1
|
Obare LM, Temu T, Mallal SA, Wanjalla CN. Inflammation in HIV and Its Impact on Atherosclerotic Cardiovascular Disease. Circ Res 2024; 134:1515-1545. [PMID: 38781301 PMCID: PMC11122788 DOI: 10.1161/circresaha.124.323891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8+ and CD4+ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| | - Tecla Temu
- Department of Pathology, Harvard Medical School, Boston, MA (T.T.)
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Institute for Immunology and Infectious Diseases, Murdoch University, WA, Western Australia (S.A.M.)
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| |
Collapse
|
2
|
Schenck JK, Karl MT, Clarkson-Paredes C, Bastin A, Pushkarsky T, Brichacek B, Miller RH, Bukrinsky MI. Extracellular vesicles produced by HIV-1 Nef-expressing cells induce myelin impairment and oligodendrocyte damage in the mouse central nervous system. J Neuroinflammation 2024; 21:127. [PMID: 38741181 PMCID: PMC11090814 DOI: 10.1186/s12974-024-03124-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/04/2024] [Indexed: 05/16/2024] Open
Abstract
HIV-associated neurocognitive disorders (HAND) are a spectrum of cognitive impairments that continue to affect approximately half of all HIV-positive individuals despite effective viral suppression through antiretroviral therapy (ART). White matter pathologies have persisted in the ART era, and the degree of white matter damage correlates with the degree of neurocognitive impairment in patients with HAND. The HIV protein Nef has been implicated in HAND pathogenesis, but its effect on white matter damage has not been well characterized. Here, utilizing in vivo, ex vivo, and in vitro methods, we demonstrate that Nef-containing extracellular vesicles (Nef EVs) disrupt myelin sheaths and inflict damage upon oligodendrocytes within the murine central nervous system. Intracranial injection of Nef EVs leads to reduced myelin basic protein (MBP) staining and a decreased number of CC1 + oligodendrocytes in the corpus callosum. Moreover, cerebellar slice cultures treated with Nef EVs exhibit diminished MBP expression and increased presence of unmyelinated axons. Primary mixed brain cultures and enriched oligodendrocyte precursor cell cultures exposed to Nef EVs display a decreased number of O4 + cells, indicative of oligodendrocyte impairment. These findings underscore the potential contribution of Nef EV-mediated damage to oligodendrocytes and myelin maintenance in the pathogenesis of HAND.
Collapse
Affiliation(s)
- Jessica K Schenck
- School of Medicine and Health Sciences, The George Washington University, 2300 I St NW, Ross Hall 624, Washington, DC, 20037, USA
| | - Molly T Karl
- School of Medicine and Health Sciences, The George Washington University, 2300 I St NW, Ross Hall 624, Washington, DC, 20037, USA
| | - Cheryl Clarkson-Paredes
- School of Medicine and Health Sciences, The George Washington University, 2300 I St NW, Ross Hall 624, Washington, DC, 20037, USA
| | - Ashley Bastin
- School of Medicine and Health Sciences, The George Washington University, 2300 I St NW, Ross Hall 624, Washington, DC, 20037, USA
| | - Tatiana Pushkarsky
- School of Medicine and Health Sciences, The George Washington University, 2300 I St NW, Ross Hall 624, Washington, DC, 20037, USA
| | - Beda Brichacek
- School of Medicine and Health Sciences, The George Washington University, 2300 I St NW, Ross Hall 624, Washington, DC, 20037, USA
| | - Robert H Miller
- School of Medicine and Health Sciences, The George Washington University, 2300 I St NW, Ross Hall 624, Washington, DC, 20037, USA
| | - Michael I Bukrinsky
- School of Medicine and Health Sciences, The George Washington University, 2300 I St NW, Ross Hall 624, Washington, DC, 20037, USA.
| |
Collapse
|
3
|
Sviridov D, Bukrinsky M. Neuro-HIV-New insights into pathogenesis and emerging therapeutic targets. FASEB J 2023; 37:e23301. [PMID: 37942865 PMCID: PMC11032165 DOI: 10.1096/fj.202301239rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023]
Abstract
HIV-associated neurocognitive disorders (HAND) is a term describing a complex set of cognitive impairments accompanying HIV infection. Successful antiretroviral therapy (ART) reduces the most severe forms of HAND, but milder forms affect over 50% of people living with HIV (PLWH). Pathogenesis of HAND in the ART era remains unknown. A variety of pathogenic factors, such as persistent HIV replication in the brain reservoir, HIV proteins released from infected brain cells, HIV-induced neuroinflammation, and some components of ART, have been implicated in driving HAND pathogenesis in ART-treated individuals. Here, we propose another factor-impairment of cholesterol homeostasis and lipid rafts by HIV-1 protein Nef-as a possible contributor to HAND pathogenesis. These effects of Nef on cholesterol may also underlie the effects of other pathogenic factors that constitute the multifactorial nature of HAND pathogenesis. The proposed Nef- and cholesterol-focused mechanism may provide a long-sought unified explanation of HAND pathogenesis that takes into account all contributing factors. Evidence for the impairment by Nef of cellular cholesterol balance, potential effects of this impairment on brain cells, and opportunities to therapeutically target this element of HAND pathogenesis are discussed.
Collapse
Affiliation(s)
- Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Michael Bukrinsky
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
4
|
Proulx J, Ghaly M, Park IW, Borgmann K. HIV-1-Mediated Acceleration of Oncovirus-Related Non-AIDS-Defining Cancers. Biomedicines 2022; 10:biomedicines10040768. [PMID: 35453518 PMCID: PMC9024568 DOI: 10.3390/biomedicines10040768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 11/25/2022] Open
Abstract
With the advent of combination antiretroviral therapy (cART), overall survival has been improved, and the incidence of acquired immunodeficiency syndrome (AIDS)-defining cancers has also been remarkably reduced. However, non-AIDS-defining cancers among human immunodeficiency virus-1 (HIV-1)-associated malignancies have increased significantly so that cancer is the leading cause of death in people living with HIV in certain highly developed countries, such as France. However, it is currently unknown how HIV-1 infection raises oncogenic virus-mediated cancer risks in the HIV-1 and oncogenic virus co-infected patients, and thus elucidation of the molecular mechanisms for how HIV-1 expedites the oncogenic viruses-triggered tumorigenesis in the co-infected hosts is imperative for developing therapeutics to cure or impede the carcinogenesis. Hence, this review is focused on HIV-1 and oncogenic virus co-infection-mediated molecular processes in the acceleration of non-AIDS-defining cancers.
Collapse
|
5
|
Clauss M, Chelvanambi S, Cook C, ElMergawy R, Dhillon N. Viral Bad News Sent by EVAIL. Viruses 2021; 13:v13061168. [PMID: 34207152 PMCID: PMC8234235 DOI: 10.3390/v13061168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 01/08/2023] Open
Abstract
This article reviews the current knowledge on how viruses may utilize Extracellular Vesicle Assisted Inflammatory Load (EVAIL) to exert pathologic activities. Viruses are classically considered to exert their pathologic actions through acute or chronic infection followed by the host response. This host response causes the release of cytokines leading to vascular endothelial cell dysfunction and cardiovascular complications. However, viruses may employ an alternative pathway to soluble cytokine-induced pathologies-by initiating the release of extracellular vesicles (EVs), including exosomes. The best-understood example of this alternative pathway is human immunodeficiency virus (HIV)-elicited EVs and their propensity to harm vascular endothelial cells. Specifically, an HIV-encoded accessory protein called the "negative factor" (Nef) was demonstrated in EVs from the body fluids of HIV patients on successful combined antiretroviral therapy (ART); it was also demonstrated to be sufficient in inducing endothelial and cardiovascular dysfunction. This review will highlight HIV-Nef as an example of how HIV can produce EVs loaded with proinflammatory cargo to disseminate cardiovascular pathologies. It will further discuss whether EV production can explain SARS-CoV-2-mediated pulmonary and cardiovascular pathologies.
Collapse
Affiliation(s)
- Matthias Clauss
- IU School of Medicine, Pulmonary, Critical Care, Sleep and Occupational Medicine, Indianapolis, IN 46202, USA;
- Correspondence: or
| | - Sarvesh Chelvanambi
- Brigham and Women’s Hospital, Department of Medicine, Boston, MA 02115, USA;
| | - Christine Cook
- Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.C.); (N.D.)
| | - Rabab ElMergawy
- IU School of Medicine, Pulmonary, Critical Care, Sleep and Occupational Medicine, Indianapolis, IN 46202, USA;
| | - Navneet Dhillon
- Pulmonary and Critical Care Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.C.); (N.D.)
| |
Collapse
|
6
|
Jacobo-Albavera L, Domínguez-Pérez M, Medina-Leyte DJ, González-Garrido A, Villarreal-Molina T. The Role of the ATP-Binding Cassette A1 (ABCA1) in Human Disease. Int J Mol Sci 2021; 22:ijms22041593. [PMID: 33562440 PMCID: PMC7915494 DOI: 10.3390/ijms22041593] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
Cholesterol homeostasis is essential in normal physiology of all cells. One of several proteins involved in cholesterol homeostasis is the ATP-binding cassette transporter A1 (ABCA1), a transmembrane protein widely expressed in many tissues. One of its main functions is the efflux of intracellular free cholesterol and phospholipids across the plasma membrane to combine with apolipoproteins, mainly apolipoprotein A-I (Apo A-I), forming nascent high-density lipoprotein-cholesterol (HDL-C) particles, the first step of reverse cholesterol transport (RCT). In addition, ABCA1 regulates cholesterol and phospholipid content in the plasma membrane affecting lipid rafts, microparticle (MP) formation and cell signaling. Thus, it is not surprising that impaired ABCA1 function and altered cholesterol homeostasis may affect many different organs and is involved in the pathophysiology of a broad array of diseases. This review describes evidence obtained from animal models, human studies and genetic variation explaining how ABCA1 is involved in dyslipidemia, coronary heart disease (CHD), type 2 diabetes (T2D), thrombosis, neurological disorders, age-related macular degeneration (AMD), glaucoma, viral infections and in cancer progression.
Collapse
Affiliation(s)
- Leonor Jacobo-Albavera
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Dirección de Investigación, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City CP14610, Mexico; (L.J.-A.); (M.D.-P.); (D.J.M.-L.); (A.G.-G.)
| | - Mayra Domínguez-Pérez
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Dirección de Investigación, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City CP14610, Mexico; (L.J.-A.); (M.D.-P.); (D.J.M.-L.); (A.G.-G.)
| | - Diana Jhoseline Medina-Leyte
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Dirección de Investigación, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City CP14610, Mexico; (L.J.-A.); (M.D.-P.); (D.J.M.-L.); (A.G.-G.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Mexico City CP04510, Mexico
| | - Antonia González-Garrido
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Dirección de Investigación, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City CP14610, Mexico; (L.J.-A.); (M.D.-P.); (D.J.M.-L.); (A.G.-G.)
| | - Teresa Villarreal-Molina
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Dirección de Investigación, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City CP14610, Mexico; (L.J.-A.); (M.D.-P.); (D.J.M.-L.); (A.G.-G.)
