherosclerotic disease. Several limitations exist to the use of F-18 FDG for PET quantification of arterial inflammation. Many targets expressed on macrophage, a significant player in arterial inflammation, have the potential for use in evaluating arterial inflammation among people living with HIV infection. The review describes the clinical utility of F-18 FDG PET/CT in assessing arterial inflammation as a risk for atherosclerotic disease among people living with HIV infection. It also outlines potential newer probes that may quantify arterial inflammation in the HIV-infected population by targeting different proteins expressed on macrophages.
Collapse
Affiliation(s)
- Ismaheel O. Lawal
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
| | - Alfred O. Ankrah
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen & University of Groningen, Groningen, The Netherlands
| | - Anton C. Stoltz
- Infectious Disease Unit, Department of Internal Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
| | - Mike M. Sathekge
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
| |
Collapse
|
158
|
Subramanya V, McKay HS, Brusca RM, Palella FJ, Kingsley LA, Witt MD, Hodis HN, Tracy RP, Post WS, Haberlen SA. Inflammatory biomarkers and subclinical carotid atherosclerosis in HIV-infected and HIV-uninfected men in the Multicenter AIDS Cohort Study. PLoS One 2019; 14:e0214735. [PMID: 30946765 PMCID: PMC6448851 DOI: 10.1371/journal.pone.0214735] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/19/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND HIV-infected persons have an increased risk of atherosclerosis relative to uninfected individuals. Inflammatory processes may contribute to this risk. We evaluated the associations of 10 biomarkers of systemic inflammation (CRP, IL-6, sTNF-αR1 and 2), monocyte activation (CCL2, sCD163, sCD14), coagulation (fibrinogen, D-dimer), and endothelial dysfunction (ICAM-1) with subclinical carotid atherosclerosis among participants in the Multicenter AIDS Cohort Study (MACS). METHODS Carotid plaque and intima media thickness (IMT) in the common carotid (CCA-IMT) and bifurcation region were assessed by B mode ultrasound among 452 HIV-infected and 276 HIV-uninfected men from 2010-2013. Associations between levels of each biomarker and presence of focal plaque and IMT were assessed by logistic and linear regression models, adjusting for demographics, risk behaviors, traditional cardiovascular disease (CVD) risk factors, and HIV disease characteristics. RESULTS Compared to HIV-uninfected men, HIV-infected men had significantly higher levels of 8 of the 10 biomarkers. Overall, men with sCD163, CCL2, IL-6, and CRP levels in the highest quintile had approximately 2 times the odds of carotid plaque relative to those with levels in the lowest quintile, independent of demographic and CVD risk factors. Fibrinogen levels were positively associated with CCA-IMT while ICAM-1, CCL2, and sTNF-αR1 levels were positively associated with bifurcation-IMT. Among HIV-uninfected men, higher levels of sTNF-αR2 were positively associated with CCA-IMT, fibrinogen with bifurcation-IMT and carotid plaque, and ICAM-1 with carotid plaque. CONCLUSION In addition to greater levels of systemic inflammation, heightened monocyte activation (sCD163, CCL2) may contribute to the burden of atherosclerosis among HIV-infected persons.
Collapse
Affiliation(s)
- Vinita Subramanya
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Heather S. McKay
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Rebeccah M. Brusca
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Frank J. Palella
- Division of Infectious Diseases, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Lawrence A. Kingsley
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Mallory D. Witt
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-University of California Los Angeles, Torrance, California, United States of America
| | - Howard N. Hodis
- Atherosclerosis Research Unit, Keck School of Medicine at University of Southern California, Los Angeles, California, United States of America
| | - Russell P. Tracy
- Departments of Pathology & Laboratory Medicine, and Biochemistry, University of Vermont, Larner College of Medicine, Colchester, Vermont, United States of America
| | - Wendy S. Post
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Sabina A. Haberlen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
159
|
Hijmans JG, Stockleman K, Reiakvam W, Levy MV, Brewster LM, Bammert TD, Greiner JJ, Connick E, DeSouza CA. Effects of HIV-1 gp120 and tat on endothelial cell sensescence and senescence-associated microRNAs. Physiol Rep 2019; 6:e13647. [PMID: 29595877 PMCID: PMC5875545 DOI: 10.14814/phy2.13647] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 02/01/2018] [Accepted: 02/12/2018] [Indexed: 12/26/2022] Open
Abstract
The aim of this study was to determine, in vitro, the effects of X4 and R5 HIV‐1 gp120 and Tat on: (1) endothelial cell senescence and (2) endothelial cell microRNA (miR) expression. Endothelial cells were treated with media without and with: R5 gp120 (100 ng/mL), X4 gp120 (100 ng/mL), or Tat (500 ng/mL) for 24 h and stained for senescence‐associated β‐galactosidase (SA‐β‐gal). Cell expression of miR‐34a, miR‐217, and miR‐146a was determined by RT‐PCR. X4 and R5 gp120 and Tat significantly increased (~100%) cellular senescence versus control. X4 gp120 significantly increased cell expression of miR‐34a (1.60 ± 0.04 fold) and miR‐217 (1.52 ± 0.18), but not miR‐146a (1.25 ± 0.32). R5 gp120 significantly increased miR‐34a (1.23 ± 0.07) and decreased miR‐146a (0.56 ± 0.07). Tat significantly increased miR‐34a (1.49 ± 0.16) and decreased miR‐146a (0.55 ± 0.23). R5 and Tat had no effect on miR‐217 (1.05 ± 0.13 and 1.06 ± 0.24; respectively). HIV‐1 gp120 (X4 and R5) and Tat promote endothelial cell senescence and dysregulation of senescence‐associated miRs.
Collapse
Affiliation(s)
- Jamie G Hijmans
- Department of Integrative Physiology, Integrative Vascular Biology Laboratory, University of Colorado, Boulder, Colorado
| | - Kelly Stockleman
- Department of Integrative Physiology, Integrative Vascular Biology Laboratory, University of Colorado, Boulder, Colorado
| | - Whitney Reiakvam
- Department of Integrative Physiology, Integrative Vascular Biology Laboratory, University of Colorado, Boulder, Colorado
| | - Ma'ayan V Levy
- Department of Integrative Physiology, Integrative Vascular Biology Laboratory, University of Colorado, Boulder, Colorado
| | - Lillian M Brewster
- Department of Integrative Physiology, Integrative Vascular Biology Laboratory, University of Colorado, Boulder, Colorado
| | - Tyler D Bammert
- Department of Integrative Physiology, Integrative Vascular Biology Laboratory, University of Colorado, Boulder, Colorado
| | - Jared J Greiner
- Department of Integrative Physiology, Integrative Vascular Biology Laboratory, University of Colorado, Boulder, Colorado
| | - Elizabeth Connick
- Department of Medicine, Division of Infectious Disease, University of Arizona, Tucson, Arizona
| | - Christopher A DeSouza
- Department of Integrative Physiology, Integrative Vascular Biology Laboratory, University of Colorado, Boulder, Colorado
| |
Collapse
|
160
|
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: 2.2] [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
|
161
|
Pastori D, Mezzaroma I, Pignatelli P, Violi F, Lip GYH. Atrial fibrillation and human immunodeficiency virus type-1 infection: a systematic review. Implications for anticoagulant and antiarrhythmic therapy. Br J Clin Pharmacol 2019; 85:508-515. [PMID: 30575989 DOI: 10.1111/bcp.13837] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/01/2018] [Accepted: 12/07/2018] [Indexed: 12/31/2022] Open
Abstract
The prevalence and incidence of atrial fibrillation/flutter (AF/AFL) in patients with human immunodeficiency virus type-1 (HIV-1) infection have been poorly investigated. We performed a systematic review using PubMed and Cochrane Database of Systematic Reviews, and screening of references, searching for clinical studies reporting on the association between HIV-1 infection and AF/AFL. We also summarized the main interactions of antiretroviral agents with antithrombotic and antiarrhythmic drugs. We found a prevalence of AF/AFL ranging from 2.0% to 5.13% in patients with HIV-1, with an incidence rate of 3.6/1000 person-years. Low CD4+ count (<200-250 cells ml-1 ) and high viral load were predictors of AF/AFL. Regarding drugs interactions, nucleoside reverse transcriptase inhibitors, integrase inhibitor and maraviroc have the lowest interactions with oral anticoagulants. Among anticoagulants, dabigatran presents the most favourable profile. Most of antiarrhythmic drugs interact with protease inhibitors, with beta blockers and diltiazem having fewer interactions. The few studies available suggest a non-negligible prevalence of AF/AFL in patients with HIV-1 infection. Awareness of potential interactions with anticoagulation and antiarrhythmic drugs is needed to offer optimal management in this population.
Collapse
Affiliation(s)
- Daniele Pastori
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Ivano Mezzaroma
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Pasquale Pignatelli
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Francesco Violi
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK.,Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
162
|
Abstract
Current antiretroviral therapy (ART) effectively suppresses Human Immunodeficiency Virus type 1 (HIV-1) in infected individuals. However, even long term ART does not eradicate HIV-1 infected cells and the virus persists in cellular reservoirs. Beside memory CD4+ T cells, cells of the myeloid lineage, especially macrophages, are believed to be an important sanctuary for HIV-1. Monocytes and macrophages are key players in the innate immune response to pathogens and are recruited to sites of infection and inflammation. Due to their long life span and ability to reside in virtually every tissue, macrophages have been proposed to play a critical role in the establishment and persistence of the HIV-1 reservoir. Current HIV-1 cure strategies mainly focus on the concept of "shock and kill" to purge the viral reservoir. This approach aims to reactivate viral protein production in latently infected cells, which subsequently are eliminated as a consequence of viral replication, or recognized and killed by the immune system. Macrophage susceptibility to HIV-1 infection is dependent on the local microenvironment, suggesting that molecular pathways directing differentiation and polarization are involved. Current latency reversing agents (LRA) are mainly designed to reactivate the HIV-1 provirus in CD4+ T cells, while their ability to abolish viral latency in macrophages is largely unknown. Moreover, the resistance of macrophages to HIV-1 mediated kill and the presence of infected macrophages in immune privileged regions including the central nervous system (CNS), may pose a barrier to elimination of infected cells by current "shock and kill" strategies. This review focusses on the role of monocytes/macrophages in HIV-1 persistence. We will discuss mechanisms of viral latency and persistence in monocytes/macrophages. Furthermore, the role of these cells in HIV-1 tissue distribution and pathogenesis will be discussed.
Collapse
Affiliation(s)
- Zita Kruize
- Laboratory for Viral Immune Pathogenesis, Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Neeltje A Kootstra
- Laboratory for Viral Immune Pathogenesis, Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
163
|
Mechanisms of Cardiovascular Disease in the Setting of HIV Infection. Can J Cardiol 2018; 35:238-248. [PMID: 30825947 DOI: 10.1016/j.cjca.2018.12.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/11/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
Although the initial reports of increased cardiovascular (CV) disease in the setting of advanced AIDS were reported approximately 30 years ago, advances in antiretroviral therapy and immediate initiation of therapy on diagnosis have transformed what was once a deadly infectious disease into a chronic health condition. Accordingly, the types of CV diseases occurring in HIV have shifted from pericardial effusions and dilated cardiomyopathy to atherosclerosis and heart failure. The underlying pathophysiology of HIV-associated CV disease remains poorly understood, partly because of the rapidly evolving nature of HIV treatment and because clinical endpoints take many years to develop. The gut plays an important role in the early pathogenesis of HIV infection as HIV preferentially infects CD4+ T cells, 80% of which are located in gut mucosa. The loss of these T cells damages gut mucosa resulting in increased gut permeability and microbial translocation, which incites chronic inflammation and immune activation. Antiretroviral therapy does not cure HIV infection and immune abnormalities persist. These abnormalities correlate with mortality and CV events. The effects of antiretroviral therapy on CV risk are complex; treatment reduces inflammation and other markers of CV risk but induces lipid abnormalities, most commonly hypertriglyceridemia. On a molecular level, monocytes/macrophages, platelet reactivity, and immune cell activation, which play a role in the general population, may be heightened in the setting of HIV and contribute to HIV-associated atherosclerosis. Chronic inflammation represents an inviting therapeutic target in HIV, as it does in uninfected persons with atherosclerosis.
