1
|
Rodriguez NR, Fortune T, Hegde E, Weinstein MP, Keane AM, Mangold JF, Swartz TH. Oxidative phosphorylation in HIV-1 infection: impacts on cellular metabolism and immune function. Front Immunol 2024; 15:1360342. [PMID: 38529284 PMCID: PMC10962326 DOI: 10.3389/fimmu.2024.1360342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/26/2024] [Indexed: 03/27/2024] Open
Abstract
Human Immunodeficiency Virus Type 1 (HIV-1) presents significant challenges to the immune system, predominantly characterized by CD4+ T cell depletion, leading to Acquired Immunodeficiency Syndrome (AIDS). Antiretroviral therapy (ART) effectively suppresses the viral load in people with HIV (PWH), leading to a state of chronic infection that is associated with inflammation. This review explores the complex relationship between oxidative phosphorylation, a crucial metabolic pathway for cellular energy production, and HIV-1, emphasizing the dual impact of HIV-1 infection and the metabolic and mitochondrial effects of ART. The review highlights how HIV-1 infection disrupts oxidative phosphorylation, promoting glycolysis and fatty acid synthesis to facilitate viral replication. ART can exacerbate metabolic dysregulation despite controlling viral replication, impacting mitochondrial DNA synthesis and enhancing reactive oxygen species production. These effects collectively contribute to significant changes in oxidative phosphorylation, influencing immune cell metabolism and function. Adenosine triphosphate (ATP) generated through oxidative phosphorylation can influence the metabolic landscape of infected cells through ATP-detected purinergic signaling and contributes to immunometabolic dysfunction. Future research should focus on identifying specific targets within this pathway and exploring the role of purinergic signaling in HIV-1 pathogenesis to enhance HIV-1 treatment modalities, addressing both viral infection and its metabolic consequences.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Talia H. Swartz
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
2
|
Feng S, Tang D, Wang Y, Li X, Bao H, Tang C, Dong X, Li X, Yang Q, Yan Y, Yin Z, Shang T, Zheng K, Huang X, Wei Z, Wang K, Qi S. The mechanism of ferroptosis and its related diseases. MOLECULAR BIOMEDICINE 2023; 4:33. [PMID: 37840106 PMCID: PMC10577123 DOI: 10.1186/s43556-023-00142-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/23/2023] [Indexed: 10/17/2023] Open
Abstract
Ferroptosis, a regulated form of cellular death characterized by the iron-mediated accumulation of lipid peroxides, provides a novel avenue for delving into the intersection of cellular metabolism, oxidative stress, and disease pathology. We have witnessed a mounting fascination with ferroptosis, attributed to its pivotal roles across diverse physiological and pathological conditions including developmental processes, metabolic dynamics, oncogenic pathways, neurodegenerative cascades, and traumatic tissue injuries. By unraveling the intricate underpinnings of the molecular machinery, pivotal contributors, intricate signaling conduits, and regulatory networks governing ferroptosis, researchers aim to bridge the gap between the intricacies of this unique mode of cellular death and its multifaceted implications for health and disease. In light of the rapidly advancing landscape of ferroptosis research, we present a comprehensive review aiming at the extensive implications of ferroptosis in the origins and progress of human diseases. This review concludes with a careful analysis of potential treatment approaches carefully designed to either inhibit or promote ferroptosis. Additionally, we have succinctly summarized the potential therapeutic targets and compounds that hold promise in targeting ferroptosis within various diseases. This pivotal facet underscores the burgeoning possibilities for manipulating ferroptosis as a therapeutic strategy. In summary, this review enriched the insights of both investigators and practitioners, while fostering an elevated comprehension of ferroptosis and its latent translational utilities. By revealing the basic processes and investigating treatment possibilities, this review provides a crucial resource for scientists and medical practitioners, aiding in a deep understanding of ferroptosis and its effects in various disease situations.