- Correspondence:
| |
Collapse
|
7
|
Bowman E, Funderburg NT. Lipidome Abnormalities and Cardiovascular Disease Risk in HIV Infection. Curr HIV/AIDS Rep 2020; 16:214-223. [PMID: 30993515 DOI: 10.1007/s11904-019-00442-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Human immunodeficiency virus (HIV) infection and its treatment with antiretroviral therapy (ART) are associated with lipid abnormalities that may enhance cardiovascular disease risk (CVD). RECENT FINDINGS Chronic inflammation persists in HIV+ individuals, and complex relationships exist among lipids and inflammation, as immune activation may be both a cause and a consequence of lipid abnormalities in HIV infection. Advances in mass spectrometry-based techniques now allow for detailed measurements of individual lipid species; improved lipid measurement might better evaluate CVD risk compared with the prognostic value of traditional assessments. Lipidomic analyses have begun to characterize dynamic changes in lipid composition during HIV infection and following treatment with ART, and further investigation may identify novel lipid biomarkers predictive of adverse outcomes. Developing strategies to improve management of comorbidities in the HIV+ population is important, and statin therapy and lifestyle modifications, including diet and exercise, may help to improve lipid levels and mitigate CVD risk.
Collapse
Affiliation(s)
- Emily Bowman
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University College of Medicine, 453 W. 10th Ave. 535A Atwell Hall, Columbus, OH, 43210, USA
| | - Nicholas T Funderburg
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Ohio State University College of Medicine, 453 W. 10th Ave. 535A Atwell Hall, Columbus, OH, 43210, USA.
| |
Collapse
|
8
|
Abstract
Apolipoprotein A-I binding protein (AIBP) is a recently identified innate anti-inflammatory factor. Here, we show that AIBP inhibited HIV replication by targeting lipid rafts and reducing virus-cell fusion. Importantly, AIBP selectively reduced levels of rafts on cells stimulated by an inflammatory stimulus or treated with extracellular vesicles containing HIV-1 protein Nef without affecting rafts on nonactivated cells. Accordingly, fusion of monocyte-derived macrophages with HIV was sensitive to AIBP only in the presence of Nef. Silencing of endogenous AIBP significantly upregulated HIV-1 replication. Interestingly, HIV-1 replication in cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, was not inhibited by AIBP. These results suggest that AIBP is an innate anti-HIV factor that targets virus-cell fusion. Apolipoprotein A-I binding protein (AIBP) is a protein involved in regulation of lipid rafts and cholesterol efflux. AIBP has been suggested to function as a protective factor under several sets of pathological conditions associated with increased abundance of lipid rafts, such as atherosclerosis and acute lung injury. Here, we show that exogenously added AIBP reduced the abundance of lipid rafts and inhibited HIV replication in vitro as well as in HIV-infected humanized mice, whereas knockdown of endogenous AIBP increased HIV replication. Endogenous AIBP was much more abundant in activated T cells than in monocyte-derived macrophages (MDMs), and exogenous AIBP was much less effective in T cells than in MDMs. AIBP inhibited virus-cell fusion, specifically targeting cells with lipid rafts mobilized by cell activation or Nef-containing exosomes. MDM-HIV fusion was sensitive to AIBP only in the presence of Nef provided by the virus or exosomes. Peripheral blood mononuclear cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, bound less AIBP than cells from donors with other HLA genotypes and were not protected by AIBP from rapid HIV-1 replication. These results provide the first evidence for the role of Nef exosomes in regulating HIV-cell fusion by modifying lipid rafts and suggest that AIBP is an innate factor that restricts HIV replication by targeting lipid rafts.
Collapse
|
9
|
Comorbidities of HIV infection: role of Nef-induced impairment of cholesterol metabolism and lipid raft functionality. AIDS 2020; 34:1-13. [PMID: 31789888 PMCID: PMC6903377 DOI: 10.1097/qad.0000000000002385] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Combination antiretroviral therapy has dramatically changed the outcome of HIV infection, turning it from a death sentence to a manageable chronic disease. However, comorbidities accompanying HIV infection, such as metabolic and cardio-vascular diseases, as well as cognitive impairment, persist despite successful virus control by combination antiretroviral therapy and pose considerable challenges to clinical management of people living with HIV. These comorbidities involve a number of pathological processes affecting a variety of different tissues and cells, making it challenging to identify a common cause(s) that would link these different diseases to HIV infection. In this article, we will present evidence that impairment of cellular cholesterol metabolism may be a common factor driving pathogenesis of HIV-associated comorbidities. Potential implications for therapeutic approaches are discussed.
Collapse
|
10
|
Sopeyin A, Zhou L, Li M, Barakat L, Paintsil E. Dysregulation of sterol regulatory element-binding protein 2 gene in HIV treatment-experienced individuals. PLoS One 2019; 14:e0226573. [PMID: 31846498 PMCID: PMC6917281 DOI: 10.1371/journal.pone.0226573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/28/2019] [Indexed: 12/11/2022] Open
Abstract
Although antiretroviral therapy (ART) has resulted in a marked decrease in AIDS-related morbidity and mortality, the therapeutic benefit is often limited by side effects such as metabolic derangement such as lipodystrophy and hyperlipidemia and cardiovascular diseases. These side effects are pervasive in people living with HIV (PLWH). However, the underlying mechanisms are not completely understood. We investigated the effects of ART on cholesterol biosynthesis genes. This is a retrospective analysis of data and specimens collected during a cross-sectional, case-control study of ART-induced toxicity. Cases were HIV treatment-experienced individuals with HIV viral suppression and no diagnosis of ART-associated toxicity (n = 18), and controls were HIV-uninfected individuals (n = 18). The mRNA expressions of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) and ATP binding cassette transporter A1 (ABCA1) were significantly upregulated in cases (HIV+) compared to controls (HIV-), as well as the corresponding protein expression level of HMGCR. We observed dysregulation between sterol regulatory element-binding protein 2 (SREBP-2, sensory control) and HMGCR and low-density lipoprotein receptor (LDLR) pathways. Dysregulation of cholesterol biosynthesis genes may predate clinical manifestation of ART-induced lipid abnormalities.
Collapse
Affiliation(s)
- Anuoluwapo Sopeyin
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Lei Zhou
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Min Li
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Lydia Barakat
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Elijah Paintsil
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- School of Public Health, Yale University, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
11
|
Exosomes containing HIV protein Nef reorganize lipid rafts potentiating inflammatory response in bystander cells. PLoS Pathog 2019; 15:e1007907. [PMID: 31344124 PMCID: PMC6657916 DOI: 10.1371/journal.ppat.1007907] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/11/2019] [Indexed: 01/21/2023] Open
Abstract
HIV infection has a profound effect on “bystander” cells causing metabolic co-morbidities. This may be mediated by exosomes secreted by HIV-infected cells and containing viral factors. Here we show that exosomes containing HIV-1 protein Nef (exNef) are rapidly taken up by macrophages releasing Nef into the cell interior. This caused down-regulation of ABCA1, reduction of cholesterol efflux and sharp elevation of the abundance of lipid rafts through reduced activation of small GTPase Cdc42 and decreased actin polymerization. Changes in rafts led to re-localization of TLR4 and TREM-1 to rafts, phosphorylation of ERK1/2, activation of NLRP3 inflammasome, and increased secretion of pro-inflammatory cytokines. The effects of exNef on lipid rafts and on inflammation were reversed by overexpression of a constitutively active mutant of Cdc42. Similar effects were observed in macrophages treated with exosomes produced by HIV-infected cells or isolated from plasma of HIV-infected subjects, but not with exosomes from cells and subjects infected with ΔNef-HIV or uninfected subjects. Mice injected with exNef exhibited monocytosis, reduced ABCA1 in macrophages, increased raft abundance in monocytes and augmented inflammation. Thus, Nef-containing exosomes potentiated pro-inflammatory response by inducing changes in cholesterol metabolism and reorganizing lipid rafts. These mechanisms may contribute to HIV-associated metabolic co-morbidities. HIV infects only a limited repertoire of cells expressing HIV receptors. Nevertheless, co-morbidities of HIV infection, such as atherosclerosis, dementia, renal impairment, myocardial pathology, abnormal haematopoiesis and others, involve dysfunction of cells that can not be infected by HIV. These co-morbidities persist even after successful application of antiretroviral therapy, when no virus is found in the blood. Many co-morbidities of HIV have a common element in their pathogenesis, impairment of cholesterol metabolism. In this study we show that HIV protein Nef released from infected cells in extracellular vesicles is taken up by un-infected (‘bystander’) cells impairing cholesterol metabolism in these cells. This impairment causes formation of excessive lipid rafts, re-localization of the inflammatory receptors into rafts, and triggers inflammation. These mechanisms may contribute to HIV-associated metabolic co-morbidities. Our work demonstrates how a single viral factor released from infected cells into circulation may cause a pleiotropy of pathogenic responses.
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to examine the evidence describing adipose tissue as a reservoir for HIV-1 and how this often expansive anatomic compartment contributes to HIV persistence. RECENT FINDINGS Memory CD4 T cells and macrophages, the major host cells for HIV, accumulate in adipose tissue during HIV/SIV infection of humans and rhesus macaques. Whereas HIV and SIV proviral DNA is detectable in CD4 T cells of multiple fat depots in virtually all infected humans and monkeys examined, viral RNA is less frequently detected, and infected macrophages may be less prevalent in adipose tissue. However, based on viral outgrowth assays, adipose-resident CD4 T cells are latently infected with virus that is replication-competent and infectious. Additionally, adipocytes interact with CD4 T cells and macrophages to promote immune cell activation and inflammation which may be supportive for HIV persistence. Antiviral effector cells, such as CD8 T cells and NK/NKT cells, are abundant in adipose tissue during HIV/SIV infection and typically exceed CD4 T cells, whereas B cells are largely absent from adipose tissue of humans and monkeys. Additionally, CD8 T cells in adipose tissue of HIV patients are activated and have a late differentiated phenotype, with unique TCR clonotypes of less diversity relative to blood CD8 T cells. With respect to the distribution of antiretroviral drugs in adipose tissue, data is limited, but there may be class-specific penetration of fat depots. The trafficking of infected immune cells within adipose tissues is a common event during HIV/SIV infection of humans and monkeys, but the virus may be mostly transcriptionally dormant. Viral replication may occur less in adipose tissue compared to other major reservoirs, such as lymphoid tissue, but replication competence and infectiousness of adipose latent virus are comparable to other tissues. Due to the ubiquitous nature of adipose tissue, inflammatory interactions among adipocytes and CD4 T cells and macrophages, and selective distribution of antiretroviral drugs, the sequestration of infected immune cells within fat depots likely represents a major challenge for cure efforts.