Collapse
|
164
|
Sarfo FS, Nichols M, Agyei B, Singh A, Ennin E, Nyantakyi AD, Asibey SO, Tagge R, Gebregziabher M, Jenkins C, Ovbiagele B. Burden of subclinical carotid atherosclerosis and vascular risk factors among people living with HIV in Ghana. J Neurol Sci 2018; 397:103-111. [PMID: 30599299 DOI: 10.1016/j.jns.2018.12.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/12/2018] [Accepted: 12/18/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND The burden of cardiovascular disease (CVD) among people living with HIV (PLWH) in sub-Saharan Africa is projected to rise due to a rapid epidemiological transition and improved treatment of HIV infection on the sub-continent. OBJECTIVE The Evaluation of Vascular Event Risk while on Long-term Anti-retroviral Suppressive Therapy (EVERLAST) Study sought to assess the extent of subclinical atherosclerosis and characterize the nature of CVD risk factors among HIV patients on Antiretroviral therapy (ART) in Ghana. METHODS We conducted a cross-sectional study involving HIV patients on antiretroviral therapy (n = 250) in comparison with HIV positive ART naïve (n = 201), and HIV uninfected controls (n = 250). We assessed prevalence of hypertension, dyslipidemia, diabetes mellitus, central obesity, and carotid atherosclerosis using B-mode carotid Doppler ultrasonography. We assessed factors associated with subclinical atherosclerosis defined by a carotid intimal media thickness (CIMT) cut-off of ≥0.78 mm among PLWH using a logistic regression model. RESULTS Mean age of PLWH on combination ART (cART) was 45.7 ± 8.6 years, 42.9 ± 8.8 years among PLWH not on cART, and 44.9 ± 9.5 years among HIV negative controls of which 81.2%, 81.6% and 81.1% respectively were females. Prevalence of subclinical atherosclerosis at the common carotid artery in the three groups was 67.6%, 66.7% and 62.4%, p = 0.43. Among PLWH, raised serum total cholesterol (OR 1.16, 95% CI: 1.00-1.35) and triglycerides (OR 1.32, 95% CI: 1.01-1.73) were significantly associated with subclinical atherosclerosis. Prevalence of vascular risk factors among PLWH on cART, PLWH cART naïve, and HIV negative controls respectively were as follows: dyslipidemia- 79.5%, 83.1%, and 73.5%, p = 0.04; hypertension- 40.2%, 23.4%, and 44.9%, p < 0.0001; central obesity-61.8%, 66.7%, and 78.2%, p < 0.0001; diabetes mellitus-6.8%, 5.5% and 4.9%, p = 0.53. CONCLUSION Overall while there is a high baseline prevalence of CVD risk factors in the Ghanaian population, serum lipid derangements appear to be more prevalent among HIV infected patients, and are linked to sub-clinical atherosclerosis. Future studies need to confirm these findings, explore the underlying pathophysiology, and optimize treatment strategies to avert untoward CVD outcomes.
Collapse
Affiliation(s)
- Fred Stephen Sarfo
- Kwame Nkrumah University of Science & Technology, Kumasi, Ghana; Komfo Anokye Teaching Hospital, Kumasi, Ghana.
| | | | - Benedict Agyei
- Kwame Nkrumah University of Science & Technology, Kumasi, Ghana
| | - Arti Singh
- Kwame Nkrumah University of Science & Technology, Kumasi, Ghana
| | | | | | | | - Raelle Tagge
- Medical University of South Carolina, Charleston, USA
| | | | | | | |
Collapse
|
165
|
Ryan T, Affandi JS, Gahungu N, Dwivedi G. Noninvasive Cardiovascular Imaging: Emergence of a Powerful Tool for Early Identification of Cardiovascular Risk in People Living With HIV. Can J Cardiol 2018; 35:260-269. [PMID: 30825948 DOI: 10.1016/j.cjca.2018.11.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/06/2018] [Accepted: 11/20/2018] [Indexed: 12/31/2022] Open
Abstract
Antiretroviral therapy (ART) has been pivotal in prolonging the lifespan of people living with HIV (PLWH). However, this also simultaneously increases their risk of cardiovascular disease (CVD) either related to ART, aging, hypertension, immunosenescence, inflammation, immune activation, or other comorbidities. Although the use of risk markers has greatly enhanced the field of cardiovascular (CV) medicine and improved the prognosis and early diagnosis in the general population, this strategy has not been clearly elucidated in PLWH. Developing accurate risk algorithms for PLWH requires an innate understanding of mechanistic factors influencing their risks. Early identification of CV risk will significantly enhance the prospects of PLWH living longer and relatively healthily. Herein, we discuss the use of multimodality noninvasive CV imaging as robust markers for ameliorating CV risk. The ability to prognosticate CV risk and hence prevent CV events in PLWH would represent an important advance in CV medicine, allowing precise detection and early institution of preventative strategies. Using novel CV imaging modalities and strategies would have a positive impact on precision medicine in this patient cohort.
Collapse
Affiliation(s)
- Timothy Ryan
- Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Jacquita S Affandi
- School of Public Health, Curtin University, Bentley, Western Australia, Australia; Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
| | - Nestor Gahungu
- Royal Perth Hospital, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Fiona Stanley Hospital, Murdoch, Western Australia, Australia; Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia; The University of Western Australia, Crawley, Western Australia, Australia.
| |
Collapse
|
166
|
Mosepele M, Regan S, Massaro J, Meigs JB, Zanni MV, D'Agostino RB, Grinspoon SK, Triant VA. Impact of the American College of Cardiology/American Heart Association Cholesterol Guidelines on Statin Eligibility Among Human Immunodeficiency Virus-Infected Individuals. Open Forum Infect Dis 2018; 5:ofy326. [PMID: 30619912 PMCID: PMC6306565 DOI: 10.1093/ofid/ofy326] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 12/11/2018] [Indexed: 12/16/2022] Open
Abstract
Background Individuals with human immunodeficiency virus (HIV) face elevated cardiovascular disease (CVD) risk. There are limited data regarding the application of the American College of Cardiology/American Heart Association (ACC/AHA) cholesterol guidelines in HIV compared with non-HIV patients. Methods Human immunodeficiency virus-infected and demographically similar control patients were assessed for statin recommendation status by ACC/AHA and the National Cholesterol Education Program Adult Treatment Program III (ATPIII), indication for statin recommendation, actual statin prescription, and CVD event. Outcomes were atherosclerotic CVD for ACC/AHA and coronary heart disease for ATPIII. Results In a clinical care cohort of 1394 patients infected with HIV, 38.6% (538 of 1394) of patients were recommended for statin therapy by the ACC/AHA guidelines compared with 20.1% (280 of 1394) by the ATPIII guidelines. Of those recommended for statin therapy, actual statin prescription rates were 42.8% (230 of 538) for ACC/AHA and 66.4% (186 of 280) for ATPIII. Among patients infected with HIV with an incident CVD event during follow-up, statin therapy was recommended for 59.2% (42 of 71) of patients by ACC/AHA and 35.2% (25 of 71) by ATPIII, versus 71.6% (141 of 197) by ACC/AHA and 43.1% (85 of 197) by ATPIII in the control group. Conclusions In an HIV clinical care cohort, the ACC/AHA cholesterol guidelines recommend a higher proportion of patients for statin therapy and identify an increased proportion of patients with a CVD event compared with ATPIII. However, 40% of patients with a CVD event would not have been recommended for statin therapy by ACC/AHA, compared with 29% for controls. This gap in identification of patients infected with HIV at high CVD risk underscores the need for HIV-specific cardiovascular prevention strategies.
Collapse
Affiliation(s)
- Mosepele Mosepele
- Faculty of Medicine, University of Botswana.,Department of Immunology and Infectious Disease, Harvard School of Public Health, Boston, Massachusetts
| | - Susan Regan
- Division of General Internal Medicine, Massachusetts General Hospital, Boston
| | - Joseph Massaro
- Mathematics and Statistics Department, Boston University College of Arts and Sciences, Massachusetts
| | - James B Meigs
- Division of General Internal Medicine, Massachusetts General Hospital, Boston
| | - Markella V Zanni
- Program in Nutritional Metabolism, Massachusetts General Hospital, Boston
| | - Ralph B D'Agostino
- Mathematics and Statistics Department, Boston University College of Arts and Sciences, Massachusetts
| | - Steven K Grinspoon
- Program in Nutritional Metabolism, Massachusetts General Hospital, Boston
| | - Virginia A Triant
- Division of General Internal Medicine, Massachusetts General Hospital, Boston.,Division of Infectious Diseases, Massachusetts General Hospital, Boston
| |
Collapse
|
167
|
Yanagawa B, Verma S, Dwivedi G, Ruel M. Cardiac Surgery in HIV Patients: State of the Art. Can J Cardiol 2018; 35:320-325. [PMID: 30744921 DOI: 10.1016/j.cjca.2018.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 11/27/2022] Open
Abstract
The clinical status of HIV infection has changed dramatically with the introduction of combined antiretroviral therapy. Patients with HIV are now living long enough to be susceptible to chronic illnesses, such as coronary disease and nonischemic cardiomyopathy, which can be consequences of the combined antiretroviral therapy treatment itself. Cardiovascular diseases are a major source of morbidity and mortality in HIV-positive patients. Increasingly, such patients might be candidates for the full range of cardiac surgical interventions, including coronary bypass, valve surgery, and heart transplantation. There has been a shift from offering palliative procedures such as pericardial window and balloon valvuloplasty, to more conventional and durable surgical therapies in HIV-positive patients. We herein provide an overview of the contemporary outcomes of cardiac surgery in this complex and unique patient population. We review some of the ethical issues around the selection and surgical care of HIV-positive patients. We also discuss strategies to best protect the surgical treatment team from the risks of HIV transmission. Finally, we highlight the need for involvement of dedicated infectious disease professionals in a multidisciplinary heart team approach, aiming at the comprehensive care of these unique and complex patients.
Collapse
Affiliation(s)
- Bobby Yanagawa
- Division of Cardiac Surgery, St Michael's Hospital, Toronto, Ontario, Canada.
| | - Subodh Verma
- Division of Cardiac Surgery, St Michael's Hospital, Toronto, Ontario, Canada
| | - Girish Dwivedi
- Harry Perkins Institute of Medical Research, The University of Western Australia, Perth, Australia
| | - Marc Ruel
- Ottawa Heart Institute, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
168
|
Waters DD, Hsue PY. Lipid Abnormalities in Persons Living With HIV Infection. Can J Cardiol 2018; 35:249-259. [PMID: 30704819 DOI: 10.1016/j.cjca.2018.11.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/02/2018] [Accepted: 11/06/2018] [Indexed: 01/16/2023] Open
Abstract
Lipid abnormalities are prevalent among persons living with HIV infection and contribute to increasing the risk of cardiovascular events. Antiretroviral therapy (ART) is associated with lipid abnormalities, most commonly hypertriglyceridemia, but also increases in low-density lipoprotein cholesterol and total cholesterol. Different classes of ART, and different drugs within classes, have differing effects on lipid levels, but in general newer drugs have more favourable effects compared with older ones. Low-level inflammation and chronic immune activation act on lipids through a variety of mechanisms to make them more atherogenic. As a consequence, risk is higher than would be expected for any given cholesterol level. Clinical outcome trials of cholesterol-lowering therapies have not yet been completed in people living with HIV, so that treatment decisions depend on extrapolation from studies in uninfected populations. Traditional risk assessment tools underestimate cardiovascular risk in individuals with HIV. Statins are the mainstay of lipid-lowering drug treatment; however, drug-drug interactions with ART must be considered. Simvastatin and lovastatin are contraindicated in patients taking protease inhibitors, and the dose of atorvastatin and rosuvastatin should be limited to 40 mg and 10 mg/d with some ART combinations. Switching from older forms of ART to lipid-friendly newer ones is a useful strategy as long as virologic suppression is maintained, but adding a statin lowers low-density lipoprotein cholesterol more effectively. Studies indicate that lipid abnormalities are not treated as aggressively in individuals living with HIV as they are in uninfected people, making this an opportunity to improve care.
