1
|
Valyaeva AA, Tikhomirova MA, Feng J, Zharikova AA, Potashnikova DM, Musinova YR, Mironov AA, Vassetzky YS, Sheval EV. Compensatory reactions of B cells in response to chronic HIV-1 Tat exposure. J Cell Physiol 2024:e31459. [PMID: 39373061 DOI: 10.1002/jcp.31459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 09/13/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024]
Abstract
Patients infected with human immunodeficiency virus-1 (HIV-1) have an increased incidence of B-cell lymphoma, even though HIV-1 does not infect B cells. The development of B-cell lymphomas appears to be related to the action of the HIV-1 transactivator protein (Tat), which is released from HIV-infected cells and penetrates uninfected B cells, affecting host cell gene expression. Upon chronic HIV-1 infection, Tat acts on the cells for a long time, probably allowing the cells to adapt to the presence of the viral protein. The aim of this work was to identify and study the mechanism of adaptation of cells to prolonged (chronic) exposure to HIV-1 Tat. We performed a comparative analysis of cells expressing Tat under the action of either an inducible promoter or a constitutive promoter, allowing us to model acute and chronic Tat effects, respectively. We found that the acute action of Tat leads to the suppression of cell proliferation, probably due to the downregulation of genes associated with replication and protein synthesis. In the case of chronic action of Tat, cell proliferation was restored and the expression of genes associated with the implementation of protective (antiviral) functions of the cell was increased. Analysis using proteasome inhibitors showed that in the case of chronic action, intense Tat proteolysis occurred, which could be the main mechanism of B-cell adaptation. Thus, B cells have a powerful mechanism to adapt to the entry of HIV-1 Tat, the efficiency of which may determine the frequency of lymphomagenesis in HIV-1-infected patients.
Collapse
Affiliation(s)
- Anna A Valyaeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Maria A Tikhomirova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Junyi Feng
- CNRS, UMR 9018, Université Paris-Saclay, Institut Gustave Roussy, Villejuif, France
| | - Anastasia A Zharikova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Daria M Potashnikova
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Yana R Musinova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey A Mironov
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Yegor S Vassetzky
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
- CNRS, UMR 9018, Université Paris-Saclay, Institut Gustave Roussy, Villejuif, France
| | - Eugene V Sheval
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
2
|
Zhu J, Cirincione AB, Strauss MJ, Davis SE, Eans SO, Tribbitt DK, Alshakhshir N, McLaughlin JP. Impact of HIV-1 tat protein on methamphetamine-induced inhibition of vesicular monoamine transporter2-mediated dopamine transport and methamphetamine conditioned place preference in HIV-1 tat transgenic mice. Eur J Pharmacol 2024; 984:177030. [PMID: 39366503 DOI: 10.1016/j.ejphar.2024.177030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/06/2024]
Abstract
Perturbation of dopamine transmission has been implicated as a contributing factor in HIV-1 associated neurocognitive disorders with concurrent methamphetamine (METH) abuse. We have demonstrated that the HIV-1 protein, transactivator of transcription (Tat), decreases dopamine transport through inhibition of vesicular monoamine transporter2 (VMAT2). This study determined the effects of Tat protein on METH-inhibited VMAT2 function and METH-conditioned place preference (CPP). In vitro exposure of isolated mouse whole brain vesicles to recombinant Tat1-86 or METH displayed a concentration-dependent inhibition of the vesicular [3H]Dopamine uptake, in which a combination of Tat and METH induced a greater reduction of dopamine uptake compared to Tat or METH alone. In vivo, the maximal velocity (Vmax) of vesicular [3H]Dopamine uptake was decreased in inducible Tat transgenic (iTat-tg) mice harvested after treatment with either 21-day doxycycline (Dox) or 14-day METH (3 mg/kg, i.p., daily), whereas these mice treated with both Dox and METH displayed an additive reduction of the Vmax compared to either Tat or METH alone. Moreover, Dox-induced Tat expression increased METH-CPP in an exposure-dependent manner, with iTat-tg mice demonstrating a 2.3-fold potentiation of METH-CPP compared with Tat null control mice upon administration of Dox for 14 days. Furthermore, a 7-day administration of Dox reinstated extinguished METH-CPP. Collectively, these results suggest a synergistic effect of Tat protein and METH on inhibition of VMAT2-mediated DA transport, potentially contributing to potentiation of METH-CPP in iTat-tg mice.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA.