Collapse
Affiliation(s)
- Jacob Couturier
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St., MSB 2.112, Houston, TX, 77030, USA
| | - Dorothy E Lewis
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin St., MSB 2.112, Houston, TX, 77030, USA.
| |
Collapse
|
13
|
Low H, Hoang A, Pushkarsky T, Dubrovsky L, Dewar E, Di Yacovo MS, Mukhamedova N, Cheng L, Downs C, Simon G, Saumoy M, Hill AF, Fitzgerald ML, Nestel P, Dart A, Hoy J, Bukrinsky M, Sviridov D. HIV disease, metabolic dysfunction and atherosclerosis: A three year prospective study. PLoS One 2019; 14:e0215620. [PMID: 30998801 PMCID: PMC6472799 DOI: 10.1371/journal.pone.0215620] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022] Open
Abstract
HIV infection is known to be associated with cardiometabolic abnormalities; here we investigated the progression and causes of these abnormalities. Three groups of participants were recruited: HIV-negative subjects and two groups of treatment-naïve HIV-positive subjects, one group initiating antiretroviral treatment, the other remaining untreated. Intima-media thickness (cIMT) increased in HIV-positive untreated group compared to HIV-negative group, but treatment mitigated the difference. We found no increase in diabetes-related metabolic markers or in the level of inflammation in any of the groups. Total cholesterol, low density lipoprotein cholesterol and apoB levels were lower in HIV-positive groups, while triglyceride and Lp(a) levels did not differ between the groups. We found a statistically significant negative association between viral load and plasma levels of total cholesterol, LDL cholesterol, HDL cholesterol, apoA-I and apoB. HIV-positive patients had hypoalphalipoproteinemia at baseline, and we found a redistribution of sub-populations of high density lipoprotein (HDL) particles with increased proportion of smaller HDL in HIV-positive untreated patients, which may result from increased levels of plasma cholesteryl ester transfer protein in this group. HDL functionality declined in the HIV-negative and HIV-positive untreated groups, but not in HIV-positive treated group. We also found differences between HIV-positive and negative groups in plasma abundance of several microRNAs involved in lipid metabolism. Our data support a hypothesis that cardiometabolic abnormalities in HIV infection are caused by HIV and that antiretroviral treatment itself does not influence key cardiometabolic parameters, but mitigates those affected by HIV.
Collapse
Affiliation(s)
- Hann Low
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Anh Hoang
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Tatiana Pushkarsky
- Department of Microbiology, Immunology and Tropical Diseases, George Washington University, Washington, DC, United States of America
| | - Larisa Dubrovsky
- Department of Microbiology, Immunology and Tropical Diseases, George Washington University, Washington, DC, United States of America
| | - Elizabeth Dewar
- The Heart Centre, Alfred Hospital, Melbourne, VIC, Australia
| | - Maria-Silvana Di Yacovo
- HIV and STD Unit, Infectious Disease Service, Hospital Universitari de Bellvitge, Instituto de Investigación Biomédica de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain
| | | | - Lesley Cheng
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Catherine Downs
- Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
| | - Gary Simon
- Division of Infectious Diseases, Department of Medicine, George Washington University, Washington, DC, United States of America
| | - Maria Saumoy
- HIV and STD Unit, Infectious Disease Service, Hospital Universitari de Bellvitge, Instituto de Investigación Biomédica de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain
| | - Andrew F. Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Michael L. Fitzgerald
- Lipid Metabolism Unit, Centre for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Paul Nestel
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Anthony Dart
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- The Heart Centre, Alfred Hospital, Melbourne, VIC, Australia
| | - Jennifer Hoy
- Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Michael Bukrinsky
- Department of Microbiology, Immunology and Tropical Diseases, George Washington University, Washington, DC, United States of America
| | - Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Abstract
BACKGROUND HIV-associated atherosclerosis is a major comorbidity due, in part, to systemic effects of the virus on cholesterol metabolism. HIV protein Nef plays an important role in this pathology by impairing maturation of the main cellular cholesterol transporter ATP-Binding Cassette (ABCA) 1. ABCA1 maturation critically depends on calnexin, an integral endoplasmic reticulum membrane chaperone, and Nef binds to the cytoplasmic domain of calnexin and impairs interaction of calnexin with ABCA1. Overarching goal of the present study was to model Nef-calnexin interaction interface, and identify small molecule compounds potentially inhibiting this interaction. METHODS Molecular dynamics was utilized to build structure model of calnexin cytoplasmic domain, followed by global docking combined with application of QASDOM software developed by us for efficient analysis of receptor-ligand complexes. Structure-based virtual screening was performed for all sites identified by docking. A soluble analogue of a compound from the screening results list was tested for ability to down-regulate ABCA1. RESULTS We identified major interaction sites in calnexin and reciprocal sites in Nef. Virtual screening yielded a number of small-molecule compounds potentially blocking a calnexin site. Interestingly, one of the compounds, NSC13987, was previously identified by us as an inhibitor targeting a Nef site. An analogue of NSC13987, AMS-55, potently reversed the negative effect of Nef on ABCA1 abundance. CONCLUSIONS We have modelled Nef-calnexin interaction, predicted small molecule compounds that can potentially inhibit this interaction, and experimentally tested one of these compounds, confirming its effectiveness. These findings provide a platform for searching for new therapeutic agents to treat HIV-associated comorbidities.
Collapse
|
15
|
McNamara RP, Costantini LM, Myers TA, Schouest B, Maness NJ, Griffith JD, Damania BA, MacLean AG, Dittmer DP. Nef Secretion into Extracellular Vesicles or Exosomes Is Conserved across Human and Simian Immunodeficiency Viruses. mBio 2018; 9:e02344-17. [PMID: 29437924 PMCID: PMC5801467 DOI: 10.1128/mbio.02344-17] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 01/10/2018] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) or exosomes have been implicated in the pathophysiology of infections and cancer. The negative regulatory factor (Nef) encoded by simian immunodeficiency virus (SIV) and human immunodeficiency virus (HIV) plays a critical role in the progression to AIDS and impairs endosomal trafficking. Whether HIV-1 Nef can be loaded into EVs has been the subject of controversy, and nothing is known about the connection between SIV Nef and EVs. We find that both SIV and HIV-1 Nef proteins are present in affinity-purified EVs derived from cultured cells, as well as in EVs from SIV-infected macaques. Nef-positive EVs were functional, i.e., capable of membrane fusion and depositing their content into recipient cells. The EVs were able to transfer Nef into recipient cells. This suggests that Nef readily enters the exosome biogenesis pathway, whereas HIV virions are assembled at the plasma membrane. It suggests a novel mechanism by which lentiviruses can influence uninfected and uninfectable, i.e., CD4-negative, cells.IMPORTANCE Extracellular vesicles (EVs) transfer biologically active materials from one cell to another, either within the adjacent microenvironment or further removed. EVs also package viral RNAs, microRNAs, and proteins, which contributes to the pathophysiology of infection. In this report, we show that both human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) incorporate the virus-encoded Nef protein into EVs, including EVs circulating in the blood of SIV-infected macaques and that this presents a novel mechanism of Nef transfer to naive and even otherwise non-infectable cells. Nef is dispensable for viral replication but essential for AIDS progression in vivo Demonstrating that Nef incorporation into EVs is conserved across species implicates EVs as novel mediators of the pathophysiology of HIV. It could help explain the biological effects that HIV has on CD4-negative cells and EVs could become biomarkers of disease progression.
Collapse
Affiliation(s)
- Ryan P McNamara
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lindsey M Costantini
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - T Alix Myers
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Blake Schouest
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Jack D Griffith
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Blossom A Damania
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Andrew G MacLean
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
16
|
Mwangi WN, Vasoya D, Kgosana LB, Watson M, Nair V. Differentially expressed genes during spontaneous lytic switch of Marek's disease virus in lymphoblastoid cell lines determined by global gene expression profiling. J Gen Virol 2017; 98:779-790. [PMID: 28475033 PMCID: PMC5657026 DOI: 10.1099/jgv.0.000744] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Marek's disease virus (MDV), an alphaherpesvirus of poultry, causes Marek's disease and is characterized by visceral CD4+TCRαβ+ T-cell lymphomas in susceptible hosts. Immortal cell lines harbouring the viral genome have been generated from ex vivo cultures of MD tumours. As readily available sources of large numbers of cells, MDV-transformed lymphoblastoid cell lines (LCLs) are extremely valuable for studies of virus-host interaction. While the viral genome in most cells is held in a latent state, minor populations of cells display spontaneous reactivation identifiable by the expression of lytic viral genes. Spontaneous reactivation in these cells presents an opportunity to investigate the biological processes involved in the virus reactivation. For detailed characterization of the molecular events associated with reactivation, we used two lymphoblastoid cell lines derived from lymphomas induced by pRB1B-UL47eGFP, a recombinant MDV engineered to express enhanced green fluorescent protein (EGFP) fused with the UL47. We used fluorescence-activated cell sorting to purify the low-frequency EGFP-positive cells with a spontaneously activating viral genome from the majority EGFP-negative cells and analysed their gene expression profiles by RNA-seq using Illumina HiSeq2500. Ingenuity pathway analysis on more than 2000 differentially expressed genes between the lytically infected (EGFP-positive) and latently infected (EGFP-negative) cell populations identified the biological pathways involved in the reactivation. Virus-reactivating cells exhibited differential expression of a significant number of viral genes, with hierarchical differences in expression levels. Downregulation of a number of host genes including those directly involved in T-cell activation, such as CD3, CD28, ICOS and phospholipase C, was also noticed in the LCL undergoing lytic switch.
Collapse
Affiliation(s)
- William N Mwangi
- Avian Viral Diseases Programme, UK-China Centre of Excellence on Avian Disease Research, The Pirbright Institute, Pirbright, Surrey, UK
| | - Deepali Vasoya
- Division of Genetics and Genomics, The Roslin Institute, R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Lydia B Kgosana
- Avian Viral Diseases Programme, UK-China Centre of Excellence on Avian Disease Research, The Pirbright Institute, Pirbright, Surrey, UK
| | - Mick Watson
- Division of Genetics and Genomics, The Roslin Institute, R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Venugopal Nair
- Avian Viral Diseases Programme, UK-China Centre of Excellence on Avian Disease Research, The Pirbright Institute, Pirbright, Surrey, UK
| |
Collapse
|
17
|
Pushkarsky T, Shilov E, Kruglova N, Naumann R, Brichacek B, Jennelle L, Sviridov D, Kruglov A, Nedospasov SA, Bukrinsky M. Short Communication: Accumulation of Neutral Lipids in Liver and Aorta of Nef-Transgenic Mice. AIDS Res Hum Retroviruses 2017; 33:57-60. [PMID: 27649790 DOI: 10.1089/aid.2016.0128] [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] [Indexed: 12/19/2022] Open
Abstract
HIV-infected individuals are at high risk of developing atherosclerosis and cardiovascular disease, in part, due to HIV-induced impairment of cholesterol metabolism. In vitro studies demonstrated that HIV-1 protein Nef inhibits activity of ABCA1, the main cellular cholesterol transporter, leading to cholesterol accumulation in macrophages and conversion of these cells into foam cells, characteristic for atherosclerosis. However, the mechanisms of Nef-mediated effects on cholesterol metabolism in vivo are not well characterized. In this study, we generated Nef-transgenic mice and evaluated the accumulation of neutral lipids in liver and aorta of these animals. Nef expression was low in all transgenic mice, with some mice carrying the Nef transgene, but not expressing the Nef RNA. Using Oil Red O staining, we demonstrated increased levels of neutral lipids in liver and aorta of mice expressing Nef relative to transgenic animals, with no detectable Nef expression or control wild-type mice. These results provide direct evidence that Nef promotes cholesterol deposition in tissues.