Collapse
Affiliation(s)
- David D Waters
- Division of Cardiology, Zuckerberg San Francisco General Hospital, and Department of Medicine, University of California, San Francisco, California, USA.
| | - Priscilla Y Hsue
- Division of Cardiology, Zuckerberg San Francisco General Hospital, and Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
169
|
Zanni MV, Toribio M, Robbins GK, Burdo TH, Lu MT, Ishai AE, Feldpausch MN, Martin A, Melbourne K, Triant VA, Suchindran S, Lee H, Hoffmann U, Williams KC, Tawakol A, Grinspoon SK. Effects of Antiretroviral Therapy on Immune Function and Arterial Inflammation in Treatment-Naive Patients With Human Immunodeficiency Virus Infection. JAMA Cardiol 2018; 1:474-80. [PMID: 27438325 DOI: 10.1001/jamacardio.2016.0846] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
IMPORTANCE Individuals with human immunodeficiency virus (HIV) infection receiving combined antiretroviral therapy (ART) have an increased risk of myocardial infarction. Effects of ART on arterial inflammation among treatment-naive individuals with HIV are unknown. OBJECTIVE To determine the effects of newly initiated ART on arterial inflammation and other immune/inflammatory indices in ART-naive patients with HIV infection. DESIGN, SETTING, PARTICIPANTS Twelve treatment-naive HIV-infected individuals underwent fludeoxyglucose F 18 ([18F]-FDG) positron emission tomographic scanning for assessment of arterial inflammation, coronary computed tomographic angiography for assessment of subclinical atherosclerosis, and systemic immune and metabolic phenotyping before and 6 months after the initiation of therapy with elvitegravir, cobicistat, emtricitabine, and tenofovir disoproxil fumarate (combined ART). Systemic immune and metabolic factors were also assessed in 12 prospectively recruited individuals without HIV serving as controls. The study began July 24, 2012, and was completed May 7, 2015. INTERVENTIONS Combined ART in the HIV-infected cohort. MAIN OUTCOMES AND MEASURES The primary outcome was change in aortic target-background ratio (TBR) on [18F]-FDG-PET with combined ART in the HIV-infected group. RESULTS For the 12 participants with HIV infection (mean (SD) age, 35 [11] years), combined ART suppressed viral load (mean [SD] log viral load, from 4.3 [0.6] to 1.3 [0] copies/mL; P < .001), increased the CD4+ T-cell count (median [IQR], from 461 [332-663] to 687 [533-882] cells/mm3; P < .001), and markedly reduced percentages of circulating activated CD4+ T cells (human leukocyte antigen-D related [HLA-DR]+CD38+CD4+) (from 3.7 [1.8-5.0] to 1.3 [0.3-2.0]; P = .008) and CD8+ T cells (HLA-DR+CD38+CD8+) (from 18.3 [8.1-27.0] to 4.0 [1.5-7.8]; P = .008), increased the percentage of circulating classical CD14+CD16- monocytes (from 85.8 [83.7-90.8] to 91.8 [87.5-93.2]; P = .04), and reduced levels of CXCL10 (mean [SD] log CXCL10, from 2.4 [0.4] to 2.2 [0.4] pg/mL; P = .03). With combined ART, uptake of [18F]-FDG in the axillary lymph nodes, as measured by TBR, decreased from a median (IQR) of 3.7 (1.3-7.0) at baseline to 1.4 (0.9-1.9; P = .01) at study end. In contrast, no significant decrease was seen in aortic TBR in response to combined ART (mean [SD], 1.9 [0.2]; median [IQR], 2.0 [1.8-2.1] at baseline to 2.2 [0.4]; 2.1 [1.9-2.6], respectively, at study end; P = .04 by 2-way test, P = .98 for test of decrease by 1-way test). Changes in aortic TBR during combined ART were significantly associated with changes in lipoprotein-associated phospholipase A2 (n = 10; r = 0.67; P = .03). Coronary plaque increased among 3 participants with HIV infection with baseline plaque and developed de novo in 1 participant during combined ART. CONCLUSIONS AND RELEVANCE Newly initiated combined ART in treatment-naive individuals with HIV infection had discordant effects to restore immune function without reducing arterial inflammation. Complementary strategies to reduce arterial inflammation among ART-treated HIV-infected individuals may be needed.
Collapse
Affiliation(s)
- Markella V Zanni
- Program in Nutritional Metabolism, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Mabel Toribio
- Program in Nutritional Metabolism, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Gregory K Robbins
- Infectious Disease Division, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Tricia H Burdo
- Department of Biology, Boston College, Chestnut Hill, Massachusetts
| | - Michael T Lu
- Cardiac Magnetic Resonance-Positron Emission Tomography-Computed Tomography Program and Division of Cardiology, Department of Radiology, Boston, Massachusetts
| | - Amorina E Ishai
- Cardiac Magnetic Resonance-Positron Emission Tomography-Computed Tomography Program and Division of Cardiology, Department of Radiology, Boston, Massachusetts
| | - Meghan N Feldpausch
- Program in Nutritional Metabolism, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Amanda Martin
- Program in Nutritional Metabolism, Massachusetts General Hospital and Harvard Medical School, Boston
| | | | - Virginia A Triant
- Infectious Disease Division, Massachusetts General Hospital and Harvard Medical School, Boston6General Internal Medicine Division, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Sujit Suchindran
- Infectious Disease Division, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Udo Hoffmann
- Cardiac Magnetic Resonance-Positron Emission Tomography-Computed Tomography Program and Division of Cardiology, Department of Radiology, Boston, Massachusetts
| | | | - Ahmed Tawakol
- Cardiac Magnetic Resonance-Positron Emission Tomography-Computed Tomography Program and Division of Cardiology, Department of Radiology, Boston, Massachusetts
| | - Steven K Grinspoon
- Program in Nutritional Metabolism, Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
170
|
Hoogeveen RM, Nahrendorf M, Riksen NP, Netea MG, de Winther MPJ, Lutgens E, Nordestgaard BG, Neidhart M, Stroes ESG, Catapano AL, Bekkering S. Monocyte and haematopoietic progenitor reprogramming as common mechanism underlying chronic inflammatory and cardiovascular diseases. Eur Heart J 2018; 39:3521-3527. [PMID: 29069365 PMCID: PMC6174026 DOI: 10.1093/eurheartj/ehx581] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/11/2017] [Accepted: 10/12/2017] [Indexed: 12/19/2022] Open
Abstract
A large number of cardiovascular events are not prevented by current therapeutic regimens. In search for additional, innovative strategies, immune cells have been recognized as key players contributing to atherosclerotic plaque progression and destabilization. Particularly the role of innate immune cells is of major interest, following the recent paradigm shift that innate immunity, long considered to be incapable of learning, does exhibit immunological memory mediated via epigenetic reprogramming. Compelling evidence shows that atherosclerotic risk factors promote immune cell migration by pre-activation of circulating innate immune cells. Innate immune cell activation via metabolic and epigenetic reprogramming perpetuates a systemic low-grade inflammatory state in cardiovascular disease (CVD) that is also common in other chronic inflammatory disorders. This opens a new therapeutic area in which metabolic or epigenetic modulation of innate immune cells may result in decreased systemic chronic inflammation, alleviating CVD, and its co-morbidities.
Collapse
Affiliation(s)
- Renate M Hoogeveen
- Department of Vascular Medicine, Academic Medical Centre, Meibergdreef 9, Amsterdam, The Netherlands
| | - Matthias Nahrendorf
- Center for Systems Biology and Department of Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, 55 Fruit Street Boston, MA, USA
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 8, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 8, Nijmegen, The Netherlands
| | - Menno P J de Winther
- Department of Medical Biochemistry, Academic Medical Centre, Meibergdreef 9, Amsterdam, The Netherlands
| | - Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Pettenkoferstraße 9, Munich, Germany
| | - Børge G Nordestgaard
- The Copenhagen General Population Study and Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Ringvej 75, Herlev, Copenhagen, Denmark
| | - Michel Neidhart
- Center of Experimental Rheumatology, University Hospital Zurich, Schlieren, Switzerland
| | - Erik S G Stroes
- Department of Vascular Medicine, Academic Medical Centre, Meibergdreef 9, Amsterdam, The Netherlands
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan and IRCCS Multimedica, Via Balzaretti, Milano, Italy
| | - Siroon Bekkering
- Department of Vascular Medicine, Academic Medical Centre, Meibergdreef 9, Amsterdam, The Netherlands
- Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 8, Nijmegen, The Netherlands
| |
Collapse
|
171
|
Ahmed D, Roy D, Cassol E. Examining Relationships between Metabolism and Persistent Inflammation in HIV Patients on Antiretroviral Therapy. Mediators Inflamm 2018; 2018:6238978. [PMID: 30363715 PMCID: PMC6181007 DOI: 10.1155/2018/6238978] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 09/06/2018] [Indexed: 12/30/2022] Open
Abstract
With the advent of antiretroviral therapy (ART), HIV-infected individuals are now living longer and healthier lives. However, ART does not completely restore health and treated individuals are experiencing increased rates of noncommunicable diseases such as dyslipidemia, insulin resistance, type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease. While it is well known that persistent immune activation and inflammation contribute to the development of these comorbid diseases, the mechanisms underlying this chronic activation remain incompletely understood. In this review, we will discuss emerging evidence that suggests that alterations in cellular metabolism may play a central role in driving this immune dysfunction in HIV patients on ART.
Collapse
Affiliation(s)
- Duale Ahmed
- Department of Biology, Carleton University, Ottawa, Ontario, Canada
| | - David Roy
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
172
|
Shah ASV, Stelzle D, Lee KK, Beck EJ, Alam S, Clifford S, Longenecker CT, Strachan F, Bagchi S, Whiteley W, Rajagopalan S, Kottilil S, Nair H, Newby DE, McAllister DA, Mills NL. Global Burden of Atherosclerotic Cardiovascular Disease in People Living With HIV: Systematic Review and Meta-Analysis. Circulation 2018; 138:1100-1112. [PMID: 29967196 PMCID: PMC6221183 DOI: 10.1161/circulationaha.117.033369] [Citation(s) in RCA: 627] [Impact Index Per Article: 89.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 06/05/2018] [Indexed: 01/23/2023]
Abstract
BACKGROUND With advances in antiretroviral therapy, most deaths in people with HIV are now attributable to noncommunicable illnesses, especially cardiovascular disease. We determine the association between HIV and cardiovascular disease, and estimate the national, regional, and global burden of cardiovascular disease attributable to HIV. METHODS We conducted a systematic review across 5 databases from inception to August 2016 for longitudinal studies of cardiovascular disease in HIV infection. A random-effects meta-analysis across 80 studies was used to derive the pooled rate and risk of cardiovascular disease in people living with HIV. We then estimated the temporal changes in the population-attributable fraction and disability-adjusted life-years (DALYs) from HIV-associated cardiovascular disease from 1990 to 2015 at a regional and global level. National cardiovascular DALYs associated with HIV for 2015 were derived for 154 of the 193 United Nations member states. The main outcome measure was the pooled estimate of the rate and risk of cardiovascular disease in people living with HIV and the national, regional, and global estimates of DALYs from cardiovascular disease associated with HIV. RESULTS In 793 635 people living with HIV and a total follow-up of 3.5 million person-years, the crude rate of cardiovascular disease was 61.8 (95% CI, 45.8-83.4) per 10 000 person-years. In comparison with individuals without HIV, the risk ratio for cardiovascular disease was 2.16 (95% CI, 1.68-2.77). Over the past 26 years, the global population-attributable fraction from cardiovascular disease attributable to HIV increased from 0.36% (95% CI, 0.21%-0.56%) to 0.92% (95% CI, 0.55%-1.41%), and DALYs increased from 0.74 (95% CI, 0.44-1.16) to 2.57 (95% CI, 1.53-3.92) million. There was marked regional variation with most DALYs lost in sub-Saharan Africa (0.87 million, 95% CI, 0.43-1.70) and the Asia Pacific (0.39 million, 95% CI, 0.23-0.62) regions. The highest population-attributable fraction and burden were observed in Swaziland, Botswana, and Lesotho. CONCLUSIONS People living with HIV are twice as likely to develop cardiovascular disease. The global burden of HIV-associated cardiovascular disease has tripled over the past 2 decades and is now responsible for 2.6 million DALYs per annum with the greatest impact in sub-Saharan Africa and the Asia Pacific regions. CLINICAL TRIAL REGISTRATION URL: https://www.crd.york.ac.uk/prospero . Unique identifier: CRD42016048257.
Collapse
Affiliation(s)
- Anoop S V Shah
- BHF Centre for Cardiovascular Science, University of Edinburgh, Scotland (A.S.V.S., K.K.L., S.A., S.C., F.S., D.E.N., N.L.M.)
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Scotland (A.S.V.S., H.N.)
| | - Dominik Stelzle
- Joint United Nations Programme on HIV/AIDS, Geneva, Switzerland (D.S., E.J.B.)