| | - Abagail B Cirincione
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Matthew J Strauss
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Sarah E Davis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Shainnel O Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Danielle K Tribbitt
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Nadine Alshakhshir
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| |
Collapse
|
3
|
Duffy BC, King KM, Nepal B, Nonnemacher MR, Kortagere S. Acute Administration of HIV-1 Tat Protein Drives Glutamatergic Alterations in a Rodent Model of HIV-Associated Neurocognitive Disorders. Mol Neurobiol 2024; 61:8467-8480. [PMID: 38514527 DOI: 10.1007/s12035-024-04113-8] [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: 01/20/2024] [Accepted: 03/12/2024] [Indexed: 03/23/2024]
Abstract
HIV-1-associated neurocognitive disorders (HAND) are a major comorbidity of HIV-1 infection, marked by impairment of executive function varying in severity. HAND affects nearly half of people living with HIV (PLWH), with mild forms predominating since the use of anti-retroviral therapies (ART). The HIV-1 transactivator of transcription (Tat) protein is found in the cerebrospinal fluid of patients adherent to ART, and its administration or expression in animals causes cognitive symptoms. Studies of Tat interaction with the N-methyl-D-aspartate receptor (NMDAR) suggest that glutamate toxicity contributes to Tat-induced impairments. To identify changes in regional glutamatergic circuitry underlying cognitive impairment, we injected recombinant Tat86 or saline to medial prefrontal cortex (mPFC) of male Sprague-Dawley rats. Rats were assessed with behavioral tasks that involve intact functioning of mPFC including the novel object recognition (NOR), spatial object recognition (SOR), and temporal order (TO) tasks at 1 and 2 postoperative weeks. Following testing, mPFC tissue was collected and analyzed by RT-PCR. Results showed Tat86 in mPFC-induced impairment in SOR, and upregulation of Grin1 and Grin2a transcripts. To further understand the mechanism of Tat toxicity, we assessed the effects of full-length Tat101 on gene expression in mPFC by RNA sequencing. The results of RNAseq suggest that glutamatergic effects of Tat86 are maintained with Tat101, as Grin2a was upregulated in Tat101-injected tissue, among other differentially expressed genes. Spatial learning and memory impairment and Grin2a upregulation suggest that exposure to Tat protein drives adaptation in mPFC, altering the function of circuitry supporting spatial learning and memory.
Collapse
Affiliation(s)
- Brenna C Duffy
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Kirsten M King
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Binod Nepal
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA.
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA.
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Cui BC, Aksenova M, Sikirzhytskaya A, Odhiambo D, Korunova E, Sikirzhytski V, Ji H, Altomare D, Broude E, Frizzell N, Booze R, Wyatt MD, Shtutman M. Suppression of HIV-TAT and cocaine-induced neurotoxicity and inflammation by cell penetrable itaconate esters. J Neurovirol 2024; 30:337-352. [PMID: 38884890 PMCID: PMC11512888 DOI: 10.1007/s13365-024-01216-9] [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: 11/17/2023] [Revised: 04/20/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024]
Abstract
HIV-associated neurological disorder (HAND) is a serious complication of HIV infection marked by neurotoxicity induced by viral proteins like Tat. Substance abuse exacerbates neurocognitive impairment in people living with HIV. There is an urgent need for therapeutic strategies to combat HAND comorbid with Cocaine Use Disorder (CUD). Our analysis of HIV and cocaine-induced transcriptomes in primary cortical cultures revealed significant overexpression of the macrophage-specific gene aconitate decarboxylase 1 (Acod1). The ACOD1 protein converts the tricarboxylic acid intermediate cis-aconitate into itaconate during the activation of inflammation. Itaconate then facilitates cytokine production and activates anti-inflammatory transcription factors, shielding macrophages from infection-induced cell death. However, the immunometabolic function of itaconate was unexplored in HIV and cocaine-exposed microglia. We assessed the potential of 4-octyl-itaconate (4OI), a cell-penetrable ester form of itaconate known for its anti-inflammatory properties. When primary cortical cultures exposed to Tat and cocaine were treated with 4OI, microglial cell number increased and the morphological altercations induced by Tat and cocaine were reversed. Microglial cells also appeared more ramified, resembling the quiescent microglia. 4OI treatment inhibited secretion of the proinflammatory cytokines IL-1α, IL-1β, IL-6, and MIP1-α induced by Tat and cocaine. Transcriptome profiling determined that Nrf2 target genes were significantly activated in Tat and 4OI treated cultures relative to Tat alone. Further, genes associated with cytoskeleton dynamics in inflammatory microglia were downregulated by 4OI treatment. Together, the results strongly suggest 4-octyl-itaconate holds promise as a potential candidate for therapeutic development to treat HAND coupled with CUD comorbidities.