Collapse
Affiliation(s)
- Tatiana Pushkarsky
- George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | | | | | - Ronald Naumann
- Max-Planck Institute for Molecular Genetics, Dresden, Germany
| | - Beda Brichacek
- George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Lucas Jennelle
- George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Andrei Kruglov
- Lomonosov Moscow State University, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- German Rheumatism Research Center, Berlin, Germany
| | - Sergei A. Nedospasov
- Lomonosov Moscow State University, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- German Rheumatism Research Center, Berlin, Germany
| | - Michael Bukrinsky
- George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Studies have shown that chronic inflammatory disorders, such as rheumatoid arthritis, systemic lupus erythematosus, and psoriasis are associated with an increased risk of atherosclerotic cardiovascular disease. The mechanism by which inflammation increases cardiovascular disease is likely multifactorial but changes in HDL structure and function that occur during inflammation could play a role. RECENT FINDINGS HDL levels decrease with inflammation and there are marked changes in HDL-associated proteins. Serum amyloid A markedly increases whereas apolipoprotein A-I, lecithin:cholesterol acyltransferase, cholesterol ester transfer protein, paraoxonase 1, and apolipoprotein M decrease. The exact mechanism by which inflammation decreases HDL levels is not defined but decreases in apolipoprotein A-I production, increases in serum amyloid A, increases in endothelial lipase and secretory phospholipase A2 activity, and decreases in lecithin:cholesterol acyltransferase activity could all contribute. The changes in HDL induced by inflammation reduce the ability of HDL to participate in reverse cholesterol transport and protect LDL from oxidation. SUMMARY During inflammation multiple changes in HDL structure occur leading to alterations in HDL function. In the short term, these changes may be beneficial resulting in an increase in cholesterol in peripheral cells to improve host defense and repair but over the long term these changes may increase the risk of atherosclerosis.
Collapse
Affiliation(s)
- Kenneth R Feingold
- Metabolism Section, Department of Veterans Affairs Medical Center, University of California San Francisco, San Francisco, California, USA
| | | |
Collapse
|
19
|
Does persistent HIV replication explain continued lymphoma incidence in the era of effective antiretroviral therapy? Curr Opin Virol 2016; 20:71-77. [PMID: 27665065 DOI: 10.1016/j.coviro.2016.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 12/16/2022]
Abstract
Non-Hodgkin lymphomas are highly increased in incidence in individuals infected with HIV, and this continues to be the case in spite of highly effective combined antiretroviral therapy (cART). New evidence has demonstrated that while successful virtual recovery of CD4 counts and elimination of HIV from peripheral blood can be achieved with cART, viral replication can still occur in lymphoid tissues. In addition, recent studies have suggested that adipose tissue provides an additional reservoir for HIV-infected macrophages and T lymphocytes even in the context of successful cART therapy. In this review article, we discuss possible mechanisms leading to the development of lymphoma in the cART era.
Collapse
|
20
|
Hunegnaw R, Vassylyeva M, Dubrovsky L, Pushkarsky T, Sviridov D, Anashkina AA, Üren A, Brichacek B, Vassylyev DG, Adzhubei AA, Bukrinsky M. Interaction Between HIV-1 Nef and Calnexin: From Modeling to Small Molecule Inhibitors Reversing HIV-Induced Lipid Accumulation. Arterioscler Thromb Vasc Biol 2016; 36:1758-71. [PMID: 27470515 DOI: 10.1161/atvbaha.116.307997] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 07/13/2016] [Indexed: 01/22/2023]
Abstract
OBJECTIVE HIV-infected patients are at an increased risk of developing atherosclerosis, in part because of downmodulation and functional impairment of ATP-binding cassette A1 (ABCA1) cholesterol transporter by the HIV-1 protein Nef. The mechanism of this effect involves Nef interacting with an ER chaperone calnexin and disrupting calnexin binding to ABCA1, leading to ABCA1 retention in ER, its degradation and resulting suppression of cholesterol efflux. However, molecular details of Nef-calnexin interaction remained unknown, limiting the translational impact of this finding. APPROACH AND RESULTS Here, we used molecular modeling and mutagenesis to characterize Nef-calnexin interaction and to identify small molecule compounds that could block it. We demonstrated that the interaction between Nef and calnexin is direct and can be reconstituted using recombinant proteins in vitro with a binding affinity of 89.1 nmol/L measured by surface plasmon resonance. The cytoplasmic tail of calnexin is essential and sufficient for interaction with Nef, and binds Nef with an affinity of 9.4 nmol/L. Replacing lysine residues in positions 4 and 7 of Nef with alanines abrogates Nef-calnexin interaction, prevents ABCA1 downregulation by Nef, and preserves cholesterol efflux from HIV-infected cells. Through virtual screening of the National Cancer Institute library of compounds, we identified a compound, 1[(7-oxo-7H-benz[de]anthracene-3-yl)amino]anthraquinone, which blocked Nef-calnexin interaction, partially restored ABCA1 activity in HIV-infected cells, and reduced foam cell formation in a culture of HIV-infected macrophages. CONCLUSION This study identifies potential targets that can be exploited to block the pathogenic effect of HIV infection on cholesterol metabolism and prevent atherosclerosis in HIV-infected subjects.
Collapse
Affiliation(s)
- Ruth Hunegnaw
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Marina Vassylyeva
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Larisa Dubrovsky
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Tatiana Pushkarsky
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Dmitri Sviridov
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Anastasia A Anashkina
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Aykut Üren
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Beda Brichacek
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Dmitry G Vassylyev
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü)
| | - Alexei A Adzhubei
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü).
| | - Michael Bukrinsky
- From the George Washington University School of Medicine and Health Sciences, Washington, DC (R.H., L.D., T.P., B.B., A.A.A., M.B.); University of Alabama School of Medicine and Dentistry, Birmingham, (M.V., D.V.); Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.S.); Engelhardt Institute of Molecular Biology RAS, Moscow, Russia (A.A. Anashkina, A.A. Adzhubei); and Georgetown University Medical Center, Lombardi Comprehensive Cancer Center, Washington, DC (A.Ü).
| |
Collapse
|
21
|
Couturier J, Agarwal N, Nehete PN, Baze WB, Barry MA, Jagannadha Sastry K, Balasubramanyam A, Lewis DE. Infectious SIV resides in adipose tissue and induces metabolic defects in chronically infected rhesus macaques. Retrovirology 2016; 13:30. [PMID: 27117277 PMCID: PMC4847269 DOI: 10.1186/s12977-016-0260-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/07/2016] [Indexed: 12/20/2022] Open
Abstract
Background HIV reservoirs pose major challenges to viral eradication. The main cellular reservoirs include CD4 T cells and macrophages, whereas anatomic reservoirs are thought to be primarily lymphoid tissues. Adipose tissue represents a potentially important non-lymphoid location for HIV replication and persistence because the stromal-vascular-fraction (AT-SVF) contains activated innate and adaptive immune cells that increase in number during infections, obesity, and chronic inflammation. Results Adipose tissue from two groups of SHIV-SF162p3-infected (~4 weeks acute infection) or SIVmac251-infected (~38 weeks chronic infection) rhesus macaques (N = 8 for each group) were studied for immune cell content, viral infectiousness, and metabolic health. The AT-SVF cells from SHIV-infected monkeys contained abundant memory CD4 and CD8 T cells, with fewer NKT cells and macrophages, and no B cells. Proviral DNA (Gag and Env) was readily detectable by nested PCR in AT-SVF cells from multiple adipose depots (subcutaneous and visceral) of acutely infected monkeys, but mostly from visceral fat. More importantly, viral outgrowth assays using input CD4 T cells derived from AT-SVF cells or peripheral blood of chronically infected monkeys resulted in robust replication of infectious virus from both AT-SVF and peripheral blood CD4 T cells. Chronically infected monkeys also experienced adipocyte dysfunction (suppression of major adipogenic genes) and systemic dyslipidemia (decreased serum total cholesterol and free fatty acids, and increased triglycerides), similar to metabolic abnormalities of HIV patients. Conclusions Adipose tissues of SIV-infected rhesus macaques become major compartments for infected immune cells, which in turn induce defects in adipose tissue metabolism. Electronic supplementary material The online version of this article (doi:10.1186/s12977-016-0260-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jacob Couturier
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Neeti Agarwal
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Diabetes Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Pramod N Nehete
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
| | - Wallace B Baze
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
| | - Michael A Barry
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - K Jagannadha Sastry
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA.,Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ashok Balasubramanyam
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Diabetes Research Center, Baylor College of Medicine, Houston, TX, USA.,Endocrine Service, Ben Taub General Hospital, Houston, TX, USA
| | - Dorothy E Lewis
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA. .,Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
22
|
Abstract
Cholesterol is an essential component of the cellular membranes and, by extension, of the HIV envelope membrane, which is derived from the host cell plasma membrane. Depletion of the cellular cholesterol has an inhibitory effect on HIV assembly, reduces infectivity of the produced virions, and makes the cell less susceptible to HIV infection. It is not surprising that the virus has evolved to gain access to cellular proteins regulating cholesterol metabolism. One of the key mechanisms used by HIV to maintain high levels of cholesterol in infected cells is Nef-mediated inhibition of cholesterol efflux and the cholesterol transporter responsible for this process, ABCA1. In this chapter, we describe methods to investigate these effects of HIV-1 infection.