- Center for Global Health, Department of Neurology, Technical University, Munich, Germany (D.S.)
| | - Kuan Ken Lee
- BHF Centre for Cardiovascular Science, University of Edinburgh, Scotland (A.S.V.S., K.K.L., S.A., S.C., F.S., D.E.N., N.L.M.)
| | - Eduard J Beck
- Joint United Nations Programme on HIV/AIDS, Geneva, Switzerland (D.S., E.J.B.)
| | - Shirjel Alam
- BHF Centre for Cardiovascular Science, University of Edinburgh, Scotland (A.S.V.S., K.K.L., S.A., S.C., F.S., D.E.N., N.L.M.)
| | - Sarah Clifford
- BHF Centre for Cardiovascular Science, University of Edinburgh, Scotland (A.S.V.S., K.K.L., S.A., S.C., F.S., D.E.N., N.L.M.)
| | - Chris T Longenecker
- Division of Cardiovascular Medicine, Case Western Reserve School of Medicine, Cleveland, OH (C.T.L., S.R.)
| | - Fiona Strachan
- BHF Centre for Cardiovascular Science, University of Edinburgh, Scotland (A.S.V.S., K.K.L., S.A., S.C., F.S., D.E.N., N.L.M.)
| | - Shashwatee Bagchi
- Division of Infectious Diseases and Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD (S.B., S.K.)
| | - William Whiteley
- Centre for Clinical Brain Sciences (W.W.), University of Edinburgh, United Kingdom
| | - Sanjay Rajagopalan
- Division of Cardiovascular Medicine, Case Western Reserve School of Medicine, Cleveland, OH (C.T.L., S.R.)
| | - Shyamasundaran Kottilil
- Division of Infectious Diseases and Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD (S.B., S.K.)
| | - Harish Nair
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Scotland (A.S.V.S., H.N.)
| | - David E Newby
- BHF Centre for Cardiovascular Science, University of Edinburgh, Scotland (A.S.V.S., K.K.L., S.A., S.C., F.S., D.E.N., N.L.M.)
| | - David A McAllister
- Institute of Health and Wellbeing, University of Glasgow, United Kingdom (D.A.M.)
| | - Nicholas L Mills
- BHF Centre for Cardiovascular Science, University of Edinburgh, Scotland (A.S.V.S., K.K.L., S.A., S.C., F.S., D.E.N., N.L.M.)
| |
Collapse
|
173
|
Zidovudine-Based Treatments Inhibit the Glycosylation of ADAM17 and Reduce CD163 Shedding From Monocytes. J Acquir Immune Defic Syndr 2018; 79:126-134. [PMID: 29794822 DOI: 10.1097/qai.0000000000001769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
BACKGROUND sCD163, a biomarker of monocyte-macrophage activation, has been identified as a predictor of all-cause mortality in treated HIV-infected individuals. Nevertheless, little is known about whether different antiretroviral drugs differentially regulate sCD163 levels and monocyte activation. METHODS A total of 123 patients receiving zidovudine (ZDV)-based (n = 55) or tenofovir disoproxil fumarate (TDF)-based (n = 68) antiretroviral regimens were enrolled, and their viral loads, CD4 counts, as well as plasma sCD163 and sCD14 levels were quantified. Twenty-eight (14 in each group) patients donated additional blood samples for flow cytometry and gene expression analyses using purified monocytes. THP-1 cultures were also used to investigate the effect of ZDV on ADAM17, which is responsible for CD163 shedding. RESULTS As compared to the TDF-treated group, the ZDV-treated group had lower plasma sCD163 levels and higher CD163 expression on CD14++CD16 monocytes. Five metabolic-inflammatory genes exhibited significantly different expression levels between purified monocytes of the ZDV and TDF groups (IL-6, 2.90-fold lower in ZDV group, P < 0.001; iNOS, 1.81-fold higher; CX3CR1, 1.72-fold lower; MIP-1β, 1.10-fold lower; and PPARγ-1, 1.36-fold higher, P < 0.05). Moreover, we show that ZDV treatment increases the surface expression of CD163 in cultured THP-1 cells, accompanied by the inhibition of glycosylation and surface expression of ADAM17. CONCLUSIONS Compared with TDF treatment, ZDV treatment causes lower plasma sCD163 levels, probably by inhibiting the glycosylation of ADAM17 and CD163 shedding. Our results show that ZDV functions as an ADAM17 inhibitor in vivo and extend our understanding of its immune-modulatory effects and adverse effects.
Collapse
|
174
|
Toribio M, Fitch KV, Stone L, Zanni MV, Lo J, de Filippi C, Sponseller CA, Lee H, Grundberg I, Thompson MA, Aberg JA, Grinspoon SK. Assessing statin effects on cardiovascular pathways in HIV using a novel proteomics approach: Analysis of data from INTREPID, a randomized controlled trial. EBioMedicine 2018; 35:58-66. [PMID: 30174281 PMCID: PMC6156703 DOI: 10.1016/j.ebiom.2018.08.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/16/2018] [Accepted: 08/16/2018] [Indexed: 12/11/2022] Open
Abstract
Background People with HIV (PWH) demonstrate increased cardiovascular disease (CVD), due in part to increased immune activation, inflammation, and endothelial dysfunction. Methods In a randomized trial (INTREPID), 252 HIV-infected participants with dyslipidemia and no history of coronary artery disease were randomized (1:1) to pitavastatin 4 mg vs. pravastatin 40 mg for 52 weeks. Using a proteomic discovery approach, 92 proteins biomarkers were assessed using Proximity Extension Assay technology to determine the effects of statins on key atherosclerosis and CVD pathways among PWH. 225 participants had specimens available for biomarker analysis pre- and post-baseline. Findings The mean age was 49.5 ± 8.0 (mean ± SD), LDL-C 155 ± 25 mg/dl and CD4 count 620 ± 243 cell/mm3. Among all participants, three proteins significantly decreased: tissue factor pathway inhibitor [TFPI; t-statistic = −6.38, FDR p-value<0.0001], paraoxonase 3 [PON3; t-statistic = −4.64, FDR p-value = 0.0003], and LDL-receptor [LDLR; t-statistic = −4.45, FDR p-value = 0.0004]; and two proteins significantly increased galectin-4 [Gal-4; t-statistic = 3.50, FDR p-value = 0.01] and insulin-like growth factor binding protein 2 [IGFBP-2; t-statistic = 3.21, FDR p-value = 0.03]. The change in TFPI was significantly different between the pitavastatin and pravastatin groups. Among all participants, change in TFPI related to the change in LDL-C (r = 0.43, P < 0.0001) and change in Lp-PLA2 (r = 0.29, P < 0.0001). Interpretation Using a proteomics approach, we demonstrated that statins led to a significant reduction in the levels of TFPI, PON3, and LDLR and an increase in Gal-4 and IGFBP-2, key proteins involved in coagulation, redox signaling, oxidative stress, and glucose metabolism. Pitavastatin led to a greater reduction in TFPI than pravastatin. These data highlight potential novel mechanisms of statin effects among PWH. Fund This work was supported by an investigator-initiated grant to S.K.G. from KOWA Pharmaceuticals America, Inc. and the National Institutes of Health [P30 DK040561; Nutrition Obesity Research Center at Harvard]. M.T. was support by National Institutes of Health [5KL2TR001100-05; Harvard Catalyst KL2 grant]. Among PWH, statins significantly decreased three proteins [tissue factor pathway inhibitor (TFPI), paraoxonase 3 (PON3), and LDL-receptor (LDLR)]. Among PWH, statins significantly increased galectin-4 (Gal-4) and insulin-like growth factor binding protein 2 (IGFBP-2). The proteins significantly affected by statin therapy are involved in important pathways in atherosclerosis and cardiovascular disease. The change in TFPI was directly related to the change in LDL-C and a systemic marker of arterial inflammation (Lp-PLA2).
Collapse
Affiliation(s)
- Mabel Toribio
- Massachusetts General Hospital, Program in Nutritional Metabolism and Harvard Medical School (MT, KVF, LS, MVZ, JL, SKG), Boston, MA, USA
| | - Kathleen V Fitch
- Massachusetts General Hospital, Program in Nutritional Metabolism and Harvard Medical School (MT, KVF, LS, MVZ, JL, SKG), Boston, MA, USA
| | - Lauren Stone
- Massachusetts General Hospital, Program in Nutritional Metabolism and Harvard Medical School (MT, KVF, LS, MVZ, JL, SKG), Boston, MA, USA
| | - Markella V Zanni
- Massachusetts General Hospital, Program in Nutritional Metabolism and Harvard Medical School (MT, KVF, LS, MVZ, JL, SKG), Boston, MA, USA
| | - Janet Lo
- Massachusetts General Hospital, Program in Nutritional Metabolism and Harvard Medical School (MT, KVF, LS, MVZ, JL, SKG), Boston, MA, USA
| | | | | | - Hang Lee
- Massachusetts General Hospital, Biostatistics Center, and Harvard Medical School (HL), Boston, MA, USA
| | | | | | - Judith A Aberg
- Mount Sinai Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai (JAA), New York, NY, USA
| | - Steven K Grinspoon
- Massachusetts General Hospital, Program in Nutritional Metabolism and Harvard Medical School (MT, KVF, LS, MVZ, JL, SKG), Boston, MA, USA.
| |
Collapse
|
175
|
D'Antoni ML, Mitchell BI, McCurdy S, Byron MM, Ogata-Arakaki D, Chow D, Mehta NN, Boisvert WA, Lefebvre E, Shikuma CM, Ndhlovu LC, Baumer Y. Cenicriviroc inhibits trans-endothelial passage of monocytes and is associated with impaired E-selectin expression. J Leukoc Biol 2018; 104:1241-1252. [PMID: 30088682 DOI: 10.1002/jlb.5a0817-328rrr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 01/23/2023] Open
Abstract
Incidences of cardiovascular diseases (CVD) are high among virologically suppressed HIV-infected individuals. Monocyte activation and trafficking are key mechanisms in the evolution of CVD. We studied the ability of cenicriviroc (CVC), a dual C-C chemokine receptor type 2 (CCR2) and CCR5 antagonist, to influence the migration of monocytes from HIV-infected individuals on antiretroviral therapy (ART). Monocytes were derived from 23 ART-suppressed HIV-infected and 16 HIV-uninfected donors. In a trans-endothelial migration model, monocytes, and human aortic endothelial cells (HAoECs) were exposed to cenicriviroc and migrated monocytes, quantified. Expression of CCR2 and CCR5 on monocytes and adhesion molecules (E-selectin, ICAM-1, VCAM-1, PECAM-1, and CD99) on HAoECs were measured. The single antagonists, BMS-22 (CCR2), and maraviroc (CCR5), served as controls. When both HAoECs and monocytes together were exposed to the antagonists, cenicriviroc led to a greater decrease in monocyte migration compared to BMS-22 or vehicle in both HIV-infected and HIV-uninfected groups (P < 0.05), with maraviroc having no inhibitory effect. Cenicriviroc treatment of HAoECs alone decreased monocyte migration in the HIV-infected group when compared to vehicle (P < 0.01). Inhibition of migration was not evident when monocytes alone were exposed to cenicriviroc, BMS-22 or maraviroc. Incubation of HAoECs with cenicriviroc decreased E-selectin expression (P = 0.045) but had limited effects on the other adhesion molecules. Cenicriviroc inhibits monocyte trans-endothelial migration more effectively than single chemokine receptor blockade, which may be mediated via disruption of monocyte-endothelial tethering through reduced E-selectin expression. Cenicriviroc should be considered as a therapeutic intervention to reduce detrimental monocyte trafficking.
Collapse
Affiliation(s)
- Michelle L D'Antoni
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA.,Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Brooks I Mitchell
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA.,Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Sara McCurdy
- Department of Medicine, Center for Cardiovascular Research, University of Hawaii, Hawaii, USA
| | - Mary Margaret Byron
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA.,Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | | | - Dominic Chow
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - William A Boisvert
- Department of Medicine, Center for Cardiovascular Research, University of Hawaii, Hawaii, USA
| | | | | | - Lishomwa C Ndhlovu
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA.,Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Yvonne Baumer
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA.,Department of Medicine, Center for Cardiovascular Research, University of Hawaii, Hawaii, USA.,Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
176
|
Affiliation(s)
- Sasha A Fahme
- From the Center for Global Health, Weill Cornell Medical College, New York, NY (S.A.F., R.P.)
- Department of Internal Medicine, Weill Bugando School of Medicine, Mwanza, Tanzania (S.A.F., R.P.)
| | - Gerald S Bloomfield
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC (G.S.B.)
| | - Robert Peck
- From the Center for Global Health, Weill Cornell Medical College, New York, NY (S.A.F., R.P.)
- Department of Internal Medicine, Weill Bugando School of Medicine, Mwanza, Tanzania (S.A.F., R.P.)
- Mwanza Interventions Trial Unit, Tanzania (R.P.)
| |
Collapse
|
177
|
Feinstein MJ, Poole B, Engel Gonzalez P, Pawlowski AE, Schneider D, Provias TS, Palella FJ, Achenbach CJ, Lloyd-Jones DM. Differences by HIV serostatus in coronary artery disease severity and likelihood of percutaneous coronary intervention following stress testing. J Nucl Cardiol 2018; 25:872-883. [PMID: 27739037 PMCID: PMC5391305 DOI: 10.1007/s12350-016-0689-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/15/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND HIV-infected persons develop coronary artery disease (CAD) more commonly and earlier than uninfected persons; however, the role of non-invasive testing to stratify CAD risk in HIV is not well defined. METHODS AND RESULTS Patients were selected from a single-center electronic cohort of HIV-infected patients and uninfected controls matched 1:2 on age, sex, race, and type of cardiovascular testing performed. Patients with abnormal echocardiographic or nuclear stress testing who subsequently underwent coronary angiography were included. Logistic regressions were used to assess differences by HIV serostatus in two co-primary endpoints: (1) severe CAD (≥70% stenosis of at least one coronary artery) and (2) performance of percutaneous coronary intervention (PCI). HIV-infected patients (N = 189) were significantly more likely to undergo PCI following abnormal stress test when compared with uninfected persons (N = 319) after adjustment for demographics, CAD risk factors, previous coronary intervention, and stress test type (OR 1.85, 95% CI 1.12-3.04, P = 0.003). No associations between HIV serostatus and CAD were statistically significant, although there was a non-significant trend toward greater CAD for HIV-infected patients. CONCLUSIONS HIV-infected patients with abnormal cardiovascular stress testing who underwent subsequent coronary angiography did not have a significantly greater CAD burden than uninfected controls, but were significantly more likely to receive PCI.