Collapse
Affiliation(s)
- B Celia Cui
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Marina Aksenova
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Aliaksandra Sikirzhytskaya
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Diana Odhiambo
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Elizaveta Korunova
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Vitali Sikirzhytski
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Hao Ji
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Diego Altomare
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Eugenia Broude
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Norma Frizzell
- Department of Pharmacology, Physiology and Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29208, USA
| | - Rosemarie Booze
- Department of Psychology, College of Arts and Sciences, University of South Carolina, Columbia, SC, 29208, USA
| | - Michael D Wyatt
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Michael Shtutman
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA.
| |
Collapse
|
5
|
Deshetty UM, Chatterjee N, Buch S, Periyasamy P. HIV-1 Tat-Mediated Human Müller Glial Cell Senescence Involves Endoplasmic Reticulum Stress and Dysregulated Autophagy. Viruses 2024; 16:903. [PMID: 38932195 PMCID: PMC11209317 DOI: 10.3390/v16060903] [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: 05/20/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Antiretroviral treatments have notably extended the lives of individuals with HIV and reduced the occurrence of comorbidities, including ocular manifestations. The involvement of endoplasmic reticulum (ER) stress in HIV-1 pathogenesis raises questions about its correlation with cellular senescence or its role in initiating senescent traits. This study investigated how ER stress and dysregulated autophagy impact cellular senescence triggered by HIV-1 Tat in the MIO-M1 cell line (human Müller glial cells). Cells exposed to HIV-1 Tat exhibited increased vimentin expression combined with markers of ER stress (BiP, p-eIF2α), autophagy (LC3, Beclin-1, p62), and the senescence marker p21 compared to control cells. Western blotting and staining techniques like SA-β-gal were employed to examine these markers. Additionally, treatments with ER stress inhibitor 4-PBA before HIV-1 Tat exposure led to a decreased expression of ER stress, senescence, and autophagy markers. Conversely, pre-treatment with the autophagy inhibitor 3-MA resulted in reduced autophagy and senescence markers but did not alter ER stress markers compared to control cells. The findings suggest a link between ER stress, dysregulated autophagy, and the initiation of a senescence phenotype in MIO-M1 cells induced by HIV-1 Tat exposure.
Collapse
Affiliation(s)
- Uma Maheswari Deshetty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA;
| | - Nivedita Chatterjee
- Vision Research Foundation, Sankara Netralaya, 18, College Road, Chennai 600006, India;
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA;
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA;
| |
Collapse
|
6
|
Yaseen MM, Abuharfeil NM, Darmani H. MDSC expansion during HIV infection: regulators, ART and immune reconstitution. Genes Immun 2024; 25:242-253. [PMID: 38605259 DOI: 10.1038/s41435-024-00272-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) become expanded in different pathological conditions including human immunodeficiency virus (HIV) infection and this may worsen the disease status and accelerate disease progression. In HIV infection, MDSCs suppress anti-HIV immune responses and hamper immune reconstitution. Understanding the factors and mechanisms of MDSC expansion during HIV infection is central to understanding the pathophysiology of HIV infection. This may pave the way to developing new therapeutic targets or strategies. In this work we addressed (i) the mechanisms that regulate MDSC expansion, (ii) the impact of antiretroviral therapy (ART) on the frequency of MDSCs during HIV infection; (iii) the impact of MDSCs on immune reconstitution during successful ART; and (iv) the potential of MDSCs as a therapeutic target.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
| | - Nizar Mohammad Abuharfeil
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Homa Darmani
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| |
Collapse
|
7
|
Kress TC, Ajala P, Jordan CR, Mintz J, MacArthur R, Kennard S, Antonova G, Belin de Chantemèle EJ. 12-week Dolutegravir treatment marginally reduces energy expenditure but does not increase body weight or alter vascular function in a murine model of Human Immunodeficiency Virus infection. Vascul Pharmacol 2024; 155:107288. [PMID: 38428626 PMCID: PMC11189738 DOI: 10.1016/j.vph.2024.107288] [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: 11/17/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024]
Abstract
Combination antiretroviral therapy (cART) has markedly increased life expectancy in people with HIV (PWH) but has also resulted in an increased prevalence of cardiometabolic disorders, whose etiopathology remains ill-defined. Notably, the respective contribution of cART and HIV-derived proteins to obesity and vascular alterations remain poorly understood. Therefore, we investigated the individual and combined effects of HIV-proteins and of the integrase strand transfer inhibitor Dolutegravir (DTG) on body composition and vascular reactivity. Male wildtype (WT) and HIV transgenic (Tg26) mice, received DTG or vehicle for 12 weeks. Viral proteins expression in Tg26 mice lowered fat mass, increased heat production, and induced a 2-fold increase in brown adipose tissue (BAT) uncoupling protein 1 (UCP1) expression. DTG increased the expression of markers of adipogenesis in adipocytes in culture, but also reduced heat production and BAT UCP1 and UCP3 expression in Tg26 mice. DTG increased food intake, fat percentage and protected from lean mass reduction in Tg26 mice only. However, DTG did not increase body weight in either WT or Tg26 mice. Viral protein expression reduced acetylcholine (endothelium)-mediated relaxation by 14% in mesenteric arteries preconstricted with phenylephrine. However, DTG did not impair nor improve endothelium-dependent relaxation. Together, these data indicate that DTG's effects on food intake, adipogenesis and energy expenditure are insufficient to increase body weight, even in the presence of HIV-proteins, suggesting that body weight gain in PWH involves additional factors likely including other cART components and pre-existing comorbidities. Moreover, these data rule out DTG as a source of vascular disorders in PWH.