Collapse
Affiliation(s)
- Shijian Feng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Dan Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yichang Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiang Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Bao
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chengbing Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiuju Dong
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xinna Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qinxue Yang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yun Yan
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhijie Yin
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tiantian Shang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Kaixuan Zheng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaofang Huang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zuheng Wei
- Chengdu Jinjiang Jiaxiang Foreign Languages High School, Chengdu, People's Republic of China
| | - Kunjie Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Shiqian Qi
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
3
|
Akiso M, Ameka M, Naidoo K, Langat R, Kombo J, Sikuku D, Ndung’u T, Altfeld M, Anzala O, Mureithi M. Metabolic and mitochondrial dysregulation in CD4+ T cells from HIV-positive women on combination anti-retroviral therapy. PLoS One 2023; 18:e0286436. [PMID: 37816026 PMCID: PMC10564234 DOI: 10.1371/journal.pone.0286436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/20/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND For optimal functionality, immune cells require a robust and adaptable metabolic program that is fueled by dynamic mitochondrial activity. In this study, we investigate the metabolic alterations occurring in immune cells during HIV infection and antiretroviral therapy by analyzing the uptake of metabolic substrates and mitochondrial phenotypes. By delineating changes in immune cell metabolic programming during HIV, we may identify novel potential therapeutic targets to improve anti-viral immune responses. METHODS After consent and voluntary participation was confirmed, whole blood was drawn from HIV uninfected women and women with chronic HIV infection on long-term combination antiretroviral therapy (HIV/cART). Peripheral blood mononuclear cells-derived immune cells were directly incubated with different fluorescently tagged metabolites and markers of mitochondrial activity: FITC-2-NBDG (2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl) amino]-2-deoxy-D-glucose), FITC-BODIPY (4,4-Difluoro-5,7-Dimethyl-4-Bora-3a,4a-Diaza-s-Indacene-3-Hexadecanoic Acid), FITC-MitoTracker Green and APC-MitoTracker Deep Red. The uptake of glucose and fats and the mitochondrial mass and potential were measured using flow cytometry. All values are reported quantitatively as geometric means of fluorescence intensity. RESULTS During chronic HIV infection, cellular uptake of glucose increases in HIV+ dendritic cells in particular. CD4+ T cells had the lowest uptake of glucose and fats compared to all other cells regardless of HIV status, while CD8+ T cells took up more fatty acids. Interestingly, despite the lower utilization of glucose and fats in CD4+ T cells, mitochondrial mass increased in HIV+ CD4+ T cells compared to HIV negative CD4+ T-cells. HIV+ CD4+ T cells also had the highest mitochondrial potential. CONCLUSIONS Significant disparities in the utilization of substrates by leukocytes during chronic HIV/cART exist. Innate immune cells increased utilization of sugars and fats while adaptive immune cells displayed lower glucose and fat utilization despite having a higher mitochondrial activity. Our findings suggest that cART treated HIV-infected CD4+ T cells be dysfunctional or may prefer alternative fuel sources not included in these studies. This underscores the importance of understanding the metabolic effects of HIV treatment on immune function.
Collapse
Affiliation(s)
- Matrona Akiso
- Department of Medical Microbiology & Immunology, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| | - Magdalene Ameka
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| | - Kewreshini Naidoo
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Robert Langat
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
- Division of Surgical Outcomes and Precision Medicine Research, Department of Surgery, University of Minnesota Twin Cities, United States of America
| | - Janet Kombo
- Department of Medical Microbiology & Immunology, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| | - Delories Sikuku
- Department of Medical Microbiology & Immunology, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
| | - Thumbi Ndung’u
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Marcus Altfeld
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Virus Immunology Department, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Omu Anzala
- Department of Medical Microbiology & Immunology, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| | - Marianne Mureithi
- Department of Medical Microbiology & Immunology, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya
- KAVI-Institute of Clinical Research (KAVI-ICR), University of Nairobi, Nairobi, Kenya
| |
Collapse
|
4
|
Metabolism-dependent ferroptosis promotes mitochondrial dysfunction and inflammation in CD4 + T lymphocytes in HIV-infected immune non-responders. EBioMedicine 2022; 86:104382. [PMID: 36462403 PMCID: PMC9718960 DOI: 10.1016/j.ebiom.2022.104382] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/29/2022] [Accepted: 11/09/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND HIV immune non-responders (INRs) are described as a failure to reestablish a pool of CD4+ T lymphocytes (CD4 cells) after antiretroviral therapy (ART), which is related to poor clinical results. Ferroptosis is a newly discovered form of cell death characterised by iron-dependent lipid peroxidation and the accumulation of reactive oxygen species (ROS). The mechanism of unrecoverable CD4 cells in INRs and whether ferroptosis plays a role are not fully understood. METHODS Ninety-two people living with HIV (PLHIVs) who experienced four-year ART with sustained viral suppression, including 27 INRs, 34 partial responders (PRs), and 31 complete responders (CRs); and 26 uninfected control participants (UCs) were analysed for 16 immune parameters with flow cytometry. Then plasma lipid, iron and oxidation, and antioxidant indicators were detected by ELISA, and CD4 cells were sorted out and visualised under transmission electron microscopy. Finally, ferroptosis inhibitors were added, and alterations in CD4 cell phenotype and function were observed. FINDINGS We found decreased recent thymic emigrants (RTE), over-activation and over-proliferation phenotypes, diminished killing function, decreased IL-7R and more severe inflammation; increased lipid peroxidation in the mitochondria and disruptions of the mitochondrial structure, showing typical features of ferroptosis in CD4 cells in INRs. Additionally, ferroptosis inhibitors could reduce inflammation and repair mitochondrial damage. Meanwhile, ELISA results showed increased plasma free fatty acids (FFA) and an imbalance of oxidative and antioxidant systems in INRs. Flow cytometry results displayed alterations of both transferrin receptor (CD71) and lipid transporter (CD36) expressions on the surface of CD4 cells. Mechanistically, there was a stronger correlation between CD36 expression and mitochondrial lipid peroxidation production, ferroptosis makers, and inflammation indicators; while amino acid transporter (CD98) was more related to killing functions; and CD71 was more closely related to activation status in CD4 cells. INTERPRETATION Cellular metabolism was closely correlated with its diverse functions in INRs. Ferroptosis was observed in CD4 cells of INRs, and inhibiting ferroptosis through modulating mitochondrial disorders and inflammation may offer an alternative immunological strategy for reinvigorating CD4 cells in INRs. FUNDING This research was supported by the 13th Five-year Plan, Ministry of Science and Technology of China (2018ZX10302-102), Beijing Municipal Administration of Hospitals' Ascent Plan (DFL20191802), and Beijing Municipal Administration of Hospitals Clinical Medicine Development of Special Funding Support (ZYLX202126).