Collapse
|
23
|
Siegel MO, Borkowska AG, Dubrovsky L, Roth M, Welti R, Roberts AD, Parenti DM, Simon GL, Sviridov D, Simmens S, Bukrinsky M, Fitzgerald ML. HIV infection induces structural and functional changes in high density lipoproteins. Atherosclerosis 2015; 243:19-29. [PMID: 26343868 PMCID: PMC4609619 DOI: 10.1016/j.atherosclerosis.2015.08.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 07/20/2015] [Accepted: 08/25/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Coronary artery disease is a growing clinical problem in HIV-infected subjects. The increased risk of coronary events in this population has been linked to low levels of HDL, but the effects of HIV infection and anti-retroviral treatment (ART) on HDL structure and function remain unknown. Here, we aimed to determine the composition and function of HDL particles isolated from ART-naive and ART-positive HIV-infected patients. METHODS AND RESULTS Proteomic profiling revealed decreased levels of paraoxonase (PON) 1 and PON 3 in HDL from HIV patients relative to HDL from uninfected controls (p < 0.0001), and PON activity of HDL from control group (0.13 ± 0.01 U/μl) was significantly higher than PON activity of HDL from HIV-infected untreated subjects (0.12 ± 0.01 U/μl, p = 0.0035), subjects treated with non-nucleoside reverse transcriptase inhibitor (NNRTI)-based therapy (0.11 ± 0.01 U/μl, p < 0.0001), subjects treated with protease inhibitor (PI)-based therapy with detectable viral load (0.11 ± 0.01 U/μl, p < 0.0001), and PI-treated patients with undetectable viral load (0.12 ± 0.01 U/μl, p = 0.0164). Lipidomic profiling uncovered a negative correlation between CD4 T cell counts and particle sphingomyelin, lyso-phosphatidylcholine and ether-linked phosphatidylserine content in the ART-naive (R(2) = 0.2611, p < 0.05; R(2) = 0.2722, p < 0.05; and R(2) = 0.3977, p < 0.05, respectively) but not treated HIV-infected subjects. Functional analysis demonstrated a negative correlation between cholesterol efflux capacity of HDL and viral load in the ART-naive HIV-infected group (R(2) = 0.26, p = 0.026). CONCLUSIONS Taken together, these results indicate that HIV infection associates with a number of both protein and lipid compositional changes in HDL particles. Moreover, HIV infection affects cholesterol efflux function of HDL, thus contributing to an increased risk of atherosclerosis in this patient population.
Collapse
Affiliation(s)
- Marc O Siegel
- Division of Infectious Diseases, Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Alison G Borkowska
- Lipid Metabolism Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Larisa Dubrovsky
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Mary Roth
- Kansas Lipidomics Research Center, Kansas State University, Manhattan, KS, USA
| | - Ruth Welti
- Kansas Lipidomics Research Center, Kansas State University, Manhattan, KS, USA
| | - Afsoon D Roberts
- Division of Infectious Diseases, Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - David M Parenti
- Division of Infectious Diseases, Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Gary L Simon
- Division of Infectious Diseases, Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Samuel Simmens
- Department of Epidemiology and Biostatistics, George Washington University, Milken Institute School of Public Health, Washington, DC, USA
| | - Michael Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| | - Michael L Fitzgerald
- Lipid Metabolism Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Low H, Cheng L, Di Yacovo MS, Churchill MJ, Meikle P, Bukrinsky M, Hill AF, Sviridov D. Lipid metabolism in patients infected with Nef-deficient HIV-1 strain. Atherosclerosis 2015; 244:22-8. [PMID: 26581048 DOI: 10.1016/j.atherosclerosis.2015.10.103] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/26/2015] [Accepted: 10/26/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND HIV protein Nef plays a key role in impairing cholesterol metabolism in both HIV infected and bystander cells. The existence of a small cohort of patients infected with Nef-deficient strain of HIV presented a unique opportunity to test the effect of Nef on lipid metabolism in a clinical setting. METHODS Here we report the results of a study comparing six patients infected with Nef-deficient strain of HIV (ΔNefHIV) with six treatment-naïve patients infected with wild-type HIV (WT HIV). Lipoprotein profile, size and functionality of high density lipoprotein (HDL) particles as well as lipidomic and microRNA profiles of patient plasma were analyzed. RESULTS We found that patients infected with ΔNefHIV had lower proportion of subjects with plasma HDL-C levels <1 mmol/l compared to patients infected with WT HIV. Furthermore, compared to a reference group of HIV-negative subjects, there was higher abundance of smaller under-lipidated HDL particles in plasma of patients infected with WT HIV, but not in those infected with ΔNefHIV. Lipidomic analysis of plasma revealed differences in abundance of phosphatidylserine and sphingolipids between patients infected with ΔNefHIV and WT HIV. MicroRNA profiling revealed that plasma abundance of 24 miRNAs, many of those involved in regulation of lipid metabolism, was differentially regulated by WT HIV and ΔNefHIV. CONCLUSION Our findings are consistent with HIV protein Nef playing a significant role in pathogenesis of lipid-related metabolic complications of HIV disease.
Collapse
Affiliation(s)
- Hann Low
- Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia
| | - Lesley Cheng
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Maria-Silvana Di Yacovo
- Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia; Institut de Resercha Biomedica Bellvitge, University of Barcelona, Gran Via de l'Hospitalet, 199, 08908 Hospitalet de Llobregat, Barcelona, Spain
| | - Melissa J Churchill
- Macfarlane Burnett Institute for Medical Research and Public Health, 85 Commercial Rd, Melbourne, VIC, 3004, Australia
| | - Peter Meikle
- Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia; Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Michael Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, 2300 I St. NW, Ross Hall, Washington DC, 20037, USA
| | - Andrew F Hill
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
25
|
Shahbaz S, Manicardi M, Guaraldi G, Raggi P. Cardiovascular disease in human immunodeficiency virus infected patients: A true or perceived risk? World J Cardiol 2015; 7:633-44. [PMID: 26516417 PMCID: PMC4620074 DOI: 10.4330/wjc.v7.i10.633] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 09/07/2015] [Accepted: 09/16/2015] [Indexed: 02/06/2023] Open
Abstract
After the successful introduction of highly active antiretroviral agents the survival of patients infected with the human immunodeficiency virus (HIV) in developed countries has increased substantially. This has allowed the surfacing of several chronic diseases among which cardiovascular disease (CVD) is prominent. The pathogenesis of CVD in HIV is complex and involves a combination of traditional and HIV related factors. An accurate assessment of risk of CVD in these patients is still elusive and as a consequence the most appropriate preventive and therapeutic interventions remain controversial.
Collapse
Affiliation(s)
- Shima Shahbaz
- Shima Shahbaz, Paolo Raggi, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton T6G 2B7, Alberta, Canada
| | - Marcella Manicardi
- Shima Shahbaz, Paolo Raggi, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton T6G 2B7, Alberta, Canada
| | - Giovanni Guaraldi
- Shima Shahbaz, Paolo Raggi, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton T6G 2B7, Alberta, Canada
| | - Paolo Raggi
- Shima Shahbaz, Paolo Raggi, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton T6G 2B7, Alberta, Canada
| |
Collapse
|
26
|
Lo J, Rosenberg ES, Fitzgerald ML, Bazner SB, Ihenachor EJ, Hawxhurst V, Borkowska AH, Wei J, Zimmerman CO, Burdo TH, Williams KC, Freeman MW, Grinspoon SK. High-density lipoprotein-mediated cholesterol efflux capacity is improved by treatment with antiretroviral therapy in acute human immunodeficiency virus infection. Open Forum Infect Dis 2014; 1:ofu108. [PMID: 25734176 PMCID: PMC4324225 DOI: 10.1093/ofid/ofu108] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 11/13/2014] [Indexed: 11/17/2022] Open
Abstract
Background Individuals infected with human immunodeficiency virus (HIV) have decreased high-density lipoprotein (HDL)-cholesterol and increased cardiovascular disease (CVD). Reverse cholesterol transport from macrophages may be inhibited by HIV and contribute to increased CVD. Human studies have not investigated longitudinal effects of HIV and antiretroviral therapy (ART) on cholesterol efflux. Methods Subjects with acute HIV infection were randomized to ART or not. Cholesterol efflux capacity was determined ex vivo after exposure of murine macrophages to apolipoprotein B-depleted patient sera obtained at baseline and after 12 weeks. Results After 12 weeks, HIV RNA decreased most in subjects randomized to ART. Available data on cholesterol demonstrated that efflux capacity from Abca1+/+ macrophages was increased most by sera obtained from ART-treated subjects (20.5% ± 5.0% to 24.3 % ± 6.9%, baseline to 12 weeks, P = .007; ART group [n = 6] vs 18.0 % ± 3.9% to 19.1 % ± 2.9%, baseline to 12 weeks, P = .30; untreated group [n = 6] [P = .04 ART vs untreated group]). Change in HIV RNA was negatively associated with change in Abca1+/+ macrophage cholesterol efflux (r = − 0.62, P = .03), and this finding remained significant (P = .03) after controlling for changes in HDL-cholesterol, CD4+ cells, and markers of monocyte or macrophage activation. Conclusions In subjects acutely infected with HIV, ATP-binding cassette transporter A1-mediated cholesterol efflux was stimulated to a greater degree over time by apolipoprotein B-depleted serum from subjects randomized to ART. The improvement in cholesterol efflux capacity is independently related to reduction in viral load.
Collapse
Affiliation(s)
- Janet Lo
- Program in Nutritional Metabolism
| | | | | | - Suzane B Bazner
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School , Boston
| | | | | | | | | | | | | | | | - Mason W Freeman
- Lipid Metabolism Unit/Center for Computational and Integrative Biology
| | | |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Pathogens of different taxa, from prions to protozoa, target cellular cholesterol metabolism to advance their own development and to impair host immune responses, but also causing metabolic complications, for example, atherosclerosis. This review describes recent findings of how pathogens do it. RECENT FINDINGS A common theme in interaction between pathogens and host cholesterol metabolism is pathogens targeting lipid rafts of the host plasma membrane. Many intracellular pathogens use rafts as an entry gate, taking advantage of the endocytic machinery and high abundance of outward-looking molecules that can be used as receptors. At the same time, disruption of the rafts' functional capacity, achieved by the pathogens through a number of various means, impairs the ability of the host to generate immune response, thus helping pathogen to thrive. Pathogens cannot synthesize cholesterol, and salvaging host cholesterol helps pathogens build advanced cholesterol-containing membranes and assembly platforms. Impact on cholesterol metabolism is not limited to the infected cells; proteins and microRNAs secreted by infected cells affect lipid metabolism systemically. SUMMARY Given an essential role that host cholesterol metabolism plays in pathogen development, targeting this interaction may be a viable strategy to fight infections, as well as metabolic complications of the infections.