Collapse
Affiliation(s)
- Taylor C Kress
- Vascular Biology Center, Medical College of Georgia at Augusta University,United States of America
| | - Priscilla Ajala
- Vascular Biology Center, Medical College of Georgia at Augusta University,United States of America
| | - Coleton R Jordan
- Vascular Biology Center, Medical College of Georgia at Augusta University,United States of America
| | - James Mintz
- Vascular Biology Center, Medical College of Georgia at Augusta University,United States of America
| | - Rodger MacArthur
- Department of Medicine, Medical College of Georgia at Augusta University, United States of America
| | - Simone Kennard
- Vascular Biology Center, Medical College of Georgia at Augusta University,United States of America
| | - Galina Antonova
- Vascular Biology Center, Medical College of Georgia at Augusta University,United States of America
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia at Augusta University,United States of America; Department of Medicine, Medical College of Georgia at Augusta University, United States of America.
| |
Collapse
|
8
|
Shmakova A, Tsimailo I, Kozhevnikova Y, Gérard L, Boutboul D, Oksenhendler E, Tuaillon E, Rivault A, Germini D, Vassetzky Y, Beaumelle B. HIV-1 Tat is present in the serum of people living with HIV-1 despite viral suppression. Int J Infect Dis 2024; 142:106994. [PMID: 38447753 DOI: 10.1016/j.ijid.2024.106994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024] Open
Abstract
OBJECTIVES Despite successful human immunodeficiency virus (HIV) control with combination antiretroviral therapy (cART), individuals with HIV still face health risks, including cancers, cardiovascular and neurocognitive diseases. An HIV protein, Tat, is potentially involved in these HIV-related diseases. Previous studies demonstrated circulating Tat in the blood of untreated people with HIV. Here, we measured Tat levels in the serum of cART-treated people with HIV to examine the effect of cART on Tat production. METHODS Serum samples from 63 HIV-positive and 20 HIV-seronegative individuals were analyzed using an ELISA assay that detected Tat concentrations above 2.5 ng/mL. RESULTS Among HIV-positive individuals, the Tat level ranged from 0 to 14 ng/mL. 25.4% (16 out of 63) exceeded the 2.5 ng/mL cut-off, with a median HIV Tat level of 4.518 [3.329-8.120] ng/mL. No correlation was revealed between Tat levels and CD4+ T cell counts, serum HIV RNA, p24 antigen, or anti-Tat levels. CONCLUSIONS Despite cART, circulating HIV Tat persists and may contribute to HIV-related diseases. This emphasizes the need for further research on the mechanisms of Tat action in non-infected cells where it can penetrate upon circulation in the blood.
Collapse
Affiliation(s)
- Anna Shmakova
- CNRS, UMR 9018, Université Paris-Saclay, Institut Gustave Roussy, Villejuif, 94800 France; Koltzov Institute of Developmental Biology, Moscow, 119334 Russia
| | - Ivan Tsimailo
- CNRS, UMR 9018, Université Paris-Saclay, Institut Gustave Roussy, Villejuif, 94800 France
| | - Yana Kozhevnikova
- CNRS, UMR 9018, Université Paris-Saclay, Institut Gustave Roussy, Villejuif, 94800 France
| | - Laurence Gérard
- Service d'Immunopathologie Clinique, Hôpital St Louis, APHP, Paris, 75012 France
| | - David Boutboul
- Service d'Immunopathologie Clinique, Hôpital St Louis, APHP, Paris, 75012 France
| | - Eric Oksenhendler
- Service d'Immunopathologie Clinique, Hôpital St Louis, APHP, Paris, 75012 France
| | - Edouard Tuaillon
- Pathogenesis and Control of Chronic and Emerging Infections, Montpellier University, INSERM U1058, Montpellier University Hospital, Montpellier, France
| | - Aurélie Rivault
- Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, CNRS UMR 9004, Montpellier, France
| | - Diego Germini
- CNRS, UMR 9018, Université Paris-Saclay, Institut Gustave Roussy, Villejuif, 94800 France
| | - Yegor Vassetzky
- CNRS, UMR 9018, Université Paris-Saclay, Institut Gustave Roussy, Villejuif, 94800 France; Koltzov Institute of Developmental Biology, Moscow, 119334 Russia.