Collapse
|
5
|
Mayberry CL, Logan NA, Wilson JJ, Chang CH. Providing a Helping Hand: Metabolic Regulation of T Follicular Helper Cells and Their Association With Disease. Front Immunol 2022; 13:864949. [PMID: 35493515 PMCID: PMC9047778 DOI: 10.3389/fimmu.2022.864949] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/25/2022] [Indexed: 01/02/2023] Open
Abstract
T follicular helper (Tfh) cells provide support to B cells upon arrival in the germinal center, and thus are critical for the generation of a robust adaptive immune response. Tfh express specific transcription factors and cellular receptors including Bcl6, CXCR5, PD-1, and ICOS, which are critical for homing and overall function. Generally, the induction of an immune response is tightly regulated. However, deviation during this process can result in harmful autoimmunity or the inability to successfully clear pathogens. Recently, it has been shown that Tfh differentiation, activation, and proliferation may be linked with the cellular metabolic state. In this review we will highlight recent discoveries in Tfh differentiation and explore how these cells contribute to functional immunity in disease, including autoimmune-related disorders, cancer, and of particular emphasis, during infection.
Collapse
Affiliation(s)
| | | | | | - Chih-Hao Chang
- The Jackson Laboratory, Bar Harbor, ME, United States
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- *Correspondence: Chih-Hao Chang,
| |
Collapse
|
6
|
Schank M, Zhao J, Moorman JP, Yao ZQ. The Impact of HIV- and ART-Induced Mitochondrial Dysfunction in Cellular Senescence and Aging. Cells 2021; 10:cells10010174. [PMID: 33467074 PMCID: PMC7830696 DOI: 10.3390/cells10010174] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023] Open
Abstract
According to the WHO, 38 million individuals were living with human immunodeficiency virus (HIV), 25.4 million of which were using antiretroviral therapy (ART) at the end of 2019. Despite ART-mediated suppression of viral replication, ART is not a cure and is associated with viral persistence, residual inflammation, and metabolic disturbances. Indeed, due to the presence of viral reservoirs, lifelong ART therapy is required to control viremia and prevent disease progression into acquired immune deficiency syndrome (AIDS). Successful ART treatment allows people living with HIV (PLHIV) to achieve a similar life expectancy to uninfected individuals. However, recent studies have illustrated the presence of increased comorbidities, such as accelerated, premature immune aging, in ART-controlled PLHIV compared to uninfected individuals. Studies suggest that both HIV-infection and ART-treatment lead to mitochondrial dysfunction, ultimately resulting in cellular exhaustion, senescence, and apoptosis. Since mitochondria are essential cellular organelles for energy homeostasis and cellular metabolism, their compromise leads to decreased oxidative phosphorylation (OXPHOS), ATP synthesis, gluconeogenesis, and beta-oxidation, abnormal cell homeostasis, increased oxidative stress, depolarization of the mitochondrial membrane potential, and upregulation of mitochondrial DNA mutations and cellular apoptosis. The progressive mitochondrial damage induced by HIV-infection and ART-treatment likely contributes to accelerated aging, senescence, and cellular dysfunction in PLHIV. This review discusses the connections between mitochondrial compromise and cellular dysfunction associated with HIV- and ART-induced toxicities, providing new insights into how HIV and current ART directly impact mitochondrial functions and contribute to cellular senescence and aging in PLHIV. Identifying this nexus and potential mechanisms may be beneficial in developing improved therapeutics for treating PLHIV.