Collapse
Affiliation(s)
- Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia
- Address correspondence to: Dmitri Sviridov, Baker IDI Heart and Diabetes Institute, PO Box 6492, Melbourne, VIC, 3004, Australia; Phone: +61385321363,
| | - Michael Bukrinsky
- George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| |
Collapse
|
28
|
Brichacek B, Darwish C, Popratiloff A, Dubrovsky L, Bukrinsky M. HIV-1 infection of macrophages induces retention of cholesterol transporter ABCA1 in the endoplasmic reticulum. AIDS Res Hum Retroviruses 2014; 30:947-8. [PMID: 25198127 DOI: 10.1089/aid.2014.0156] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Beda Brichacek
- George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Christina Darwish
- George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Anastas Popratiloff
- George Washington University Center for Microscopy and Image Analysis, Office of VP for Research, Washington, District of Columbia
| | - Larisa Dubrovsky
- George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Michael Bukrinsky
- George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| |
Collapse
|
29
|
Jennelle L, Hunegnaw R, Dubrovsky L, Pushkarsky T, Fitzgerald ML, Sviridov D, Popratiloff A, Brichacek B, Bukrinsky M. HIV-1 protein Nef inhibits activity of ATP-binding cassette transporter A1 by targeting endoplasmic reticulum chaperone calnexin. J Biol Chem 2014; 289:28870-84. [PMID: 25170080 DOI: 10.1074/jbc.m114.583591] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
HIV-infected patients are at increased risk of developing atherosclerosis, in part due to an altered high density lipoprotein profile exacerbated by down-modulation and impairment of ATP-binding cassette transporter A1 (ABCA1) activity by the HIV-1 protein Nef. However, the mechanisms of this Nef effect remain unknown. Here, we show that Nef interacts with an endoplasmic reticulum chaperone calnexin, which regulates folding and maturation of glycosylated proteins. Nef disrupted interaction between calnexin and ABCA1 but increased affinity and enhanced interaction of calnexin with HIV-1 gp160. The Nef mutant that did not bind to calnexin did not affect the calnexin-ABCA1 interaction. Interaction with calnexin was essential for functionality of ABCA1, as knockdown of calnexin blocked the ABCA1 exit from the endoplasmic reticulum, reduced ABCA1 abundance, and inhibited cholesterol efflux; the same effects were observed after Nef overexpression. However, the effects of calnexin knockdown and Nef on cholesterol efflux were not additive; in fact, the combined effect of these two factors together did not differ significantly from the effect of calnexin knockdown alone. Interestingly, gp160 and ABCA1 interacted with calnexin differently; although gp160 binding to calnexin was dependent on glycosylation, glycosylation was of little importance for the interaction between ABCA1 and calnexin. Thus, Nef regulates the activity of calnexin to stimulate its interaction with gp160 at the expense of ABCA1. This study identifies a mechanism for Nef-dependent inactivation of ABCA1 and dysregulation of cholesterol metabolism.
Collapse
Affiliation(s)
- Lucas Jennelle
- From the George Washington University School of Medicine and Health Sciences, Washington, D. C. 20037
| | - Ruth Hunegnaw
- From the George Washington University School of Medicine and Health Sciences, Washington, D. C. 20037
| | - Larisa Dubrovsky
- From the George Washington University School of Medicine and Health Sciences, Washington, D. C. 20037
| | - Tatiana Pushkarsky
- From the George Washington University School of Medicine and Health Sciences, Washington, D. C. 20037
| | - Michael L Fitzgerald
- the Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| | - Dmitri Sviridov
- the Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia, and
| | - Anastas Popratiloff
- the George Washington Center for Microscopy and Image Analysis, Office of VP for Research, Washington, D. C. 20037
| | - Beda Brichacek
- From the George Washington University School of Medicine and Health Sciences, Washington, D. C. 20037
| | - Michael Bukrinsky
- From the George Washington University School of Medicine and Health Sciences, Washington, D. C. 20037,
| |
Collapse
|
30
|
Mariani C, Desdouits M, Favard C, Benaroch P, Muriaux DM. Role of Gag and lipids during HIV-1 assembly in CD4(+) T cells and macrophages. Front Microbiol 2014; 5:312. [PMID: 25009540 PMCID: PMC4069574 DOI: 10.3389/fmicb.2014.00312] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 06/08/2014] [Indexed: 12/25/2022] Open
Abstract
HIV-1 is an RNA enveloped virus that preferentially infects CD4+ T lymphocytes and also macrophages. In CD4+ T cells, HIV-1 mainly buds from the host cell plasma membrane. The viral Gag polyprotein targets the plasma membrane and is the orchestrator of the HIV assembly as its expression is sufficient to promote the formation of virus-like particles carrying a lipidic envelope derived from the host cell membrane. Certain lipids are enriched in the viral membrane and are thought to play a key role in the assembly process and the envelop composition. A large body of work performed on infected CD4+ T cells has provided important knowledge about the assembly process and the membrane virus lipid composition. While HIV assembly and budding in macrophages is thought to follow the same general Gag-driven mechanism as in T-lymphocytes, the HIV cycle in macrophage exhibits specific features. In these cells, new virions bud from the limiting membrane of seemingly intracellular compartments, where they accumulate while remaining infectious. These structures are now often referred to as Virus Containing Compartments (VCCs). Recent studies suggest that VCCs represent intracellularly sequestered regions of the plasma membrane, but their precise nature remains elusive. The proteomic and lipidomic characterization of virions produced by T cells or macrophages has highlighted the similarity between their composition and that of the plasma membrane of producer cells, as well as their enrichment in acidic lipids, some components of raft lipids and in tetraspanin-enriched microdomains. It is likely that Gag promotes the coalescence of these components into an assembly platform from which viral budding takes place. How Gag exactly interacts with membrane lipids and what are the mechanisms involved in the interaction between the different membrane nanodomains within the assembly platform remains unclear. Here we review recent literature regarding the role of Gag and lipids on HIV-1 assembly in CD4+ T cells and macrophages.
Collapse
Affiliation(s)
- Charlotte Mariani
- Membrane Domains and Viral Assembly, CNRS UMR-5236, Centre d'étude d'agents Pathogènes et Biotechnologies pour la Santé Montpellier, Cedex, France
| | - Marion Desdouits
- Intracellular Transport and Immunity, Immunité et Cancer, Institut Curie - Inserm U932 Paris, France
| | - Cyril Favard
- Membrane Domains and Viral Assembly, CNRS UMR-5236, Centre d'étude d'agents Pathogènes et Biotechnologies pour la Santé Montpellier, Cedex, France
| | - Philippe Benaroch
- Intracellular Transport and Immunity, Immunité et Cancer, Institut Curie - Inserm U932 Paris, France
| | - Delphine M Muriaux
- Membrane Domains and Viral Assembly, CNRS UMR-5236, Centre d'étude d'agents Pathogènes et Biotechnologies pour la Santé Montpellier, Cedex, France
| |
Collapse
|
31
|
Monocytes as regulators of inflammation and HIV-related comorbidities during cART. J Immunol Res 2014; 2014:569819. [PMID: 25025081 PMCID: PMC4082935 DOI: 10.1155/2014/569819] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 05/15/2014] [Indexed: 12/17/2022] Open
Abstract
Combined antiretroviral therapy (cART) extends the lifespan and the quality of life for HIV-infected persons but does not completely eliminate chronic immune activation and inflammation. The low level of chronic immune activation persisting during cART-treated HIV infection is associated with the development of diseases which usually occur in the elderly. Although T-cell activation has been extensively examined in the context of cART-treated HIV infection, monocyte activation is only beginning to be recognized as an important source of inflammation in this context. Here we examine markers and sources of monocyte activation during cART-treated HIV infection and discuss the role of monocytes during cardiovascular disease, HIV-associated neurocognitive disorder, and innate immune aging.
Collapse
|
32
|
López-Calderón C, Palacios R, Cobo A, Nuño E, Ruiz J, Márquez M, Santos J. Serum ferritin in HIV-positive patients is related to immune deficiency and inflammatory activity. Int J STD AIDS 2014; 26:393-7. [PMID: 24912540 DOI: 10.1177/0956462414539669] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 05/19/2014] [Indexed: 11/17/2022]
Abstract
To analyse the prevalence of high ferritin levels in asymptomatic HIV patients and its related factors we conducted a cross-sectional study of a cohort of HIV outpatients in regular follow-up. Epidemiological, clinical, analytical and therapeutic data were collected. Patients completed a questionnaire about cardiovascular risk factors and underwent a physical examination and a 12-h fasting blood analysis. High ferritin levels were defined as a plasma ferritin level >200 µg/L in women and >300 µg/L in men. A total of 571 patients (78.1% men) were included. Median age was 43.2 years, HIV sexual transmission 68.5%, median CD4 count 474 cells/µL, 36.3% AIDS cases, 86.2% on antiretroviral therapy and 74.8% of them with undetectable viral load; 14.6% metabolic syndrome criteria, and mean cardiovascular risk at 10 years 6.67%. High ferritin levels prevalence was 11%, and related factors were a CD4 count <350 cells/µL (odds ratio, OR 2.37 [1.3-4.1], p = 0.003), ultrasensitive C-reactive protein >3 mg/L (OR 2.67 [1.5-4.7], p = 0.001) and chronic hepatitis C virus infection (OR 2.77 [1.5-4.9], p = 0.001). High ferritin levels are not uncommon in HIV patients, and they correlate with immunosuppression defined as CD4 count <350 cells/µL, higher ultrasensitive C-reactive protein and hepatitis C virus infection, and in contrast to the general population, they are not related to increased cardiovascular risk or metabolic syndrome.
Collapse
Affiliation(s)
| | - Rosario Palacios
- UGC Enfermedades Infecciosas, Hospital Virgen de la Victoria, Málaga, Spain FIMABIS, Málaga, Spain
| | - Andrés Cobo
- UGC Análisis Clínicos, Hospital Virgen de la Victoria, Málaga, Spain
| | - Enrique Nuño
- UGC Enfermedades Infecciosas, Hospital Virgen de la Victoria, Málaga, Spain FIMABIS, Málaga, Spain
| | - Josefa Ruiz
- UGC Enfermedades Infecciosas, Hospital Virgen de la Victoria, Málaga, Spain FIMABIS, Málaga, Spain
| | - Manuel Márquez
- UGC Enfermedades Infecciosas, Hospital Virgen de la Victoria, Málaga, Spain FIMABIS, Málaga, Spain
| | - Jesús Santos
- UGC Enfermedades Infecciosas, Hospital Virgen de la Victoria, Málaga, Spain FIMABIS, Málaga, Spain
| |
Collapse
|
33
|
Park IW, Fan Y, Luo X, Ryou MG, Liu J, Green L, He JJ. HIV-1 Nef is transferred from expressing T cells to hepatocytic cells through conduits and enhances HCV replication. PLoS One 2014; 9:e99545. [PMID: 24911518 PMCID: PMC4050050 DOI: 10.1371/journal.pone.0099545] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 05/16/2014] [Indexed: 12/15/2022] Open
Abstract
HIV-1 infection enhances HCV replication and as a consequence accelerates HCV-mediated hepatocellular carcinoma (HCC). However, the precise molecular mechanism by which this takes place is currently unknown. Our data showed that infectious HIV-1 failed to replicate in human hepatocytic cell lines. No discernible virus replication was observed, even when the cell lines transfected with HIV-1 proviral DNA were co-cultured with Jurkat T cells, indicating that the problem of liver deterioration in the co-infected patient is not due to the replication of HIV-1 in the hepatocytes of the HCV infected host. Instead, HIV-1 Nef protein was transferred from nef-expressing T cells to hepatocytic cells through conduits, wherein up to 16% (average 10%) of the cells harbored the transferred Nef, when the hepatocytic cells were co-cultured with nef-expressing Jurkat cells for 24 h. Further, Nef altered the size and numbers of lipid droplets (LD), and consistently up-regulated HCV replication by 1.5∼2.5 fold in the target subgenomic replicon cells, which is remarkable in relation to the initially indolent viral replication. Nef also dramatically augmented reactive oxygen species (ROS) production and enhanced ethanol-mediated up-regulation of HCV replication so as to accelerate HCC. Taken together, these data indicate that HIV-1 Nef is a critical element in accelerating progression of liver pathogenesis via enhancing HCV replication and coordinating modulation of key intra- and extra-cellular molecules for liver decay.