Collapse
Affiliation(s)
- Matthew J Feinstein
- Division of Cardiovascular Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA.
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N. Lakeshore Drive, Suite 1400, Chicago, IL, 60611, USA.
| | - Brian Poole
- Department of Medicine, Beth Israel Deaconness Medical Center, Boston, MA, USA
| | - Pedro Engel Gonzalez
- Department of Medicine, Northwestern University Feinberg School of Medicine, 251 E. Huron, Suite 3-150, Chicago, IL, 60611, USA
| | - Anna E Pawlowski
- Northwestern Medicine Enterprise Data Warehouse, Chicago, IL, 60611, USA
| | - Daniel Schneider
- Northwestern Medicine Enterprise Data Warehouse, Chicago, IL, 60611, USA
| | - Tim S Provias
- Division of Cardiovascular Diseases, Northwestern University Feinberg School of Medicine, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Frank J Palella
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, 251 E. Huron St., Suite 3-150, Chicago, IL, 60611, USA
| | - Chad J Achenbach
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, 251 E. Huron St., Suite 3-150, Chicago, IL, 60611, USA
| | - Donald M Lloyd-Jones
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N. Lakeshore Drive, Suite 1400, Chicago, IL, 60611, USA
| |
Collapse
|
178
|
Cardiovascular disease risk among women living with HIV in North America and Europe. Curr Opin HIV AIDS 2018; 12:585-593. [PMID: 28832367 DOI: 10.1097/coh.0000000000000413] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW To examine the epidemiology and mechanistic underpinnings of heightened cardiovascular disease (CVD) risk among women living with HIV (WLHIV) in North America and Europe. RECENT FINDINGS WLHIV in North America and Europe exhibit high CVD incidence rates, which are at par with those of compatriot men living with HIV. Compared with uninfected women, WLHIV in these regions face a 2-4-fold increased relative risk for myocardial infarction, stroke, and heart failure. HIV-associated CVD risk is fuelled by a negative synergy of traditional cardiometabolic risk factors and heightened systemic immune activation/inflammation. Among WLHIV, female sex and endogenous sex hormone production influence both traditional cardiometabolic risk factors and patterns of systemic immune activation/inflammation. WLHIV in North America and Europe may also experience heightened CVD risk in relation to a relatively increased prevalence of behavioral and psychosocial CVD risk factors, coupled with suboptimal therapeutic targeting of known traditional cardiometabolic risk factors. SUMMARY Additional research on sex-specific mechanisms of HIV-associated CVD - based not only out of North America and Europe but also and especially out of Africa, Asia, and South America - will inform the development of CVD prediction algorithms and prevention guidelines clinically relevant to the approximately 17 million women aging with HIV globally.
Collapse
|
179
|
Mosepele M, Mohammed T, Mupfumi L, Moyo S, Bennett K, Lockman S, Hemphill LC, Triant VA. HIV disease is associated with increased biomarkers of endothelial dysfunction despite viral suppression on long-term antiretroviral therapy in Botswana. Cardiovasc J Afr 2018; 29:155-161. [PMID: 29771268 PMCID: PMC6107727 DOI: 10.5830/cvja-2018-003] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 01/14/2018] [Indexed: 12/11/2022] Open
Abstract
Background Untreated HIV infection is associated with increased biomarkers of endothelial dysfunction. However, the predictors and degree of endothelial dysfunction among virally suppressed HIV–infected adults on long–term antiretroviral therapy (ART) have not been well studied in sub– Saharan Africa (SSA). Methods We enrolled 112 HIV–infected adults with virological suppression on long–term ART and 84 HIV–uninfected controls in Botswana. We measured plasma levels of markers of endothelial injury [soluble vascular adhesion molecule 1 (VCAM–1), intercellular adhesion molecule 1 (ICAM–1) and E–selectin] and plasma levels of biomarkers of inflammation [interleukin 6 (IL–6)] and monocyte activation (sCD163). Baseline traditional cardiovascular disease (CVD) risk factors and bilateral common carotid intima–media thickness (cIMT) were also available for all participants. We assessed whether HIV status (despite virological suppression on ART) was associated with biomarkers of endothelial dysfunction after controlling for traditional CVD risk factors in linear regression models. We additionally assessed the association between IL–6, sCD163 and cIMT with endothelial dysfunction in separate multivariate linear regression models, controlling for cIMT, among virally suppressed HIV–infected participants only. Results In multivariate analysis, HIV infection was significantly associated with increased VCAM–1 (p < 0.01) and ICAM–1 (p = 0.03) but not E–selectin (p = 0.74) levels. Within the HIV–positive group, higher sCD163 levels were associated with decreased ICAM–1 and E–selectin (p < 0.01 and p = 0.01, respectively) but not VCAM–1 (p = 0.13) levels. IL–6 was not associated with any of the biomarkers of endothelial dysfunction. Conclusion HIV disease was associated with biomarkers of endothelial dysfunction among virally suppressed adults in Botswana on long–term ART after controlling for traditional CVD risk factors. Future work should explore the clinical impact of persistent endothelial dysfunction following longterm HIV viral suppression on the risk of CVD clinical endpoints among HIV–infected patients in this setting.
Collapse
Affiliation(s)
- Mosepele Mosepele
- Department of Medicine, Faculty of Medicine, University of Botswana, Gaborone, Botswana; Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.
| | | | - Lucy Mupfumi
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Sikhulile Moyo
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Kara Bennett
- Bennett Statistical Consulting Inc, Ballston Lake, New York, USA
| | - Shahin Lockman
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana; Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA; Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Linda C Hemphill
- Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Virginia A Triant
- Division of General Internal Medicine and Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
180
|
deFilippi C, Lo J, Christenson R, Grundberg I, Stone L, Zanni MV, Lee H, Grinspoon SK. Novel mediators of statin effects on plaque in HIV: a proteomics approach. AIDS 2018; 32:867-876. [PMID: 29369166 PMCID: PMC5869115 DOI: 10.1097/qad.0000000000001762] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE HIV patients have increased atherosclerotic coronary vascular disease (ASCVD), thought to be mediated through inflammatory mechanisms. We hypothesized that among asymptomatic HIV-infected patients with subclinical coronary plaque, statin therapy would modulate unique inflammatory and cardiovascular proteins in relation to change in subclinical coronary plaque volume. We tested this hypothesis using a novel proteomics approach. DESIGN Forty HIV-infected participants were randomized to atorvastatin (40 mg/day) versus placebo, and underwent computed tomography coronary angiography to quantify plaque volume at baseline and 1 year. METHODS We used Olink Cardiovascular III and Cardiometabolic panels based on dual antibody epitope recognition with linked DNA amplification to compare change over time in 184 proteins in treatment versus placebo and in relation to change in coronary plaque volume. RESULTS Six proteins (TFPI, CCL24, NT-Pro BNP, MBL2, LTBR, PCOLCE) changed significantly in the atorvastatin versus placebo group, many in innate immune and other novel inflammatory pathways. Twenty-six proteins changed significantly in relationship to total coronary plaque volume over 1 year. Notably, many of these proteins changed only weakly in relationship to change in low-density lipoprotein (LDL). Overlapping these two broad discovery approaches, proteins involved in myocardial fibrosis/collagen formation and monocyte chemoattraction changed with statin treatment, in relationship to plaque volume, but not LDL. CONCLUSION This proof-of-concept study employing a proteomic discovery platform offers insight into statin effects on novel immune pathways relevant to ASCVD progression in HIV. Novel biomarker discovery may enhance precision medicine strategies to estimate the efficacy of targeted therapies to reduce ASCVD progression and events in HIV.
Collapse
Affiliation(s)
| | - Janet Lo
- Program in Nutritional Metabolism, MGH and Harvard Medical School, Boston
| | - Robert Christenson
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Lauren Stone
- Program in Nutritional Metabolism, MGH and Harvard Medical School, Boston
| | - Markella V Zanni
- Program in Nutritional Metabolism, MGH and Harvard Medical School, Boston
| | - Hang Lee
- MGH Biostatistics Center, MGH and Harvard Medical School, Boston, Massachusetts
| | - Steven K Grinspoon
- Program in Nutritional Metabolism, MGH and Harvard Medical School, Boston
| |
Collapse
|
181
|
Kearns AC, Robinson JA, Shekarabi M, Liu F, Qin X, Burdo TH. Caspase-1-associated immune activation in an accelerated SIV-infected rhesus macaque model. J Neurovirol 2018; 24:420-431. [PMID: 29611111 DOI: 10.1007/s13365-018-0630-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/03/2018] [Accepted: 03/06/2018] [Indexed: 02/07/2023]
Abstract
In the antiretroviral therapy (ART) era, chronic HIV infection is primarily associated with chronic inflammation driving comorbidities such as cardiovascular disease and neurocognitive impairment. Caspase-1 activation in leukocytes has been documented in HIV infection; however, whether caspase-1 activation and the downstream pro-inflammatory cytokines interleukin-1beta (IL-1β) and interleukin-18 (IL-18) contribute to chronic inflammation in HIV comorbidities remains undetermined. The relationship between the caspase-1 cascade and persistent inflammation in HIV has not been investigated. Here, we used an accelerated simian immunodeficiency virus (SIV)-infected rhesus macaque model with or without ART to investigate the dynamics of caspase-1 and immune cell activation before infection, 21 days post infection (dpi), and necropsy. Caspase-1, IL-18, IL-1β, and immune markers were measured both in the circulation and lymphoid tissues. We found a significant increase in caspase-1 and IL-18 in SIV infection that positively correlated with inflammatory monocytes and negatively correlated with CD4+ T cell counts. ART attenuated these effects at necropsy in the circulation. Further, lymph nodes from SIV+ or SIV+ART animals had increased activation of caspase-1 and potential upstream priming of the NF-κB pathway, indicating that tissue-specific immune activation persists with ART. Together, these results shed light on the interconnectedness of the caspase-1 pathway and peripheral immune activation and further indicate that ART is not sufficient for suppressing inflammation. The caspase-1 pathway may provide novel therapeutic targets to improve HIV-associated comorbidities and health outcomes in the context of viral suppression.
Collapse
Affiliation(s)
- Alison C Kearns
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, MERB 755, 3500 North Broad Street, Philadelphia, PA, 19140, USA
| | - Jake A Robinson
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, MERB 755, 3500 North Broad Street, Philadelphia, PA, 19140, USA
| | - Masoud Shekarabi
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, MERB 755, 3500 North Broad Street, Philadelphia, PA, 19140, USA
| | - Fengming Liu
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, MERB 755, 3500 North Broad Street, Philadelphia, PA, 19140, USA
| | - Xuebin Qin
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, MERB 755, 3500 North Broad Street, Philadelphia, PA, 19140, USA
| | - Tricia H Burdo
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, MERB 755, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
182
|
Butler J, Kalogeropoulos AP, Anstrom KJ, Hsue PY, Kim RJ, Scherzer R, Shah SJ, Shah SH, Velazquez EJ, Hernandez AF, Desvigne-Nickens P, Braunwald E. Diastolic Dysfunction in Individuals With Human Immunodeficiency Virus Infection: Literature Review, Rationale and Design of the Characterizing Heart Function on Antiretroviral Therapy (CHART) Study. J Card Fail 2018; 24:255-265. [PMID: 29482027 PMCID: PMC5880702 DOI: 10.1016/j.cardfail.2018.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 02/06/2018] [Accepted: 02/13/2018] [Indexed: 12/28/2022]
Abstract
Antiretroviral therapy (ART) has been associated with a shift in the epidemiology of human immunodeficiency virus (HIV)-associated cardiomyopathy from a phenotype of primarily left ventricular (LV) systolic dysfunction to LV diastolic dysfunction (DD). Patients with HIV receiving ART have higher rates of DD compared with age-matched control subjects and develop DD at a younger age. However, little is known about the natural history and pathogenesis of DD in virally suppressed HIV-infected patients. Current evidence suggests that immune processes modulate the risk for cardiac involvement in HIV-infected persons. Ongoing inflammation appears to have myocardial effects, and accelerated myocardial fibrosis appears to be a key mediator of HIV-induced DD. The Characterizing Heart Function on Antiretroviral Therapy (CHART) study aims to systematically investigate determinants, mechanisms, and consequences of DD in HIV-infected patients. We will compare ART-treated virally suppressed HIV-infected individuals with and without DD and HIV- individuals with DD regarding (1) systemic inflammation, myocardial stress, and subclinical myocardial necrosis as indicated by circulating biomarkers; (2) immune system activation as indicated by cell surface receptors; (3) myocardial fibrosis according to cardiac magnetic resonance examination; (4) markers of fibrosis and remodeling, oxidative stress, and hypercoagulability; (5) left atrial function according to echocardiographic examination; (6) myocardial stress and subclinical necrosis as indicated by circulating biomarkers; (7) proteomic and metabolic profiles; and (8) phenotype signatures derived from clinical, biomarker, and imaging data.