Collapse
Affiliation(s)
- Madison Schank
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (M.S.); (J.Z.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Juan Zhao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (M.S.); (J.Z.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Jonathan P. Moorman
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (M.S.); (J.Z.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN 37614, USA
| | - Zhi Q. Yao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (M.S.); (J.Z.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN 37614, USA
- Correspondence: ; Tel.: +423-439-8063; Fax: +423-439-7010
| |
Collapse
|
7
|
Deguit CDT, Hough M, Hoh R, Krone M, Pilcher CD, Martin JN, Deeks SG, McCune JM, Hunt PW, Rutishauser RL. Some Aspects of CD8+ T-Cell Exhaustion Are Associated With Altered T-Cell Mitochondrial Features and ROS Content in HIV Infection. J Acquir Immune Defic Syndr 2019; 82:211-219. [PMID: 31513075 PMCID: PMC6746248 DOI: 10.1097/qai.0000000000002121] [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: 01/14/2023]
Abstract
BACKGROUND Reversing or preventing T-cell exhaustion is an important treatment goal in the context of HIV disease; however, the mechanisms that regulate HIV-specific CD8 T-cell exhaustion are incompletely understood. Since mitochondrial mass (MM), mitochondrial membrane potential (MMP), and cellular reactive oxygen species (ROS) content are altered in exhausted CD8 T cells in other settings, we hypothesized that similar lesions may arise in HIV infection. METHODS We sampled cryopreserved peripheral blood mononuclear cells from HIV-uninfected (n = 10) and HIV-infected participants with varying levels and mechanisms of viral control: viremic (VL > 2000 copies/mL; n = 8) or aviremic (VL < 40 copies/mL) due to antiretroviral therapy (n = 11) or natural control (n = 9). We characterized the MM, MMP, and ROS content of bulk CD8 T cells and MHC class I tetramer+ HIV-specific CD8 T cells by flow cytometry. RESULTS We observed higher MM, MMP, and ROS content across bulk effector-memory CD8 T-cell subsets in HIV-infected compared with HIV-uninfected participants. Among HIV-specific CD8 T cells, these features did not vary by the extent or mechanism of viral control but were significantly altered in cells displaying characteristics associated with exhaustion (eg, high PD-1 expression, low CD127 expression, and impaired proliferative capacity). CONCLUSIONS While we did not find that control of HIV replication in vivo correlates with the CD8 T-cell MM, MMP, or ROS content, we did find that some features of CD8 T-cell exhaustion are associated with alterations in mitochondrial state. Our findings support further studies to probe the relationship between mitochondrial dynamics and CD8 T-cell functionality in HIV infection.
Collapse
Affiliation(s)
- Christian Deo T. Deguit
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
- Current Address: Department of Biochemistry and Molecular Biology, University of the Philippines, Manila, Philippines
| | - Michelle Hough
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
- Current Address: Department of Medicine, University of Southern California, Los Angeles, CA, U.S.A
| | - Rebecca Hoh
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
| | - Melissa Krone
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, U.S.A
| | - Christopher D. Pilcher
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, U.S.A
| | - Steven G. Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
| | - Joseph M. McCune
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
- Current Address: Bill & Melinda Gates Foundation, Seattle, WA, U.S.A
| | - Peter W. Hunt
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
| | - Rachel L. Rutishauser
- Department of Medicine, University of California, San Francisco, San Francisco, CA, U.S.A
| |
Collapse
|
8
|
Angin M, Volant S, Passaes C, Lecuroux C, Monceaux V, Dillies MA, Valle-Casuso JC, Pancino G, Vaslin B, Le Grand R, Weiss L, Goujard C, Meyer L, Boufassa F, Müller-Trutwin M, Lambotte O, Sáez-Cirión A. Metabolic plasticity of HIV-specific CD8 + T cells is associated with enhanced antiviral potential and natural control of HIV-1 infection. Nat Metab 2019; 1:704-716. [PMID: 32694646 DOI: 10.1038/s42255-019-0081-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 06/05/2019] [Indexed: 12/27/2022]
Abstract
Spontaneous control of human immunodeficiency virus (HIV) is generally associated with an enhanced capacity of CD8+ T cells to eliminate infected CD4+ T cells, but the molecular characteristics of these highly functional CD8+ T cells are largely unknown. In the present study, using single-cell analysis, it was shown that HIV-specific, central memory CD8+ T cells from spontaneous HIV controllers (HICs) and antiretrovirally treated non-controllers have opposing transcriptomic profiles. Genes linked to effector functions and survival are upregulated in cells from HICs. In contrast, genes associated with activation, exhaustion and glycolysis are upregulated in cells from non-controllers. It was shown that HIV-specific CD8+ T cells from non-controllers are largely glucose dependent, whereas those from HICs have more diverse metabolic resources that enhance both their survival potential and their capacity to develop anti-HIV effector functions. The functional efficiency of the HIV-specific CD8+ T cell response in HICs is thus engraved in their memory population and related to their metabolic programme. Metabolic reprogramming in vitro through interleukin-15 treatment abrogated the glucose dependency and enhanced the antiviral potency of HIV-specific CD8+ T cells from non-controllers.