Collapse
Affiliation(s)
- In-Woo Park
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| | - Yan Fan
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Xiaoyu Luo
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Myoung-Gwi Ryou
- Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Jinfeng Liu
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Linden Green
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Johnny J. He
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| |
Collapse
|
34
|
Olivetta E, Tirelli V, Chiozzini C, Scazzocchio B, Romano I, Arenaccio C, Sanchez M. HIV-1 Nef impairs key functional activities in human macrophages through CD36 downregulation. PLoS One 2014; 9:e93699. [PMID: 24705461 PMCID: PMC3976297 DOI: 10.1371/journal.pone.0093699] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 03/08/2014] [Indexed: 01/01/2023] Open
Abstract
Monocytes and macrophages utilize the class A and B scavenger receptors to recognize and perform phagocytosis of invading microbes before a pathogen-specific immune response is generated. HIV-1 Nef protein affects the innate immune system impairing oxidative burst response and phagocytic capacity of macrophages. Our data show that exogenous recombinant myristoylated Nef protein induces a marked CD36 downregulation in monocytes from Peripheral Blood Mononuclear Cells, in Monocyte-Derived Macrophages (MDMs) differentiated by cytokines and in MDMs contained in a mixed culture obtained expanding PBMCs under Human Erythroid Massive Amplification condition. Under the latter culture condition we identify three main populations after 6 days of expansion: lymphocytes (37.8±14.7%), erythroblasts (46.7±6.1%) and MDMs (15.7±7.5%). The Nef addition to the cell culture significantly downregulates CD36 expression in MDMs, but not in erythroid cells. Furthermore, CD36 inhibition is highly specific since it does not modify the expression levels of other MDM markers such as CD14, CD11c, CD86, CD68, CD206, Toll-like Receptor 2 and Toll-like Receptor 4. Similar results were obtained in MDMs infected with VSV-G pseudotyped HIV-1-expressing Nef. The reduced CD36 membrane expression is associated with decrease of correspondent CD36 mRNA transcript. Furthermore, Nef-induced CD36 downregulation is linked to both impaired scavenger activity with reduced capability to take up oxidized lipoproteins and to significant decreased phagocytosis of fluorescent beads and GFP-expressing Salmonella tiphymurium. In addition we observed that Nef induces TNF-α release in MDMs. Although these data suggest a possible involvement of TNF-α in mediating Nef activity, our results exclude a possible relationship between Nef-induced TNF-α release and Nef-mediated CD36 downregulation. The present work shows that HIV-1 Nef protein may have a role in the strategies elaborated by HIV-1 to alter pathogen disease outcomes, by modulating CD36 expression in macrophages, favoring the onset of opportunistic infections in HIV-1 infected people.
Collapse
Affiliation(s)
| | - Valentina Tirelli
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | | | - Beatrice Scazzocchio
- Department of Veterinary, Public Health and Food Safety, Istituto Superiore di Sanità, Rome, Italy
| | - Ignazio Romano
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Claudia Arenaccio
- National AIDS Centre, Istituto Superiore di Sanità, Rome, Italy; Department of Science, University Roma TRE, Rome, Italy
| | - Massimo Sanchez
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
35
|
Cui HL, Ditiatkovski M, Kesani R, Bobryshev YV, Liu Y, Geyer M, Mukhamedova N, Bukrinsky M, Sviridov D. HIV protein Nef causes dyslipidemia and formation of foam cells in mouse models of atherosclerosis. FASEB J 2014; 28:2828-39. [PMID: 24642731 DOI: 10.1096/fj.13-246876] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Patients with HIV are at an increased risk of cardiovascular disease. In this study we investigated the effect of Nef, a secreted HIV protein responsible for the impairment of cholesterol efflux, on the development of atherosclerosis in two animal models. ApoE(-/-) mice fed a high-fat diet and C57BL/6 mice fed a high-fat, high-cholesterol diet were injected with recombinant Nef (40 ng/injection) or vehicle, and the effects of Nef on development of atherosclerosis, inflammation, and dyslipidemia were assessed. In apoE(-/-) mice, Nef significantly increased the size of atherosclerotic lesions and caused vessel remodeling. Nef caused elevation of total cholesterol and triglyceride levels in the plasma while reducing high-density lipoprotein cholesterol levels. These changes were accompanied by a reduction of ABCA1 abundance in the liver, but not in the vessels. In C57BL/6 mice, Nef caused a significant number of lipid-laden macrophages presented in adventitia of the vessels; these cells were absent from the vessels of control mice. Nef caused sharp elevations of plasma triglyceride levels and body weight. Taken together, our findings suggest that Nef causes dyslipidemia and accumulation of cholesterol in macrophages within the vessel wall, supporting the role of Nef in pathogenesis of atherosclerosis in HIV-infected patients.-Cui, H. L., Ditiatkovski, M., Kesani, R., Bobryshev, Y. V., Liu, Y., Geyer, M., Mukhamedova, N., Bukrinsky, M., Sviridov, D. HIV protein Nef causes dyslipidemia and formation of foam cells in mouse models of atherosclerosis.
Collapse
Affiliation(s)
- Huanhuan L Cui
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Rajitha Kesani
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yuri V Bobryshev
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Yingying Liu
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Matthias Geyer
- Center for Advanced European Studies and Research (CAESAR), Bonn, Germany; and
| | | | - Michael Bukrinsky
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, District of Columbia, USA
| | - Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia;
| |
Collapse
|
36
|
Jacob D, Hunegnaw R, Sabyrzyanova TA, Pushkarsky T, Chekhov VO, Adzhubei AA, Kalebina TS, Bukrinsky M. The ABCA1 domain responsible for interaction with HIV-1 Nef is conformational and not linear. Biochem Biophys Res Commun 2014; 444:19-23. [PMID: 24406162 DOI: 10.1016/j.bbrc.2013.12.141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 12/30/2013] [Indexed: 12/11/2022]
Abstract
HIV-1 Nef is an accessory protein responsible for inactivation of a number of host cell proteins essential for anti-viral immune responses. In most cases, Nef binds to the target protein and directs it to a degradation pathway. Our previous studies demonstrated that Nef impairs activity of the cellular cholesterol transporter, ABCA1, and that Nef interacts with ABCA1. Mutation of the (2226)DDDHLK motif in the C-terminal cytoplasmic tail of ABCA1 disrupted interaction with Nef. Here, we tested Nef interaction with the ABCA1 C-terminal cytoplasmic fragment using yeast 2-hybrid system assay and co-immunoprecipitation analysis in human cells. Surprisingly, analysis in a yeast 2-hybrid system did not reveal any interaction between Nef and the C-terminal cytoplasmic fragment of ABCA1. Using co-immunoprecipitation from HEK 293T cells expressing these polypeptides, only a very weak interaction could be detected. The (2226)DDDHLK motif in the C-terminal cytoplasmic tail of ABCA1 found previously to be essential for interaction between ABCA1 and Nef is insufficient to bestow strong binding to Nef. Molecular modeling suggested that interaction with Nef may be mediated by a conformational epitope composed of the sequences within the cytoplasmic loop of ABCA1 and the C-terminal cytoplasmic domain. Studies are now underway to characterize this epitope.
Collapse
Affiliation(s)
- Daria Jacob
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, Moscow 119899, Russia
| | - Ruth Hunegnaw
- Department of Microbiology, Immunology and Tropical Medicine, GWU School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Tatyana A Sabyrzyanova
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, Moscow 119899, Russia
| | - Tatiana Pushkarsky
- Department of Microbiology, Immunology and Tropical Medicine, GWU School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Vladimir O Chekhov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov St. 32, 119991 Moscow, Russia
| | - Alexei A Adzhubei
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov St. 32, 119991 Moscow, Russia
| | - Tatyana S Kalebina
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, Moscow 119899, Russia
| | - Michael Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, GWU School of Medicine and Health Sciences, Washington, DC 20037, USA.
| |
Collapse
|
37
|
Asztalos BF, Matera R, Horvath KV, Horan M, Tani M, Polak JF, Skinner S, Wanke CA. Cardiovascular Disease-Risk Markers in HIV Patients. ACTA ACUST UNITED AC 2014; 5. [PMID: 26005590 PMCID: PMC4439003 DOI: 10.4172/2155-6113.1000317] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Objectives HIV-positive patients have an increased risk for CVD; however, the underlying mechanisms are not well understood. Our goal was to assess traditional and emerging CVD-risk factors in the CARE Study, a well-described cohort of HIV-infected adults. Methods We analyzed demographic and clinical (viral load, CD4 count, ART regimen, cIMT) data including markers of lipid and glucose homeostasis in 176 HIV-positive subjects receiving regular care for HIV infection. Results No significant association between cIMT and LDL-C level was observed. HIV patients had significantly lower level of the large α-1 HDL particles and about 3-fold higher level of the small pre β-1 HDL particles than the normal population, but these parameters were not significantly associated with cIMT. Components of the metabolic syndrome, high TG/low HDL-C, insulin resistance and high BMI, as well as viral load were significant but moderate contributors to increased cIMT. Conclusion The major lipid disorder was low HDL-C and high TG level in this HIV-positive cohort. LDL-C was not elevated. These and previously published data indicate that HIV infection and HIV medications influence CVD risk by impairing cholesterol removal (efflux) via ABCA1 from macrophages. Decreasing CVD risk in HIV patients, with impaired cholesterol efflux from macrophages, may require a lower LDL-C goal than recommended for HIV-negative patients and also a better control of TG level.