| | - Bruno Beaumelle
- Institut de Recherche en Infectiologie de Montpellier, Université de Montpellier, CNRS UMR 9004, Montpellier, France
| |
Collapse
|
9
|
White CW, Platt S, Kilpatrick LE, Dale N, Abhayawardana RS, Dekkers S, Kindon ND, Kellam B, Stocks MJ, Pfleger KDG, Hill SJ. CXCL17 is an allosteric inhibitor of CXCR4 through a mechanism of action involving glycosaminoglycans. Sci Signal 2024; 17:eabl3758. [PMID: 38502733 PMCID: PMC7615768 DOI: 10.1126/scisignal.abl3758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 02/29/2024] [Indexed: 03/21/2024]
Abstract
CXCL17 is a chemokine principally expressed by mucosal tissues, where it facilitates chemotaxis of monocytes, dendritic cells, and macrophages and has antimicrobial properties. CXCL17 is also implicated in the pathology of inflammatory disorders and progression of several cancers, and its expression is increased during viral infections of the lung. However, the exact role of CXCL17 in health and disease requires further investigation, and there is a need for confirmed molecular targets mediating CXCL17 functional responses. Using a range of bioluminescence resonance energy transfer (BRET)-based assays, here we demonstrated that CXCL17 inhibited CXCR4-mediated signaling and ligand binding. Moreover, CXCL17 interacted with neuropillin-1, a VEGFR2 coreceptor. In addition, we found that CXCL17 only inhibited CXCR4 ligand binding in intact cells and demonstrated that this effect was mimicked by known glycosaminoglycan binders, surfen and protamine sulfate. Disruption of putative GAG binding domains in CXCL17 prevented CXCR4 binding. This indicated that CXCL17 inhibited CXCR4 by a mechanism of action that potentially required the presence of a glycosaminoglycan-containing accessory protein. Together, our results revealed that CXCL17 is an endogenous inhibitor of CXCR4 and represents the next step in our understanding of the function of CXCL17 and regulation of CXCR4 signaling.
Collapse
Affiliation(s)
- Carl W. White
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, Western Australia 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
- Dimerix Limited, Melbourne, Australia
| | - Simon Platt
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
| | - Laura E. Kilpatrick
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Natasha Dale
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, Western Australia 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Rekhati S. Abhayawardana
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, Western Australia 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Sebastian Dekkers
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Nicholas D Kindon
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Barrie Kellam
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Michael J Stocks
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Kevin D. G. Pfleger
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, Western Australia 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
- Dimerix Limited, Melbourne, Australia
| | - Stephen J. Hill
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
| |
Collapse
|
10
|
Cafaro A, Schietroma I, Sernicola L, Belli R, Campagna M, Mancini F, Farcomeni S, Pavone-Cossut MR, Borsetti A, Monini P, Ensoli B. Role of HIV-1 Tat Protein Interactions with Host Receptors in HIV Infection and Pathogenesis. Int J Mol Sci 2024; 25:1704. [PMID: 38338977 PMCID: PMC10855115 DOI: 10.3390/ijms25031704] [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: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Each time the virus starts a new round of expression/replication, even under effective antiretroviral therapy (ART), the transactivator of viral transcription Tat is one of the first HIV-1 protein to be produced, as it is strictly required for HIV replication and spreading. At this stage, most of the Tat protein exits infected cells, accumulates in the extracellular matrix and exerts profound effects on both the virus and neighbor cells, mostly of the innate and adaptive immune systems. Through these effects, extracellular Tat contributes to the acquisition of infection, spreading and progression to AIDS in untreated patients, or to non-AIDS co-morbidities in ART-treated individuals, who experience inflammation and immune activation despite virus suppression. Here, we review the role of extracellular Tat in both the virus life cycle and on cells of the innate and adaptive immune system, and we provide epidemiological and experimental evidence of the importance of targeting Tat to block residual HIV expression and replication. Finally, we briefly review vaccine studies showing that a therapeutic Tat vaccine intensifies ART, while its inclusion in a preventative vaccine may blunt escape from neutralizing antibodies and block early events in HIV acquisition.