Collapse
Affiliation(s)
- Mathieu Angin
- Institut Pasteur, Unité HIV Inflammation et Persistance, Paris, France
| | - Stevenn Volant
- Institut Pasteur, Hub Bioinformatique et Biostatistique, Paris, France
| | - Caroline Passaes
- Institut Pasteur, Unité HIV Inflammation et Persistance, Paris, France
| | - Camille Lecuroux
- CEA, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department/IBFJ, Université Paris Sud, Fontenay-aux-Roses, France
| | - Valérie Monceaux
- Institut Pasteur, Unité HIV Inflammation et Persistance, Paris, France
| | | | | | | | - Bruno Vaslin
- CEA, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department/IBFJ, Université Paris Sud, Fontenay-aux-Roses, France
| | - Roger Le Grand
- CEA, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department/IBFJ, Université Paris Sud, Fontenay-aux-Roses, France
| | - Laurence Weiss
- Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Cecile Goujard
- Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Service de Médecine Interne et Immunologie Clinique, Le Kremlin-Bicêtre, France
| | - Laurence Meyer
- Centre de Recherche en Epidémiologie et Santé des Populations, Université Paris Sud, Le Kremlin-Bicêtre, France
| | - Faroudy Boufassa
- Centre de Recherche en Epidémiologie et Santé des Populations, Université Paris Sud, Le Kremlin-Bicêtre, France
| | | | - Olivier Lambotte
- CEA, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department/IBFJ, Université Paris Sud, Fontenay-aux-Roses, France
- Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Service de Médecine Interne et Immunologie Clinique, Le Kremlin-Bicêtre, France
| | - Asier Sáez-Cirión
- Institut Pasteur, Unité HIV Inflammation et Persistance, Paris, France.
| |
Collapse
|
9
|
Palmer CS, Henstridge DC, Yu D, Singh A, Balderson B, Duette G, Cherry CL, Anzinger JJ, Ostrowski M, Crowe SM. Emerging Role and Characterization of Immunometabolism: Relevance to HIV Pathogenesis, Serious Non-AIDS Events, and a Cure. THE JOURNAL OF IMMUNOLOGY 2017; 196:4437-44. [PMID: 27207806 DOI: 10.4049/jimmunol.1600120] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/20/2016] [Indexed: 01/18/2023]
Abstract
Immune cells cycle between a resting and an activated state. Their metabolism is tightly linked to their activation status and, consequently, functions. Ag recognition induces T lymphocyte activation and proliferation and acquisition of effector functions that require and depend on cellular metabolic reprogramming. Likewise, recognition of pathogen-associated molecular patterns by monocytes and macrophages induces changes in cellular metabolism. As obligate intracellular parasites, viruses manipulate the metabolism of infected cells to meet their structural and functional requirements. For example, HIV-induced changes in immune cell metabolism and redox state are associated with CD4(+) T cell depletion, immune activation, and inflammation. In this review, we highlight how HIV modifies immunometabolism with potential implications for cure research and pathogenesis of comorbidities observed in HIV-infected patients, including those with virologic suppression. In addition, we highlight recently described key methods that can be applied to study the metabolic dysregulation of immune cells in disease states.
Collapse
Affiliation(s)
- Clovis S Palmer
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia; Department of Infectious Diseases, Monash University, Melbourne, Victoria 3004, Australia;
| | - Darren C Henstridge
- Cellular and Molecular Metabolism Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Di Yu
- Laboratory of Molecular Immunomodulation, School of Biomedical Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Amit Singh
- Department of Microbiology and Cell Biology, Centre for Infectious Disease and Research, Indian Institute of Science, Bangalore 560012, India
| | - Brad Balderson
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia
| | - Gabriel Duette
- Instituto de Investigaciones Biomedicas en Retrovirus y SIDA, Facultad de Medicina, C1121ABG Buenos Aires, Argentina
| | - Catherine L Cherry
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia; Department of Infectious Diseases, Monash University, Melbourne, Victoria 3004, Australia; Infectious Diseases Department, The Alfred Hospital, Melbourne, Victoria 3004, Australia; School of Physiology, University of the Witwatersrand, Johannesburg, Gauteng 2000, South Africa; and
| | - Joshua J Anzinger
- Department of Microbiology, University of the West Indies, Mona, Jamaica
| | - Matias Ostrowski
- Instituto de Investigaciones Biomedicas en Retrovirus y SIDA, Facultad de Medicina, C1121ABG Buenos Aires, Argentina
| | - Suzanne M Crowe
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia; Department of Infectious Diseases, Monash University, Melbourne, Victoria 3004, Australia; Infectious Diseases Department, The Alfred Hospital, Melbourne, Victoria 3004, Australia
| |
Collapse
|
10
|
Yeoh HL, Cheng AC, Cherry CL, Weir JM, Meikle PJ, Hoy JF, Crowe SM, Palmer CS. Immunometabolic and Lipidomic Markers Associated With the Frailty Index and Quality of Life in Aging HIV+ Men on Antiretroviral Therapy. EBioMedicine 2017; 22:112-121. [PMID: 28754302 PMCID: PMC5552224 DOI: 10.1016/j.ebiom.2017.07.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/15/2017] [Accepted: 07/17/2017] [Indexed: 01/09/2023] Open
Abstract