Collapse
Affiliation(s)
- Javed Butler
- Cardiology Division, Department of Medicine, Stony Brook University, Stony Brook, New York.
| | | | - Kevin J Anstrom
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Priscilla Y Hsue
- Cardiology Division, Department of Medicine, University of California, San Francisco, California
| | - Raymond J Kim
- Cardiology Division, Department of Medicine, Duke University, Durham, North Carolina
| | - Rebecca Scherzer
- UCSF Department of Medicine and San Francisco Department of Veterans Administration, San Francisco, California
| | - Sanjiv J Shah
- Cardiology Division, Department of Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois
| | - Svati H Shah
- Cardiology Division, Department of Medicine, Duke University, Durham, North Carolina
| | - Eric J Velazquez
- Duke Clinical Research Institute, Duke University, Durham, North Carolina; Cardiology Division, Department of Medicine, Duke University, Durham, North Carolina
| | - Adrian F Hernandez
- Cardiology Division, Department of Medicine, Duke University, Durham, North Carolina
| | - Patrice Desvigne-Nickens
- Division of Cardiovascular Sciences, National, Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Eugene Braunwald
- Cardiology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
183
|
Shimamoto Y, Nio-Kobayashi J, Watarai H, Nagano M, Saito N, Takahashi E, Higuchi H, Kobayashi A, Kimura T, Kitamura H. Generation and validation of novel anti-bovine CD163 monoclonal antibodies ABM-1A9 and ABM-2D6. Vet Immunol Immunopathol 2018; 198:6-13. [DOI: 10.1016/j.vetimm.2018.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/14/2018] [Accepted: 02/12/2018] [Indexed: 12/28/2022]
|
184
|
Hileman CO, McComsey GA. Short Communication: The Effect of Rosuvastatin on Vascular Disease Differs by Smoking Status in Treated HIV Infection. AIDS Res Hum Retroviruses 2018; 34:282-285. [PMID: 28974102 DOI: 10.1089/aid.2017.0164] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Smoking is an important contributor to cardiovascular disease risk and is highly prevalent in the HIV population. In the Stopping Atherosclerosis and Treating Unhealthy Bone with Rosuvastatin in HIV trial (SATURN-HIV), a 96-week, randomized placebo-controlled study testing the effect of rosuvastatin on subclinical vascular disease and immune activation in HIV-infected adults, rosuvastatin improved immune activation and arrested common carotid artery intima media thickness (CCA IMT) progression. In this exploratory analysis, ANOVA was used to test for effect modification by smoking. One-hundred forty-seven adults were included (72 in rosuvastatin group; 75 in placebo group). Groups were similar at baseline. Overall, mean ± SD age was 45.4 ± 9.9 years, 115 (78%) were men and 100 (68%) were African American. Ninety-three (63%) were current smokers (mean ± SD 0.6 ± 0.44 packs/day) and another 24 (16%) were smokers in the past. There were statistically significant randomization group by smoking status interactions for 0-24 (p = .01) and 0-48 (p < .01) week changes in proportion of activated CD4+ T cells and for 0-48 (p < .01) and 0-96 (trend only; p = .06) week changes in CCA IMT. No effect modification by smoking was detected for changes in markers of inflammation or monocyte activation. The beneficial effect of rosuvastatin on CCA IMT was not apparent in smokers although T cell activation improved to a greater degree in this subgroup.
Collapse
Affiliation(s)
- Corrilynn O. Hileman
- Division of Infectious Diseases, Department of Medicine, MetroHealth Medical Center, Cleveland, Ohio
- Case Western Reserve University, Cleveland, Ohio
| | - Grace A. McComsey
- Case Western Reserve University, Cleveland, Ohio
- Departments of Pediatrics and Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
185
|
McGettrick P, Barco EA, Mallon PWG. Ageing with HIV. Healthcare (Basel) 2018; 6:healthcare6010017. [PMID: 29443936 PMCID: PMC5872224 DOI: 10.3390/healthcare6010017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/29/2018] [Accepted: 02/07/2018] [Indexed: 12/13/2022] Open
Abstract
The population of people living with HIV (PLWH) is growing older with an estimated 4 million over the age of 50 years, a figure which has doubled since the introduction of effective antiretroviral therapy (ART) and which is increasing globally. Despite effective ART, PLWH still experience excess morbidity and mortality compared to the general population with increased prevalence of age-related, non-AIDS illnesses (NAI) such as cardiovascular disease, malignancies, cognitive impairment and reduced bone mineral density, which impact disability and everyday functioning. This review will discuss the challenges presented by comorbidities in ageing PLWH and discuss the aetiology and management of age-related illnesses in this vulnerable population.
Collapse
Affiliation(s)
- Padraig McGettrick
- HIV Molecular Research Group, UCD School of Medicine, University College Dublin, Dublin, Ireland.
- Mater Misericordae University Hospital, Eccles street, Dublin 7, Ireland.
| | - Elena Alvarez Barco
- HIV Molecular Research Group, UCD School of Medicine, University College Dublin, Dublin, Ireland.
| | - Patrick W G Mallon
- HIV Molecular Research Group, UCD School of Medicine, University College Dublin, Dublin, Ireland.
- Mater Misericordae University Hospital, Eccles street, Dublin 7, Ireland.
| |
Collapse
|
186
|
Mallard J, Williams K. An SIV macaque model of SIV and HAND: the need for adjunctive therapies in HIV that target activated monocytes and macrophages. J Neurovirol 2018; 24:213-219. [PMID: 29435829 DOI: 10.1007/s13365-018-0616-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 12/24/2022]
Abstract
Non-human primate models of AIDS and neuroAIDS are critical to study HIV infection of the CNS, neuropathology, and immune activation and macrophage accumulation that occurs in HAND. SIV, similar to HIV, infects CD4+ T lymphocytes and monocytes/macrophages. Virus enters the CNS early, and macrophage activation correlates with CNS disease, as well as inflammation outside of the CNS. Antiretroviral in HIV+ humans and SIV+ Rhesus macaques results in non-detectable plasma virus, decreased or non-detectable viral RNA or protein in the CNS. But, viral DNA rebounds following therapy interruption, demonstrating the presence of replication competent virus in the CNS within myeloid cells. In this brief review, we discuss our findings using a Rhesus macaque model of SIV-associated CNS infection and pathology, focusing on monocyte/macrophage activation and the link between CNS and cardiac disease. We conclude with recent studies using adjunctive therapy targeting monocytes/macrophages with ART to prevent or diminish CNS pathology that may be associated with HAND.
Collapse
Affiliation(s)
- Jaclyn Mallard
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | - Kenneth Williams
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
187
|
Cournoyer JM, Garms AP, Thiessen KN, Bowers MT, Johnson MD, Relf MV. Cardiovascular Disease and HIV: Pathophysiology, Treatment Considerations, and Nursing Implications. Crit Care Nurse 2018; 36:37-46. [PMID: 27694356 DOI: 10.4037/ccn2016839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
HIV infection has progressed from an acute, terminal disease to a chronic illness with cardiovascular disease as the leading cause of death among persons living with HIV. As persons living with HIV infection continue to become older, traditional risk factors for atherosclerosis compounded by the pathophysiological effects of HIV infection and antiretroviral therapy markedly increase the risk for cardiovascular disease. Further, persons living with HIV are also at high risk for cardiomyopathy. Critical care nurses must recognize the risk factors for cardiovascular disease and the pathophysiology and complex treatment options in order to manage care of these patients and facilitate multidisciplinary collaboration. Two case studies are used to highlight the treatment options and nursing considerations associated with cardiovascular disease among persons living with HIV.
Collapse
Affiliation(s)
- Justin M Cournoyer
- Justin M. Cournoyer is a clinical nurse I in the pediatric cardiac intensive care unit, Duke University Hospital, Durham, North Carolina.Aven P. Garms is a clinical nurse I in the intensive care nursery, Duke University Hospital.Kimberly N. Thiessen is a staff nurse in the pediatric emergency department, WakeMed Health and Hospitals, Raleigh, North Carolina.Margaret T. Bowers is an associate professor and the faculty coordinator of the adult/geriatric nurse practitioner program and the lead faculty member for the adult/geriatric nurse practitioner-cardiovascular specialty at Duke University School of Nursing, Durham, North Carolina.Melissa D. Johnson is an associate professor in medicine, Duke University Medical Center, Durham, North Carolina, and Campbell University, Buies Creek, North Carolina.Michael V. Relf is an associate professor and the associate dean for global and community affairs, Duke University School of Nursing
| | - Aven P Garms
- Justin M. Cournoyer is a clinical nurse I in the pediatric cardiac intensive care unit, Duke University Hospital, Durham, North Carolina.Aven P. Garms is a clinical nurse I in the intensive care nursery, Duke University Hospital.Kimberly N. Thiessen is a staff nurse in the pediatric emergency department, WakeMed Health and Hospitals, Raleigh, North Carolina.Margaret T. Bowers is an associate professor and the faculty coordinator of the adult/geriatric nurse practitioner program and the lead faculty member for the adult/geriatric nurse practitioner-cardiovascular specialty at Duke University School of Nursing, Durham, North Carolina.Melissa D. Johnson is an associate professor in medicine, Duke University Medical Center, Durham, North Carolina, and Campbell University, Buies Creek, North Carolina.Michael V. Relf is an associate professor and the associate dean for global and community affairs, Duke University School of Nursing
| | - Kimberly N Thiessen
- Justin M. Cournoyer is a clinical nurse I in the pediatric cardiac intensive care unit, Duke University Hospital, Durham, North Carolina.Aven P. Garms is a clinical nurse I in the intensive care nursery, Duke University Hospital.Kimberly N. Thiessen is a staff nurse in the pediatric emergency department, WakeMed Health and Hospitals, Raleigh, North Carolina.Margaret T. Bowers is an associate professor and the faculty coordinator of the adult/geriatric nurse practitioner program and the lead faculty member for the adult/geriatric nurse practitioner-cardiovascular specialty at Duke University School of Nursing, Durham, North Carolina.Melissa D. Johnson is an associate professor in medicine, Duke University Medical Center, Durham, North Carolina, and Campbell University, Buies Creek, North Carolina.Michael V. Relf is an associate professor and the associate dean for global and community affairs, Duke University School of Nursing
| | - Margaret T Bowers
- Justin M. Cournoyer is a clinical nurse I in the pediatric cardiac intensive care unit, Duke University Hospital, Durham, North Carolina.Aven P. Garms is a clinical nurse I in the intensive care nursery, Duke University Hospital.Kimberly N. Thiessen is a staff nurse in the pediatric emergency department, WakeMed Health and Hospitals, Raleigh, North Carolina.Margaret T. Bowers is an associate professor and the faculty coordinator of the adult/geriatric nurse practitioner program and the lead faculty member for the adult/geriatric nurse practitioner-cardiovascular specialty at Duke University School of Nursing, Durham, North Carolina.Melissa D. Johnson is an associate professor in medicine, Duke University Medical Center, Durham, North Carolina, and Campbell University, Buies Creek, North Carolina.Michael V. Relf is an associate professor and the associate dean for global and community affairs, Duke University School of Nursing
| | - Melissa D Johnson
- Justin M. Cournoyer is a clinical nurse I in the pediatric cardiac intensive care unit, Duke University Hospital, Durham, North Carolina.Aven P. Garms is a clinical nurse I in the intensive care nursery, Duke University Hospital.Kimberly N. Thiessen is a staff nurse in the pediatric emergency department, WakeMed Health and Hospitals, Raleigh, North Carolina.Margaret T. Bowers is an associate professor and the faculty coordinator of the adult/geriatric nurse practitioner program and the lead faculty member for the adult/geriatric nurse practitioner-cardiovascular specialty at Duke University School of Nursing, Durham, North Carolina.Melissa D. Johnson is an associate professor in medicine, Duke University Medical Center, Durham, North Carolina, and Campbell University, Buies Creek, North Carolina.Michael V. Relf is an associate professor and the associate dean for global and community affairs, Duke University School of Nursing
| | - Michael V Relf
- Justin M. Cournoyer is a clinical nurse I in the pediatric cardiac intensive care unit, Duke University Hospital, Durham, North Carolina.Aven P. Garms is a clinical nurse I in the intensive care nursery, Duke University Hospital.Kimberly N. Thiessen is a staff nurse in the pediatric emergency department, WakeMed Health and Hospitals, Raleigh, North Carolina.Margaret T. Bowers is an associate professor and the faculty coordinator of the adult/geriatric nurse practitioner program and the lead faculty member for the adult/geriatric nurse practitioner-cardiovascular specialty at Duke University School of Nursing, Durham, North Carolina.Melissa D. Johnson is an associate professor in medicine, Duke University Medical Center, Durham, North Carolina, and Campbell University, Buies Creek, North Carolina.Michael V. Relf is an associate professor and the associate dean for global and community affairs, Duke University School of Nursing.
| |
Collapse
|
188
|
High Prevalence of Hypertension in Ethiopian and Non-Ethiopian HIV-Infected Adults. Int J Hypertens 2018; 2018:8637101. [PMID: 29623220 PMCID: PMC5830020 DOI: 10.1155/2018/8637101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/03/2018] [Indexed: 02/08/2023] Open
Abstract
Objectives Prevalence of hypertension has not been studied in the Ethiopian HIV-infected population, which represents 60% of the patients in our AIDS unit. Our aim was to identify risk factors and characterize the prevalence of hypertension in the population monitored at our unit. Methods A retrospective chart review categorized subjects according to their blood pressure levels. Hypertension prevalence was determined and stratified according to variables perceived to contribute to elevated blood pressure. Results The prevalence of hypertension in our study population was significantly higher compared to the general population (53% versus 20%, P < 0.0001) and was associated with known risk factors and not with patients' viral load and CD4 levels. Ethiopian HIV-infected adults had a prominently higher rate of blood pressure rise over time as compared to non-Ethiopians (P = 0.016). Conclusions The high prevalence of hypertension in this cohort and the rapid increase in blood pressure in Ethiopians are alarming. We could not attribute high prevalence to HIV-related factors and we presume it is part of the metabolic syndrome. The lifelong cardiovascular risk associated with HIV infection mandates hypertension screening and close monitoring in this population.