Collapse
Affiliation(s)
- Aurelio Cafaro
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (L.S.); (R.B.); (M.C.); (F.M.); (S.F.); (M.R.P.-C.); (A.B.); (P.M.)
| | | | | | | | | | | | | | | | | | | | - Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.S.); (L.S.); (R.B.); (M.C.); (F.M.); (S.F.); (M.R.P.-C.); (A.B.); (P.M.)
| |
Collapse
|
11
|
Cui BC, Aksenova M, Sikirzhytskaya A, Odhiambo D, Korunova E, Sikirzhytski V, Ji H, Altomare D, Broude E, Frizzell N, Booze R, Wyatt MD, Shtutman M. Suppression of HIV and cocaine-induced neurotoxicity and inflammation by cell penetrable itaconate esters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.25.559154. [PMID: 37808776 PMCID: PMC10557618 DOI: 10.1101/2023.09.25.559154] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
HIV-associated neurological disorder (HAND) is a serious complication of HIV infection, marked by neurotoxicity induced by viral proteins like Tat. Substance abuse exacerbates neurocognitive impairment in people living with HIV. There is an urgent need for effective therapeutic strategies to combat HAND comorbid with Cocaine Use Disorder (CUD). Our analysis of the HIV and cocaine-induced transcriptomes in primary cortical cultures revealed a significant overexpression of the macrophage-specific gene, aconitate decarboxylase 1 (Acod1), caused by the combined insults of HIV and cocaine. ACOD1 protein converts the tricarboxylic acid intermediate cis-aconitate into itaconate during the activation of inflammation. The itaconate produced facilitates cytokine production and subsequently activates anti-inflammatory transcription factors, shielding macrophages from infection-induced cell death. While the role of itaconate' in limiting inflammation has been studied in peripheral macrophages, its immunometabolic function remains unexplored in HIV and cocaine-exposed microglia. We assessed in this model system the potential of 4-octyl-itaconate (4OI), a cell-penetrable esterified form of itaconate known for its potent anti-inflammatory properties and potential therapeutic applications. We administered 4OI to primary cortical cultures exposed to Tat and cocaine. 4OI treatment increased the number of microglial cells in both untreated and Tat±Cocaine-treated cultures and also reversed the morphological altercations induced by Tat and cocaine. In the presence of 4OI, microglial cells also appeared more ramified, resembling the quiescent microglia. Consistent with these results, 4OI treatment inhibited the secretion of the proinflammatory cytokines IL-1α, IL-1β, IL-6, and MIP1-α induced by Tat and cocaine. Transcriptome profiling further determined that Nrf2 target genes such as NAD(P)H quinone oxidoreductase 1 (Nqo1), Glutathione S-transferase Pi (Gstp1), and glutamate cysteine ligase catalytic (Gclc), were most significantly activated in Tat-4OI treated cultures, relative to Tat alone. Further, genes associated with cytoskeleton dynamics in inflammatory microglia were downregulated by 4OI treatment. Together, the results strongly suggest 4-octyl-itaconate holds promise as a potential candidate for therapeutic development aimed at addressing HAND coupled with CUD comorbidities.
Collapse
Affiliation(s)
- B. Celia Cui
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Marina Aksenova
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Aliaksandra Sikirzhytskaya
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Diana Odhiambo
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Elizaveta Korunova
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Vitali Sikirzhytski
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Hao Ji
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Diego Altomare
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Eugenia Broude
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Norma Frizzell
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | - Rosemarie Booze
- Department of Psychology, College of Arts and Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Michael D. Wyatt
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Michael Shtutman
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
12
|
Flounlacker KM, Hahn YK, Xu R, Simons CA, Tian T, Hauser KF, Knapp PE. Myelin regulatory factor is a target of individual and interactive effects of HIV-1 Tat and morphine in the striatum and pre-frontal cortex. J Neurovirol 2023; 29:15-26. [PMID: 36853588 DOI: 10.1007/s13365-022-01107-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 11/11/2022] [Accepted: 11/25/2022] [Indexed: 03/01/2023]
Abstract
HIV-associated neurocognitive disorders (HAND) remain pervasive even with increased efficacy/use of antiretroviral therapies. Opioid use/abuse among HIV + individuals is documented to exacerbate CNS deficits. White matter (WM) alterations, including myelin pallor, and volume/structural alterations detected by diffusion tensor imaging are common observations in HIV + individuals, and studies in non-human primates suggest that WM may harbor virus. Using transgenic mice that express the HIV-1 Tat protein, we examined in vivo effects of 2-6 weeks of Tat and morphine exposure on WM using genomic and biochemical methods. RNA sequencing of striatal tissue at 2 weeks revealed robust changes in mRNAs associated with oligodendrocyte precursor populations and myelin integrity, including those for transferrin, the atypical oligodendrocyte marker N-myc downstream regulated 1 (Ndrg1), and myelin regulatory factor (Myrf/Mrf), an oligodendrocyte-specific transcription factor with a significant role in oligodendrocyte differentiation/maturation. Western blots conducted after 6-weeks exposure in 3 brain regions (striatum, corpus callosum, pre-frontal cortex) revealed regional differences in the effect of Tat and morphine on Myrf levels, and on levels of myelin basic protein (MBP), whose transcription is regulated by Myrf. Responses included individual and interactive effects. Although baseline and post-treatment levels of Myrf and MBP differed between brain regions, post-treatment MBP levels in striatum and pre-frontal cortex were compatible with changes in Myrf activity. Additionally, the Myrf regulatory ubiquitin ligase Fbxw7 was identified as a novel target in our model. These results suggest that Myrf and Fbxw7 contribute to altered myelin gene regulation in HIV.