Chronic immune activation persists despite antiretroviral therapy (ART) in HIV+ individuals and underpins an increased risk of age-related co-morbidities. We assessed the Frailty Index in older HIV+ Australian men on ART. Immunometabolic markers on monocytes and T cells were analyzed using flow cytometry, plasma innate immune activation markers by ELISA, and lipidomic profiling by mass spectrometry. The study population consisted of 80 HIV+ men with a median age of 59 (IQR, 56-65), and most had an undetectable viral load (92%). 24% were frail, and 76% were non-frail. Frailty was associated with elevated Glucose transporter-1 (Glut1) expression on the total monocytes (p=0.04), increased plasma levels of innate immune activation marker sCD163 (OR, 4.8; CI 1.4-15.9, p=0.01), phosphatidylethanolamine PE(36:3) (OR, 5.1; CI 1.7-15.5, p=0.004) and triacylglycerol TG(16:1_18:1_18:1) (OR, 3.4; CI 1.3-9.2, p=0.02), but decreased expression of GM3 ganglioside, GM3(d18:1/18:0) (OR, 0.1; CI 0.0-0.6, p=0.01) and monohexosylceramide HexCerd(d18:1/22:0) (OR, 0.1; CI 0.0-0.5, p=0.004). There is a strong inverse correlation between quality of life and the concentration of PE(36:3) (ρ=-0.33, p=0.004) and PE(36:4) (ρ=-0.37, p=0.001). These data suggest that frailty is associated with increased innate immune activation and abnormal lipidomic profile. These markers should be investigated in larger, longitudinal studies to determine their potential as biomarkers for frailty.
Collapse
Affiliation(s)
- Hui-Ling Yeoh
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Allen C Cheng
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Department of Epidemiology and Preventive Medicine, Monash University, Level 6, The Alfred Centre (Alfred Hospital), 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Catherine L Cherry
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; School of Physiology, University of the Witwatersrand, 1 Jan Smuts Avenue, Braamfontein, 2000 Johannesburg, South Africa
| | - Jacquelyn M Weir
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Jennifer F Hoy
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Suzanne M Crowe
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Clovis S Palmer
- Department of Infectious Diseases, The Alfred and Monash University, Level 2, Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia; Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
11
|
Piggott DA, Erlandson KM, Yarasheski KE. Frailty in HIV: Epidemiology, Biology, Measurement, Interventions, and Research Needs. Curr HIV/AIDS Rep 2016; 13:340-348. [PMID: 27549318 PMCID: PMC5131367 DOI: 10.1007/s11904-016-0334-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Frailty is a critical aging-related syndrome marked by diminished physiologic reserve and heightened vulnerability to stressors, predisposing to major adverse clinical outcomes, including hospitalization, institutionalization, disability, and death in the general population of older adults. As the proportion of older adults living with HIV increases in the era of antiretroviral therapy, frailty is increasingly recognized to be of significant clinical and public health relevance to the HIV-infected population. This article reviews current knowledge on the epidemiology and biology of frailty and its potential role as a target for reducing disparities in outcomes in HIV; conceptual frameworks and current approaches to frailty measurement; existing data on frailty interventions; and important areas for future research focus necessary to develop and advance effective strategies to prevent or ameliorate frailty and its marked adverse consequences among people living with HIV.
Collapse
Affiliation(s)
- Damani A Piggott
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, MD, USA
| | - Kristine M Erlandson
- Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Kevin E Yarasheski
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8127, St. Louis, MO, 63110, USA.
| |
Collapse
|
12
|
Protein carbonyl content: a novel biomarker for aging in HIV/AIDS patients. Braz J Infect Dis 2016; 21:35-41. [PMID: 27821249 PMCID: PMC9425472 DOI: 10.1016/j.bjid.2016.09.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/24/2016] [Accepted: 09/21/2016] [Indexed: 12/26/2022] Open
Abstract
Background The major complications of “treated” Human Immunodeficiency Virus (HIV) infection are cardiovascular disease, malignancy, renal disease, liver disease, bone disease, and perhaps neurological complications, which are phenomena of the normal aging process occurring at an earlier age in the HIV-infected population. The present study is aimed to explore protein carbonyl content as a biomarker for detecting oxidative DNA damage induced ART toxicity and/or accelerated aging in HIV/AIDS patients. Objective To investigate the potential of carbonyl content as a biomarker for detecting oxidative Deoxyribonucleic acid (DNA) damage induced Antiretroviral Theraphy (ART) toxicity and/or accelerated aging in HIV/AIDS patients. Methods In this case–control study a total 600 subjects were included. All subjects were randomly selected and grouped as HIV-negative (control group) (n = 300), HIV-infected ART naive (n = 100), HIV-infected on first line ART (n = 100), and HIV-infected on second line ART (n = 100). Seronegative control subjects were age- and sex-matched with the ART naive patients and the two other groups. Carbonyl protein was determined by the method described in Levine et al. DNA damage marker 8-OH-dG was determined using 8-hydroxy-2-deoxy Guanosine StressXpress ELA Kit by StressMarq Biosciences. Results Protein carbonyl content levels and oxidative DNA damage were significantly higher (p < 0.05) in HIV-infected patients on second line ART and HIV-infected patients on first line ART than ART naive patients and controls. In a linear regression analysis, increased protein carbonyl content was positively associated with increased DNA damage (OR: 0.356; 95% CI: 0.287–0.426) p < 0.05. Conclusions Carbonyl content may has a role as a biomarker for detecting oxidative DNA damage induced ART toxicity and/or accelerated aging in HIV/AIDS patients. Larger studies are warranted to elucidate the role of carbonyl content as a biomarker for premature aging in HIV/AIDS patients.