Collapse
|
189
|
Gootenberg DB, Paer JM, Luevano JM, Kwon DS. HIV-associated changes in the enteric microbial community: potential role in loss of homeostasis and development of systemic inflammation. Curr Opin Infect Dis 2018; 30:31-43. [PMID: 27922852 PMCID: PMC5325247 DOI: 10.1097/qco.0000000000000341] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Supplemental Digital Content is available in the text Purpose of review Despite HIV therapy advances, average life expectancy in HIV-infected individuals on effective treatment is significantly decreased relative to uninfected persons, largely because of increased incidence of inflammation-related diseases, such as cardiovascular disease and renal dysfunction. The enteric microbial community could potentially cause this inflammation, as HIV-driven destruction of gastrointestinal CD4+ T cells may disturb the microbiota–mucosal immune system balance, disrupting the stable gut microbiome and leading to further deleterious host outcomes. Recent findings Varied enteric microbiome changes have been reported during HIV infection, but unifying patterns have emerged. Community diversity is decreased, similar to pathologies such as inflammatory bowel disease, obesity, and Clostridium difficile infection. Many taxa frequently enriched in HIV-infected individuals, such as Enterobacteriaceae and Erysipelotrichaceae, have pathogenic potential, whereas depleted taxa, such as Bacteroidaceae and Ruminococcaceae, are more linked with anti-inflammatory properties and maintenance of gut homeostasis. The gut viral community in HIV has been found to contain a greater abundance of pathogenesis-associated Adenoviridae and Anelloviridae. These bacterial and viral changes correlate with increased systemic inflammatory markers, such as serum sCD14, sCD163, and IL-6. Summary Enteric microbial community changes may contribute to chronic HIV pathogenesis, but more investigation is necessary, especially in the developing world population with the greatest HIV burden (Video, Supplemental Digital Content 1, which includes the authors’ summary of the importance of the work).
Collapse
Affiliation(s)
- David B Gootenberg
- aRagon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Cambridge bHarvard Medical School, Boston cDivision of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
190
|
Abstract
OBJECTIVE To determine the association of smoking and HIV status with tissue-specific inflammation measured by flurodeoxyglucose positron emission tomography (PET). DESIGN A cross-sectional study. METHODS We prospectively enrolled 55 HIV study participants on stable antiretroviral therapy and 19 age-matched HIV-uninfected controls without known cardiovascular disease. We measured aortic target-to-background ratio (TBR) and spleen standardized uptake values (SUV) 3-h post-FDG, and used regression models to examine the independent association of HIV and smoking status with PET variables. RESULTS Overall, median (interquartile range) age was 50 (42-55) years; 81% were men and 54% were current smokers (median 0.5 packs/day, 25 pack-years]. Median CD4 of HIV study participants was 690 cells/ml and 88% had HIV-1 RNA less than 20 c/ml; 43% were on a protease inhibitor. In fully adjusted models, HIV was associated with 0.16 (95% confidence interval 0.04-0.27; P = 0.009) higher aortic TBR, whereas current smoking was marginally associated with a lower TBR [-0.11 (95% confidence interval -0.23 to 0.01); P = 0.07]. Spleen SUVmean was not associated with HIV or smoking, and there was no evidence for an HIVsmoking interaction for aortic or spleen models (all P > 0.1). Spleen SUVmean was positively associated with biomarkers of inflammation and coronary artery calcium, but adjustment for traditional cardiovascular disease risk factors attenuated these relationships. CONCLUSION The FDG-PET study of HIV study participants suggests that HIV is associated with increased aortic inflammation independent of traditional risk factors, but smoking is not. Future studies should continue to explore the mechanistic roles of smoking and inflammation at various stages of clinical and subclinical atherosclerotic vascular disease in HIV.
Collapse
|
191
|
GRINSPOON STEVEN. NOVEL MECHANISMS AND ANTI-INFLAMMATORY STRATEGIES TO REDUCE CARDIOVASCULAR RISK IN HUMAN IMMUNODEFICIENCY VIRUS. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2018; 129:140-154. [PMID: 30166708 PMCID: PMC6116636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cardiovascular disease (CVD) rates are 50% to 100% higher in human immunodeficiency virus (HIV) patients. Traditional risks account for only 25% of this excess risk. Excess CVD risk in HIV may relate in part to increases in ectopic adipose depots. CVD in HIV-infection is characterized by atypical highly vulnerable plaque lesions, which are inflamed, in tight relationship to immune, and monocyte activation pathways. Using 18fluorine-2-deoxy-D-glucose positron-emission tomography imaging techniques, we have shown increased arterial inflammation which persists even after effective antiretroviral therapy. More recent studies, using a novel macrophage specific imaging agent in humans, have shown highly increased inflammatory patterns in large vessels in HIV. In addition, statins may be uniquely valuable among HIV patients, not only lowering low-density lipoprotein, but also reducing monocyte chemo-attraction, and immune activation pathways. These data have led the National Institutes of Health to fund the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE), the first large multicenter primary CVD prevention trial in HIV, aimed at assessing inflammatory mechanisms of CVD in HIV.
Collapse
Affiliation(s)
- STEVEN GRINSPOON
- Correspondence and reprint requests: Steven Grinspoon, MD, Harvard Medical School,
5 Longfellow Place, Room 207, Boston, Massachusetts, 02114617-724-9109617-724-8998
| |
Collapse
|
192
|
Buscher K, Marcovecchio P, Hedrick CC, Ley K. Patrolling Mechanics of Non-Classical Monocytes in Vascular Inflammation. Front Cardiovasc Med 2017; 4:80. [PMID: 29312957 PMCID: PMC5742122 DOI: 10.3389/fcvm.2017.00080] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/23/2017] [Indexed: 12/13/2022] Open
Abstract
Non-classical monocytes have emerged as the preeminent vascular housekeepers. Continuous intravascular screening is enabled by slow patrolling on the endothelium and allows a rapid response to local perturbations. Intravital imaging has been crucial to elucidate the molecular mechanisms and migratory phenotype of patrolling. In this review, we discuss technical requirements of intravital microscopy such as imaging modalities, labeling strategies, and data analysis. We further focus on patrolling kinetics and adhesion receptors in different organs and vascular beds including arteries during homeostasis and vascular inflammation and define pertinent questions in the field.
Collapse
Affiliation(s)
- Konrad Buscher
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States.,Department of Internal Medicine, Nephrology and Rheumatology, University Hospital Münster, Münster, Germany
| | - Paola Marcovecchio
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| |
Collapse
|
193
|
Kaplan-Lewis E, Aberg JA, Lee M. Atherosclerotic Cardiovascular Disease and Anti-Retroviral Therapy. Curr HIV/AIDS Rep 2017; 13:297-308. [PMID: 27562769 DOI: 10.1007/s11904-016-0331-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In the current era of available therapy for human immunodeficiency virus (HIV), life expectancy for persons living with HIV (PLWH) nears that of the general population. Atherosclerotic cardiovascular disease (ASCVD) has become a particular burden for PLWH and society at large. PLWH have historically been shown to have an excess of cardiovascular risk and subsequent events when compared to the general population. Potential explanations include the increased prevalence of traditional risk factors, direct inflammatory and immunological effects from the HIV virus itself, and metabolic adverse effects of anti-retroviral therapy (ART). Over the past few years, there has been building evidence that chronic inflammation and immune activation independent of virologic suppression contribute significantly to excess ASCVD risk. Although independent agents and combination therapies have varying metabolic effects, the evidence from major randomized controlled trials (RCTs) supports the benefits of early initiation of ART. In this review, we will discuss the epidemiology of ASCVD in HIV-infected patients compared with the general population, give an overview of potential pathogenesis of high-risk plaque in HIV-infected patients, discuss different metabolic effects of individual anti-retrovirals, and discuss the limitations in current screening models for assessing cardiovascular disease (CVD) risk and future directions for treatment.
Collapse
Affiliation(s)
- Emma Kaplan-Lewis
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1090, New York, NY, 10029, USA
| | - Judith A Aberg
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1090, New York, NY, 10029, USA
| | - Mikyung Lee
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1090, New York, NY, 10029, USA.
| |
Collapse
|
194
|
Nowlin BT, Wang J, Schafer JL, Autissier P, Burdo TH, Williams KC. Monocyte subsets exhibit transcriptional plasticity and a shared response to interferon in SIV-infected rhesus macaques. J Leukoc Biol 2017; 103:141-155. [PMID: 29345061 DOI: 10.1002/jlb.4a0217-047r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 09/26/2017] [Accepted: 10/16/2017] [Indexed: 12/24/2022] Open
Abstract
The progression to AIDS is influenced by changes in the biology of heterogeneous monocyte subsets. Classical (CD14++CD16-), intermediate (CD14++CD16+), and nonclassical (CD14+CD16++) monocytes may represent progressive stages of monocyte maturation or disparate myeloid lineages with different turnover rates and function. To investigate the relationship between monocyte subsets and the response to SIV infection, we performed microarray analysis of monocyte subsets in rhesus macaques at three time points: prior to SIV infection, 26 days postinfection, and necropsy with AIDS. Genes with a 2-fold change between monocyte subsets (2023 genes) or infection time points (424 genes) were selected. We identify 172 genes differentially expressed among monocyte subsets in both uninfected and SIV-infected animals. Classical monocytes express genes associated with inflammatory responses and cell proliferation. Nonclassical monocytes express genes associated with activation, immune effector functions, and cell cycle inhibition. The classical and intermediate subsets are most similar at all time points, and transcriptional similarity between intermediate and nonclassical monocytes increases with AIDS. Cytosolic sensors of nucleic acids, restriction factors, and IFN-stimulated genes are induced in all three subsets with AIDS. We conclude that SIV infection alters the transcriptional relationship between monocyte subsets and that the innate immune response to SIV infection is conserved across monocyte subsets.
Collapse
Affiliation(s)
- Brian T Nowlin
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - John Wang
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Jamie L Schafer
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Patrick Autissier
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Tricia H Burdo
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Kenneth C Williams
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| |
Collapse
|
195
|
Muenchhoff M, Adland E, Karimanzira O, Crowther C, Pace M, Csala A, Leitman E, Moonsamy A, McGregor C, Hurst J, Groll A, Mori M, Sinmyee S, Thobakgale C, Tudor-Williams G, Prendergast AJ, Kloverpris H, Roider J, Leslie A, Shingadia D, Brits T, Daniels S, Frater J, Willberg CB, Walker BD, Ndung'u T, Jooste P, Moore PL, Morris L, Goulder P. Nonprogressing HIV-infected children share fundamental immunological features of nonpathogenic SIV infection. Sci Transl Med 2017; 8:358ra125. [PMID: 27683550 DOI: 10.1126/scitranslmed.aag1048] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/22/2016] [Indexed: 12/14/2022]
Abstract
Disease-free infection in HIV-infected adults is associated with human leukocyte antigen-mediated suppression of viremia, whereas in the sooty mangabey and other healthy natural hosts of simian immunodeficiency virus (SIV), viral replication continues unabated. To better understand factors preventing HIV disease, we investigated pediatric infection, where AIDS typically develops more rapidly than in adults. Among 170 nonprogressing antiretroviral therapy-naïve children aged >5 years maintaining normal-for-age CD4 T cell counts, immune activation levels were low despite high viremia (median, 26,000 copies/ml). Potent, broadly neutralizing antibody responses in most of the subjects and strong virus-specific T cell activity were present but did not drive pediatric nonprogression. However, reduced CCR5 expression and low HIV infection in long-lived central memory CD4 T cells were observed in pediatric nonprogressors. These children therefore express two cardinal immunological features of nonpathogenic SIV infection in sooty mangabeys-low immune activation despite high viremia and low CCR5 expression on long-lived central memory CD4 T cells-suggesting closer similarities with nonpathogenetic mechanisms evolved over thousands of years in natural SIV hosts than those operating in HIV-infected adults.