Collapse
Affiliation(s)
- Kelly M Flounlacker
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA.
| | - Yun Kyung Hahn
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA
| | - Ruqiang Xu
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA
| | - Chloe A Simons
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA
| | - Tao Tian
- Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Kurt F Hauser
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA.,Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.,The Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Pamela E Knapp
- Departments of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, 1101 E. Marshall St, Richmond, VA, 23298-0709, USA.,Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.,The Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| |
Collapse
|
13
|
Kalita E, Panda M, Rao A, Prajapati VK. Exploring the role of secretory proteins in the human infectious diseases diagnosis and therapeutics. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:231-269. [PMID: 36707203 DOI: 10.1016/bs.apcsb.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Secretory proteins are playing important role during the host-pathogen interaction to develop the infection or protection into the cell. Pathogens developing infectious disease to human being are taken up by host macrophages or number of immune cells, play an important role in physiological, developmental and immunological function. At the same time, infectious agents are also secreting various proteins to neutralize the resistance caused by host cells and also helping the pathogens to develop the infection. Secretory proteins (secretome) are only developed at the time of host-pathogen interaction, therefore they become very important to develop the targeted and potential therapeutic strategies. Pathogen specific secretory proteins released during interaction with host cell provide opportunity to develop point of care and rapid diagnostic kits. Proteins secreted by pathogens at the time of interaction with host cell have also been found as immunogenic in nature and numbers of vaccines have been developed to control the spread of human infectious diseases. This chapter highlights the importance of secretory proteins in the development of diagnostic and therapeutic strategies to fight against human infectious diseases.
Collapse
Affiliation(s)
- Elora Kalita
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Mamta Panda
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Abhishek Rao
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India.
| |
Collapse
|
14
|
Davis SE, Ferris MJ, Ananthan S, Augelli-Szafran CE, Zhu J. Novel Allosteric Modulator Southern Research Institute-32743 Reverses HIV-1 Transactivator of Transcription-Induced Increase in Dopamine Release in the Caudate Putamen of Inducible Transactivator of Transcription Transgenic Mice. J Pharmacol Exp Ther 2023; 384:306-314. [PMID: 36456195 PMCID: PMC9875314 DOI: 10.1124/jpet.122.001291] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/30/2022] [Accepted: 11/07/2022] [Indexed: 12/04/2022] Open
Abstract
Development of neurocognitive disorder in human immunodeficiency virus (HIV)-infected patients has been linked to dysregulation of dopamine by the HIV-1 transactivator of transcription (Tat) protein, a negative allosteric modulator of dopamine transporter (DAT). Using fast scan cyclic voltammetry, the present study determined the effects of in vivo Tat expression on dopamine release in the caudate putamen of inducible Tat transgenic (iTat-tg) mice and the impact of a novel DAT allosteric modulator, Southern Research Institute (SRI)-32743, on the Tat effect. We found that 7- or 14-day doxycycline (Dox)-induced Tat expression in iTat-tg mice resulted in a 2-fold increase in phasic but not tonic stimulated baseline dopamine release relative to saline control mice. To determine whether the Tat-induced increase in dopamine release is mediated by DAT regulation, we examined the effect of an in vitro applied DAT inhibitor, nomifensine, on the dopamine release. Nomifensine (1 nM-10 µM) concentration-dependently enhanced phasic stimulated dopamine release in both saline- and Dox-treated iTat-tg mice, while the magnitude of the nomifensine-mediated dopamine release was unchanged between saline and Dox treatment groups. A single systemic administration of SRI-32743 prior to animal sacrifice reversed the increased dopamine release in the baseline of phasic dopamine release and nomifensine-augmented dopamine levels in Dox-treated iTat-tg mice, while SRI-32743 alone did not alter baseline of dopamine release. These findings suggest that Tat expression induced an increase in extracellular dopamine levels by not only inhibiting DAT-mediated dopamine transport but also stimulating synaptic dopamine release. Thus, DAT allosteric modulators may serve as a potential therapeutic intervention for HIV infection-dysregulated dopamine system observed in HIV-1 positive individuals. SIGNIFICANCE STATEMENT: HIV infection-induced dysregulation of the dopaminergic system has been implicated in the development of neurocognitive impairments observed in HIV positive patients. Understanding the mechanisms underlying HIV-1 Tat protein-induced alteration of extracellular dopamine levels will provide insights into the development of molecules that can attenuate Tat interaction with targets in the dopaminergic system. Here, we determined whether Tat alters dopamine release and how the novel DAT allosteric modulator, SRI-32743, impacts dopamine neurotransmission to attenuate Tat-induced effects on extracellular dopamine dynamics.