Collapse
|
13
|
Saxena M, Busca A, Holcik M, Kumar A. Bacterial DNA Protects Monocytic Cells against HIV-Vpr-Induced Mitochondrial Membrane Depolarization. THE JOURNAL OF IMMUNOLOGY 2016; 196:3754-67. [PMID: 26969755 DOI: 10.4049/jimmunol.1402379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 02/19/2016] [Indexed: 12/26/2022]
Abstract
Monocytes and macrophages are important HIV reservoirs, as they exhibit marked resistance to apoptosis upon infection. However, the mechanism underlying resistance to apoptosis in these cells is poorly understood. Using HIV-viral protein R-52-96 aa peptide (Vpr), we show that primary monocytes and THP-1 cells treated with Vpr are highly susceptible to mitochondrial depolarization, but develop resistance following stimulation with bacterial DNA or CpG oligodeoxynucleotide. We have shown that Vpr-induced mitochondrial depolarization is mediated by TNFR-associated factor-1 (TRAF-1) and TRAF-2 degradation and subsequent activation of caspase-8, Bid, and Bax. To provide the mechanism governing such resistance to mitochondrial depolarization, our results show that prior stimulation with CpG oligodeoxynucleotide or Escherichia coli DNA prevented: 1) TRAF-1/2 downregulation; 2) activation of caspase-8, Bid, and Bax; and 3) subsequent mitochondrial depolarization and release of apoptosis-inducing factor and cytochrome c Furthermore, this protection was mediated by upregulation of antiapoptotic protein (c-IAP-2) through calmodulin-dependent kinase-II activation. Thus, c-IAP-2 may prevent Vpr-mediated mitochondrial depolarization through stabilizing TRAF-1/2 expression and sequential inhibition of caspase-8, Bid, and Bax.
Collapse
Affiliation(s)
- Mansi Saxena
- Department of Biochemistry, Microbiology and Immunology, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada
| | - Aurelia Busca
- Department of Biochemistry, Microbiology and Immunology, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada
| | - Martin Holcik
- Research Institute, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada; and
| | - Ashok Kumar
- Department of Biochemistry, Microbiology and Immunology, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada; Research Institute, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada; and Department of Pathology and Laboratory Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario K1H 8L1, Canada
| |
Collapse
|
14
|
Hydrogen Sulfide and Cellular Redox Homeostasis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6043038. [PMID: 26881033 PMCID: PMC4736422 DOI: 10.1155/2016/6043038] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 11/23/2015] [Accepted: 12/01/2015] [Indexed: 01/06/2023]
Abstract
Intracellular redox imbalance is mainly caused by overproduction of reactive oxygen species (ROS) or weakness of the natural antioxidant defense system. It is involved in the pathophysiology of a wide array of human diseases. Hydrogen sulfide (H2S) is now recognized as the third “gasotransmitters” and proved to exert a wide range of physiological and cytoprotective functions in the biological systems. Among these functions, the role of H2S in oxidative stress has been one of the main focuses over years. However, the underlying mechanisms for the antioxidant effect of H2S are still poorly comprehended. This review presents an overview of the current understanding of H2S specially focusing on the new understanding and mechanisms of the antioxidant effects of H2S based on recent reports. Both inhibition of ROS generation and stimulation of antioxidants are discussed. H2S-induced S-sulfhydration of key proteins (e.g., p66Shc and Keap1) is also one of the focuses of this review.