Collapse
Affiliation(s)
- Maximilian Muenchhoff
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. Max von Pettenkofer-Institute, Department of Virology, Ludwig-Maximilians-University Munich, Munich, Germany. German Center for Infection Research (DZIF), Partner Site Munich, Germany
| | - Emily Adland
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Owen Karimanzira
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Carol Crowther
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Matthew Pace
- Institute for Emerging Infections, Oxford Martin School, University of Oxford, Oxford, U.K. Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford OX1 3SY, U.K. Oxford National Institute of Health Research, Biomedical Research Centre, Oxford, U.K
| | - Anna Csala
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Ellen Leitman
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Angeline Moonsamy
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Callum McGregor
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Jacob Hurst
- Institute of Cancer Research, Old Brompton Road, London SW7 3RP, U.K
| | - Andreas Groll
- Department of Mathematics, Ludwig-Maximilians-University Munich, Theresienstrasse 39, 80333 Munich, Germany
| | - Masahiko Mori
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Smruti Sinmyee
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Christina Thobakgale
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | | | | | - Henrik Kloverpris
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa. Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Julia Roider
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa
| | - Alasdair Leslie
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa
| | - Delane Shingadia
- Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children, London, U.K
| | - Thea Brits
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - Samantha Daniels
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - John Frater
- Institute for Emerging Infections, Oxford Martin School, University of Oxford, Oxford, U.K. Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford OX1 3SY, U.K. Oxford National Institute of Health Research, Biomedical Research Centre, Oxford, U.K
| | - Christian B Willberg
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford OX1 3SY, U.K. Oxford National Institute of Health Research, Biomedical Research Centre, Oxford, U.K
| | - Bruce D Walker
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139-4307, USA
| | - Thumbi Ndung'u
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa. Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139-4307, USA. Max Planck Institute for Infection Biology, Berlin, Germany
| | - Pieter Jooste
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - Penny L Moore
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa. Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa. Center for the AIDS Programme of Research in South Africa (CAPRISA), 4001 Durban, South Africa
| | - Lynn Morris
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa. Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa. Center for the AIDS Programme of Research in South Africa (CAPRISA), 4001 Durban, South Africa
| | - Philip Goulder
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children, London, U.K.
| |
Collapse
|
196
|
Raghavan A, Rimmelin D, Fitch KV, Zanni MV. Sex Differences in Select Non-communicable HIV-Associated Comorbidities: Exploring the Role of Systemic Immune Activation/Inflammation. Curr HIV/AIDS Rep 2017; 14:220-228. [PMID: 29080122 PMCID: PMC6007989 DOI: 10.1007/s11904-017-0366-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF THE REVIEW The goals of this review are to (1) explore HIV-associated cardiovascular disease (CVD), neurocognitive impairment, and non-AIDS-defining cancers (NADC) as heterogeneous model disease states fuelled in part by systemic immune activation/inflammation; (2) consider sex differences in the epidemiology of these diseases in both high-resource and lower-resource settings; and (3) examine biological and environmental factors which may contribute to heightened systemic immune activation/inflammation specifically among women living with HIV (WLHIV). RECENT FINDINGS The observation that WLHIV have higher levels of systemic immune activation/inflammation than men living with HIV (MLHIV) may be relevant to sex differences in select non-communicable HIV-associated comorbidities. Heightened systemic immune activation among WLHIV may be influenced by sex-specific responses to the virus and to immunomodulatory agents, as well as by behavioral choices/comorbid conditions and perturbations in the hypothalamic-pituitary-gonadal axis. Additional research is needed to elucidate region-specific drivers of heightened systemic immune activation/inflammation among WLHIV and to determine whether WLHIV who present with one immune-mediated HIV-associated comorbidity (e.g., cognitive impairment) may be at increased risk for another (e.g., CVD, NADC). This kind of research would facilitate improved risk prediction for non-communicable HIV-associated comorbidities among WLHIV and the development of targeted immunomodulatory prevention strategies.
Collapse
Affiliation(s)
- Avanthi Raghavan
- Massachusetts General Hospital, Program in Nutritional Metabolism, Harvard Medical School
| | - Dodie Rimmelin
- Massachusetts General Hospital, Program in Nutritional Metabolism, Harvard Medical School
| | - Kathleen V. Fitch
- Massachusetts General Hospital, Program in Nutritional Metabolism, Harvard Medical School
| | - Markella V. Zanni
- Massachusetts General Hospital, Program in Nutritional Metabolism, Harvard Medical School
| |
Collapse
|
197
|
Sherman KE, Peters MG, Thomas D. Human immunodeficiency virus and liver disease: A comprehensive update. Hepatol Commun 2017; 1:987-1001. [PMID: 30838978 PMCID: PMC5721407 DOI: 10.1002/hep4.1112] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/29/2017] [Accepted: 09/19/2017] [Indexed: 12/16/2022] Open
Abstract
Among persons living with human immunodeficiency virus (HIV) infection, liver disease remains a major cause of morbidity and mortality. While the etiologies are varied and often overlapping in the individual patient, the underlying mechanisms, including oxidative stress, direct activation of stellate cells, HIV interaction with hepatocytes, and bacterial translocation with systemic immune activation, seem to be unifying characteristics. Early and fully suppressive HIV antiretroviral therapy is a mainstay of management either before or concurrent with treatment of etiologic cofactors, including hepatitis C virus, hepatitis B virus, and nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Significant barriers to care that still exist include liver disease recognition, appropriate linkage to care, ongoing substance abuse, and psychiatric comorbidities in the HIV-infected population. Emerging issues in these patients include acute and chronic hepatitis E, underreported hepatitis D, and a rising incidence of hepatocellular carcinoma. (Hepatology Communications 2017;1:987-1001).
Collapse
|
198
|
Baker JV, Sharma S, Grund B, Rupert A, Metcalf JA, Schechter M, Munderi P, Aho I, Emery S, Babiker A, Phillips A, Lundgren JD, Neaton JD, Lane HC. Systemic Inflammation, Coagulation, and Clinical Risk in the START Trial. Open Forum Infect Dis 2017; 4:ofx262. [PMID: 29308409 PMCID: PMC5751061 DOI: 10.1093/ofid/ofx262] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022] Open
Abstract
Background The Strategic Timing of AntiRetroviral Treatment (START) trial demonstrated that immediate (at CD4+ >500 cells/µL) vs deferred (to CD4+ <350 cells/µL or AIDS) antiretroviral therapy (ART) initiation reduced risk for AIDS and serious non-AIDS (SNA). We investigated associations of inflammation, coagulation, and vascular injury biomarkers with AIDS, SNA or death, and the effect of immediate ART initiation. Methods Biomarkers were measured from stored plasma prior to randomization and at month 8. Associations of baseline biomarkers with event risk were estimated with Cox regression, pooled across groups, adjusted for age, gender, and treatment group, and stratified by region. Mean changes over 8 months were estimated and compared between the immediate and deferred ART arms using analysis of covariance models, adjusted for levels at entry. Results Baseline biomarker levels were available for 4299 START participants (92%). Mean follow-up was 3.2 years. Higher levels of IL-6 and D-dimer were the only biomarkers associated with risk for AIDS, SNA or death, as well as the individual components of SNA and AIDS events (HRs ranged 1.37-1.41 per 2-fold higher level), even after adjustment for baseline CD4+ count, HIV RNA level, and other biomarkers. At month 8, biomarker levels were lower in the immediate arm by 12%-21%. Conclusions These data, combined with evidence from prior biomarker studies, demonstrate that IL-6 and D-dimer consistently predict clinical risk across a broad spectrum of CD4 counts for those both ART-naïve and treated. Research is needed to identify disease-modifying treatments that target inflammation beyond the effects of ART.
Collapse
Affiliation(s)
- Jason V Baker
- Department of Medicine University of Minnesota, Minneapolis, Minnesota.,Division of Biostatistics School of Statistics, University of Minnesota, Minneapolis, Minnesota
| | - Shweta Sharma
- Division of Infectious Diseases, Hennepin County Medical Center, Minneapolis, Minnesota
| | - Birgit Grund
- Division of Biostatistics, School of Public Health University of Minnesota, Minneapolis, Minnesota
| | - Adam Rupert
- Leidos Biomedical Research Inc., Frederick, Maryland
| | - Julia A Metcalf
- National Institute of Allergy and Infectious Diseases, Division of Clinical Research, Bethesda, Maryland
| | - Mauro Schechter
- Projeto Praça Onze, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Inka Aho
- Division of Infectious Diseases, Helsinki University Hospital, Helsinki, Finland
| | - Sean Emery
- Faculty of Medicine, University of Queensland, Brisbane, Australia.,Kirby Institute, University of New South Wales, Sydney, Australia
| | - Abdel Babiker
- MRC Clinical Trials Unit, University College London, London, UK
| | - Andrew Phillips
- HIV Epidemiology and Biostatistics Group, University College London, London, UK
| | - Jens D Lundgren
- CHIP, Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - James D Neaton
- Division of Infectious Diseases, Hennepin County Medical Center, Minneapolis, Minnesota
| | - H Clifford Lane
- National Institute of Allergy and Infectious Diseases, Division of Clinical Research, Bethesda, Maryland
| | | |
Collapse
|
199
|
HIV protease inhibitor-induced cardiac dysfunction and fibrosis is mediated by platelet-derived TGF-β1 and can be suppressed by exogenous carbon monoxide. PLoS One 2017; 12:e0187185. [PMID: 29088262 PMCID: PMC5663426 DOI: 10.1371/journal.pone.0187185] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 10/16/2017] [Indexed: 12/24/2022] Open
Abstract
Human immunodeficiency virus (HIV) infection is an independent risk factor for cardiovascular disease. This risk is magnified by certain antiretrovirals, particularly the protease inhibitor ritonavir, but the pathophysiology of this connection is unknown. We postulated that a major mechanism for antiretroviral-associated cardiac disease is pathologic fibrosis linked to platelet activation with release and activation of transforming growth factor (TGF)-β1, and that these changes could be modeled in a murine system. We also sought to intervene utilizing inhaled carbon monoxide (CO) as proof-of-concept for therapeutics capable of regulating TGF-β1 signaling and collagen autophagy. We demonstrate decreased cardiac function indices, including cardiac output, ejection fraction and stroke volume, and prominent cardiac fibrosis, in mice exposed to pharmacological doses of ritonavir. Cardiac output and fibrosis correlated with plasma TGF-β1 levels. Mice with targeted deletion of TGF-β1 in megakaryocytes/platelets (PF4CreTgfb1flox/flox) were partially protected from ritonavir-induced cardiac dysfunction and fibrosis. Inhalation of low dose CO (250ppm), used as a surrogate for upregulation of inducible heme oxygenase/endogenous CO pathways, suppressed ritonavir-induced cardiac fibrosis. This occurred in association with modulation of canonical (Smad2) and non-canonical (p38) TGF-β1 signaling pathways. In addition, CO treatment suppressed the M1 pro-inflammatory subset of macrophages and increased M2c regulatory cells in the hearts of RTV-exposed animals. The effects of CO were dependent upon autophagy as CO did not mitigate ritonavir-induced fibrosis in autophagy-deficient LC3-/- mice. These results suggest that platelet-derived TGF-β1 contributes to ritonavir-associated cardiac dysfunction and fibrosis, extending the relevance of our findings to other antiretrovirals that also activate platelets. The anti-fibrotic effects of CO are linked to alterations in TGF-β1 signaling and autophagy, suggesting a proof-of-concept for novel interventions in HIV/antiretroviral therapy-mediated cardiovascular disease.
Collapse
|
200
|
Abstract
PURPOSE OF REVIEW This review focuses on the differential effects of contemporary antiretrovirals on systemic inflammation as heightened immune activation is linked to important co-morbidities and mortality with HIV infection. RECENT FINDINGS Antiretroviral therapy (ART) reduces dramatically systemic inflammation and immune activation, but not to levels synchronous with HIV-uninfected populations. In one ART initiation trial, integrase inhibitors appear to reduce inflammation to a greater degree than non-nucleoside reverse transcriptase inhibitors (NNRTIs); however, it is not clear that there are beneficial effects on inflammation resulting from treatment with integrase inhibitors compared to PIs, between PIs and NNRTIs, between specific nucleoside reverse transcriptase inhibitors, or with maraviroc in ART-naïve patients. In ART switch studies, changing to an integrase inhibitor from a PI-, NNRTI-, or enfuvirtide-containing regimen has resulted in improvement in several markers of inflammation. Additional research is needed to conclusively state whether there are clear differences in effects of specific antiretrovirals on inflammation and immune activation in HIV.
Collapse
|