Collapse
Affiliation(s)
- Bela F Asztalos
- Lipid Metabolism Laboratory, Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA, USA ; Division of Nutrition and Infection, Department of Public Health and Community Medicine, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA, USA
| | - Robert Matera
- Lipid Metabolism Laboratory, Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA, USA
| | - Katalin V Horvath
- Lipid Metabolism Laboratory, Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA, USA
| | - Michael Horan
- Lipid Metabolism Laboratory, Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA, USA
| | - Mariko Tani
- Lipid Metabolism Laboratory, Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA, USA
| | - Joseph F Polak
- Department of Radiology, Tufts University School of Medicine, 145 Harrison Avenue, Boston, MA, USA
| | - Sally Skinner
- Division of Nutrition and Infection, Department of Public Health and Community Medicine, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA, USA
| | - Christine A Wanke
- Division of Nutrition and Infection, Department of Public Health and Community Medicine, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA, USA
| |
Collapse
|
38
|
Mansfield KG, Sasseville VG, Westmoreland SV. Molecular Localization Techniques in the Diagnosis and Characterization of Nonhuman Primate Infectious Diseases. Vet Pathol 2013; 51:110-26. [DOI: 10.1177/0300985813509386] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Molecular localization techniques remain important diagnostic and research tools for the pathologist evaluating nonhuman primate tissues. In situ hybridization and immunohistochemistry protocols have been developed for many important pathogens of nonhuman primates, including RNA and DNA viruses, prions, and bacterial, protozoal, and fungal pathogens. Such techniques will remain critical in defining the impact and relevance of novel agents on animal health and disease. A comparative pathology perspective often provides valuable insight to the best strategy for reagent development and can also facilitate interpretation of molecular localization patterns. Such a perspective is grounded in a firm understanding of microbe-host pathobiology. This review summarizes current molecular localization protocols used in the diagnosis of selected primate infectious diseases.
Collapse
Affiliation(s)
- K. G. Mansfield
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | - S. V. Westmoreland
- New England Primate Research Center, Harvard Medical School, Southborough, MA, USA
| |
Collapse
|
39
|
Lamers SL, Fogel GB, Singer EJ, Salemi M, Nolan DJ, Huysentruyt LC, McGrath MS. HIV-1 Nef in macrophage-mediated disease pathogenesis. Int Rev Immunol 2013; 31:432-50. [PMID: 23215766 DOI: 10.3109/08830185.2012.737073] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Combined anti-retroviral therapy (cART) has significantly reduced the number of AIDS-associated illnesses and changed the course of HIV-1 disease in developed countries. Despite the ability of cART to maintain high CD4+ T-cell counts, a number of macrophage-mediated diseases can still occur in HIV-infected subjects. These diseases include lymphoma, metabolic diseases, and HIV-associated neurological disorders. Within macrophages, the HIV-1 regulatory protein "Nef" can modulate surface receptors, interact with signaling pathways, and promote specific environments that contribute to each of these pathologies. Moreover, genetic variation in Nef may also guide the macrophage response. Herein, we review findings relating to the Nef-macrophage interaction and how this relationship contributes to disease pathogenesis.
Collapse
|
40
|
Bonilla H, Mcshannic J, Goldberg E, Chua D, Conner R, Fiorentino M, McComsey G. Impact of Human Immunodeficiency Virus Infection on Measures of Cardiovascular Disease in Long-Term Nonprogressors. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2013. [DOI: 10.1097/ipc.0b013e31828262f3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
41
|
Feeney ER, McAuley N, O'Halloran JA, Rock C, Low J, Satchell CS, Lambert JS, Sheehan GJ, Mallon PWG. The expression of cholesterol metabolism genes in monocytes from HIV-infected subjects suggests intracellular cholesterol accumulation. J Infect Dis 2012. [PMID: 23204179 DOI: 10.1093/infdis/jis723] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV) infection is associated with increased cardiovascular risk and reduced high-density lipoprotein cholesterol (HDL-c). In vitro, HIV impairs monocyte-macrophage cholesterol efflux, a major determinant of circulating HDL-c, by increasing ABCA1 degradation, with compensatory upregulation of ABCA1 messenger RNA (mRNA). METHODS We examined expression of genes involved in cholesterol uptake, metabolism, and efflux in monocytes from 22 HIV-positive subjects on antiretroviral therapy (ART-Treated), 30 untreated HIV-positive subjects (ART-Naive), and 22 HIV-negative controls (HIV-Neg). RESULTS HDL-c was lower and expression of ABCA1 mRNA was higher in ART-Naive subjects than in both ART-Treated and HIV-Neg subjects (both P < .01), with HDL-c inversely correlated with HIV RNA (ρ = -0.52; P < .01). Expression of genes involved in cholesterol uptake (LDLR, CD36), synthesis (HMGCR), and regulation (SREBP2, LXRA) was significantly lower in both ART-Treated and ART-Naive subjects than in HIV-Neg controls. CONCLUSIONS In vivo, increased monocyte ABCA1 expression in untreated HIV-infected patients and normalization of ABCA1 expression with virological suppression by ART supports direct HIV-induced impairment of cholesterol efflux previously demonstrated in vitro. However, decreased expression of cholesterol sensing, uptake, and synthesis genes in both untreated and treated HIV infection suggests that both HIV and ART affect monocyte cholesterol metabolism in a pattern consistent with accumulation of intramonocyte cholesterol.
Collapse
Affiliation(s)
- Eoin R Feeney
- HIV Molecular Research Group, School of Medicine and Medical Science, University College Dublin, Ireland.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Saxena SK, Shrivastava G, Tiwari S, Swamy MA, Nair MP. Modulation of HIV pathogenesis and T-cell signaling by HIV-1 Nef. Future Virol 2012; 7:609-620. [PMID: 22844345 DOI: 10.2217/fvl.12.42] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
HIV-1 Nef protein is an approximately 27-kDa myristoylated protein that is a virulence factor essential for efficient viral replication and infection in CD4(+) T cells. The functions of CD4(+) T cells are directly impeded after HIV infection. HIV-1 Nef plays a crucial role in manipulating host cellular machinery and in HIV pathogenesis by reducing the ability of infected lymphocytes to form immunological synapses by promoting virological synapses with APCs, and by affecting T-cell stimulation. This article reviews the current status of the efficient Nef-mediated spread of virus in the unreceptive environment of the immune system by altering CD4(+) T-lymphocyte signaling, intracellular trafficking, cell migration and apoptotic pathways.
Collapse
Affiliation(s)
- Shailendra K Saxena
- CSIR-Centre for Cellular & Molecular Biology, Uppal Road, Hyderabad 500007 (AP), India
| | | | | | | | | |
Collapse
|
43
|
Dubrovsky L, Van Duyne R, Senina S, Guendel I, Pushkarsky T, Sviridov D, Kashanchi F, Bukrinsky M. Liver X receptor agonist inhibits HIV-1 replication and prevents HIV-induced reduction of plasma HDL in humanized mouse model of HIV infection. Biochem Biophys Res Commun 2012; 419:95-8. [PMID: 22326260 DOI: 10.1016/j.bbrc.2012.01.137] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 01/27/2012] [Indexed: 01/24/2023]
Abstract
HIV-infected subjects are at high risk of developing atherosclerosis, in part due to virus-induced impairment of HDL metabolism. Here, using as a model of HIV infection the NOD.Cg-Prkdc(scid)IL2rg(tm1Wjl)/SzJ (NSG) mice humanized by human stem cell transplantation, we demonstrate that LXR agonist TO901317 potently reduces viral replication and prevents HIV-induced reduction of plasma HDL. These results establish that humanized mice can be used to investigate the mechanisms of HIV-induced impairment of HDL formation, a major feature of dyslipidemia associated with HIV-1 infection, and show potential benefits of developing LXR agonists for treatment of HIV-associated cardio-vascular disease.
Collapse
|
44
|
Cui HL, Grant A, Mukhamedova N, Pushkarsky T, Jennelle L, Dubrovsky L, Gaus K, Fitzgerald ML, Sviridov D, Bukrinsky M. HIV-1 Nef mobilizes lipid rafts in macrophages through a pathway that competes with ABCA1-dependent cholesterol efflux. J Lipid Res 2012; 53:696-708. [PMID: 22262807 DOI: 10.1194/jlr.m023119] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HIV infection, through the actions of viral accessory protein Nef, impairs activity of cholesterol transporter ABCA1, inhibiting cholesterol efflux from macrophages and elevating the risk of atherosclerosis. Nef also induces lipid raft formation. In this study, we demonstrate that these activities are tightly linked and affect macrophage function and HIV replication. Nef stimulated lipid raft formation in macrophage cell line RAW 264.7, and lipid rafts were also mobilized in HIV-1-infected human monocyte-derived macrophages. Nef-mediated transfer of cholesterol to lipid rafts competed with the ABCA1-dependent pathway of cholesterol efflux, and pharmacological inhibition of ABCA1 functionality or suppression of ABCA1 expression by RNAi increased Nef-dependent delivery of cholesterol to lipid rafts. Nef reduced cell-surface accessibility of ABCA1 and induced ABCA1 catabolism via the lysosomal pathway. Despite increasing the abundance of lipid rafts, expression of Nef impaired phagocytic functions of macrophages. The infectivity of the virus produced in natural target cells of HIV-1 negatively correlated with the level of ABCA1. These findings demonstrate that Nef-dependent inhibition of ABCA1 is an essential component of the viral replication strategy and underscore the role of ABCA1 as an innate anti-HIV factor.
Collapse
Affiliation(s)
- Huanhuan L Cui
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Inflammation predicts changes in high-density lipoprotein particles and apolipoprotein A1 following initiation of antiretroviral therapy. AIDS 2011; 25:2133-42. [PMID: 21857489 DOI: 10.1097/qad.0b013e32834be088] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND The effects of HIV infection and antiretroviral therapy (ART) on usual lipid levels have been reported. The effects of initiating versus deferring ART on high-density and low-density lipoprotein particle (HDL-P and LDL-P, respectively) concentrations and apolipoprotein (Apo) levels are not well described. METHODS In a subgroup of participants not taking ART at study entry who were randomized in the Strategies for Management of Antiretroviral Therapy (SMART) trial to immediately initiate ART ('viral suppression group') or to defer it ('drug conservation group'), lipoprotein particle concentrations and ApoA1 and ApoB levels were measured at baseline and at 2 and 6 months following randomization. RESULTS Compared with drug conservation group (n = 126), HDL-P and ApoA1 levels increased among viral suppression participants (n = 128) after starting ART. At 6 months, viral suppression participants had 13% higher total HDL-P (P < 0.001) and 9% higher ApoA1 (P < 0.001). LDL-P, very low density lipoprotein particle, and ApoB did not differ significantly between the viral suppression and drug conservation groups. Among viral suppression participants, predictors of HDL-P and ApoA1 increases included baseline levels of high-sensitivity C-reactive protein (hsCRP) and interleukin 6 (IL-6), but not HIV RNA level, CD4 cell count, or traditional cardiovascular disease risk factors. The effect of starting ART on changes in HDL-P and ApoA1 was greater for those with higher versus lower baseline levels of IL-6 (P = 0.001 and 0.08, respectively, for interaction) or hsCRP (P = 0.01 and 0.04, respectively, for interaction). CONCLUSION HDL-P and ApoA1 increase following ART initiation, to a degree that depends on the degree of inflammation present at entry. These findings suggest that activation of inflammatory pathways contribute to HIV-associated changes in HDL.
Collapse
|
46
|
Hierro y riesgo cardiovascular en pacientes con infección por el virus de la inmunodeficiencia humana. Med Clin (Barc) 2011; 137:141-2. [DOI: 10.1016/j.medcli.2010.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 07/29/2010] [Indexed: 11/21/2022]
|