Collapse
|
15
|
Abstract
Bioenergetics has become central to our understanding of pathological mechanisms, the
development of new therapeutic strategies and as a biomarker for disease progression
in neurodegeneration, diabetes, cancer and cardiovascular disease. A key concept is
that the mitochondrion can act as the ‘canary in the coal mine’ by
serving as an early warning of bioenergetic crisis in patient populations. We propose
that new clinical tests to monitor changes in bioenergetics in patient populations
are needed to take advantage of the early and sensitive ability of bioenergetics to
determine severity and progression in complex and multifactorial diseases. With the
recent development of high-throughput assays to measure cellular energetic function
in the small number of cells that can be isolated from human blood these clinical
tests are now feasible. We have shown that the sequential addition of
well-characterized inhibitors of oxidative phosphorylation allows a bioenergetic
profile to be measured in cells isolated from normal or pathological samples. From
these data we propose that a single value–the Bioenergetic Health Index
(BHI)–can be calculated to represent the patient's composite mitochondrial
profile for a selected cell type. In the present Hypothesis paper, we discuss how BHI
could serve as a dynamic index of bioenergetic health and how it can be measured in
platelets and leucocytes. We propose that, ultimately, BHI has the potential to be a
new biomarker for assessing patient health with both prognostic and diagnostic
value.
Collapse
|
16
|
Perrin S, Cremer J, Faucher O, Reynes J, Dellamonica P, Micallef J, Solas C, Lacarelle B, Stretti C, Kaspi E, Robaglia-Schlupp A, Tamalet CNBC, Lévy N, Poizot-Martin I, Cau P, Roll P. HIV protease inhibitors do not cause the accumulation of prelamin A in PBMCs from patients receiving first line therapy: the ANRS EP45 "aging" study. PLoS One 2012; 7:e53035. [PMID: 23285253 PMCID: PMC3532351 DOI: 10.1371/journal.pone.0053035] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 11/22/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The ANRS EP45 "Aging" study investigates the cellular mechanisms involved in the accelerated aging of HIV-1 infected and treated patients. The present report focuses on lamin A processing, a pathway known to be altered in systemic genetic progeroid syndromes. METHODS 35 HIV-1 infected patients being treated with first line antiretroviral therapy (ART, mean duration at inclusion: 2.7±1.3 years) containing boosted protease inhibitors (PI/r) (comprising lopinavir/ritonavir in 65% of patients) were recruited together with 49 seronegative age- and sex-matched control subjects (http://clinicaltrials.gov/, NCT01038999). In more than 88% of patients, the viral load was <40 copies/ml and the CD4+ cell count was >500/mm³. Prelamin A processing in peripheral blood mononuclear cells (PBMCs) from patients and controls was analysed by western blotting at inclusion. PBMCs from patients were also investigated at 12 and 24 months after enrolment in the study. PBMCs from healthy controls were also incubated with boosted lopinavir in culture medium containing various concentrations of proteins (4 to 80 g/L). RESULTS Lamin A precursor was not observed in cohort patient PBMC regardless of the PI/r used, the dose and the plasma concentration. Prelamin A was detected in PBMC incubated in culture medium containing a low protein concentration (4 g/L) but not in plasma (60-80 g/L) or in medium supplemented with BSA (40 g/L), both of which contain a high protein concentration. CONCLUSIONS Prelamin A processing abnormalities were not observed in PBMCs from patients under the PI/r first line regimen. Therefore, PI/r do not appear to contribute to lamin A-related aging in PBMCs. In cultured PBMCs from healthy donors, prelamin A processing abnormalities were only observed when the protein concentration in the culture medium was low, thus increasing the amount of PI available to enter cells. ClinicalTrials.gov NCT01038999 http://clinicaltrials.gov/ct2/show/NCT01038999.
Collapse
Affiliation(s)
- Sophie Perrin
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Jonathan Cremer
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Olivia Faucher
- Service d’Immuno-Hématologie Clinique, Centre Hospitalier Universitaire (CHU) Sainte Marguerite Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Jacques Reynes
- Département des Maladies Infectieuses et Tropicales, Centre Hospitalier Régional et Universitaire (CHRU) Gui-de-Chauliac, Montpellier, France
| | - Pierre Dellamonica
- Service d’Infectiologie, Centre Hospitalier Universitaire (CHU) L’Archet 1, Sophia-Antipolis Université, Nice, France
| | - Joëlle Micallef
- Centre d’Investigation Clinique - Unité de Pharmacologie Clinique et d’Evaluations Thérapeutiques (CIC-UPCET), Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Caroline Solas
- Laboratoire de Pharmacocinétique et de Toxicologie, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
- Inserm UMR_S 911, Aix-Marseille Université, Marseille, France
| | - Bruno Lacarelle
- Laboratoire de Pharmacocinétique et de Toxicologie, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
- Inserm UMR_S 911, Aix-Marseille Université, Marseille, France
| | - Charlotte Stretti
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Elise Kaspi
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Andrée Robaglia-Schlupp
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | | | - Nicolas Lévy
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Génetique Moléculaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Isabelle Poizot-Martin
- Département des Maladies Infectieuses et Tropicales, Centre Hospitalier Régional et Universitaire (CHRU) Gui-de-Chauliac, Montpellier, France
| | - Pierre Cau
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Patrice Roll
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
- * E-mail:
| |
Collapse
|