Collapse
Affiliation(s)
- Sarah E Davis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina (S.E.D., J.Z.); Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina (M.M.F.); and Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, Alabama (S.A., C.E.A.)
| | - Mark J Ferris
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina (S.E.D., J.Z.); Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina (M.M.F.); and Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, Alabama (S.A., C.E.A.)
| | - Subramaniam Ananthan
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina (S.E.D., J.Z.); Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina (M.M.F.); and Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, Alabama (S.A., C.E.A.)
| | - Corinne E Augelli-Szafran
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina (S.E.D., J.Z.); Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina (M.M.F.); and Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, Alabama (S.A., C.E.A.)
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina (S.E.D., J.Z.); Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina (M.M.F.); and Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, Alabama (S.A., C.E.A.)
| |
Collapse
|
15
|
Ghanam RH, Eastep GN, Saad JS. Structural Insights into the Mechanism of HIV-1 Tat Secretion from the Plasma Membrane. J Mol Biol 2023; 435:167880. [PMID: 36370804 PMCID: PMC9822876 DOI: 10.1016/j.jmb.2022.167880] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/27/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) trans-activator of transcription (Tat) is a small, intrinsically disordered basic protein that plays diverse roles in the HIV-1 replication cycle, including promotion of efficient viral RNA transcription. Tat is released by infected cells and subsequently absorbed by healthy cells, thereby contributing to HIV-1 pathogenesis including HIV-associated neurocognitive disorder. It has been shown that, in HIV-1-infected primary CD4 T-cells, Tat accumulates at the plasma membrane (PM) for secretion, a mechanism mediated by phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). However, the structural basis for Tat interaction with the PM and thereby secretion is lacking. Herein, we employed NMR and biophysical methods to characterize Tat86 (86 amino acids) interactions with PI(4,5)P2 and lipid nanodiscs (NDs). Our data revealed that Arg49, Lys50 and Lys51 (RKK motif) constitute the PI(4,5)P2 binding site, that Tat86 interaction with lipid NDs is dependent on PI(4,5)P2 and phosphatidylserine (PS), and that the arginine-rich motif (RRQRRR) preferentially interacts with PS. Furthermore, we show that Trp11, previously implicated in Tat secretion, penetrates deeply in the membrane; substitution of Trp11 severely reduced Tat86 interaction with membranes. Deletion of the entire highly basic region and Trp11 completely abolished Tat86 binding to lipid NDs. Our data support a mechanism by which HIV-1 Tat secretion from the PM is mediated by a tripartite signal consisting of binding of the RKK motif to PI(4,5)P2, arginine-rich motif to PS, and penetration of Trp11 in the membrane. Altogether, these findings provide new insights into the molecular requirements for Tat binding to membranes during secretion.
Collapse
Affiliation(s)
- Ruba H Ghanam
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Gunnar N Eastep
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Jamil S Saad
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
16
|
Lien K, Mayer W, Herrera R, Padilla NT, Cai X, Lin V, Pholcharoenchit R, Palefsky J, Tugizov SM. HIV-1 Proteins gp120 and Tat Promote Epithelial-Mesenchymal Transition and Invasiveness of HPV-Positive and HPV-Negative Neoplastic Genital and Oral Epithelial Cells. Microbiol Spectr 2022; 10:e0362222. [PMID: 36314970 PMCID: PMC9770004 DOI: 10.1128/spectrum.03622-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
The incidence of human papillomavirus (HPV)-associated anogenital and oropharyngeal cancer in human immunodeficiency virus (HIV)-infected individuals is substantially higher than in HIV-uninfected individuals. HIV may also be a risk factor for the development of HPV-negative head and neck, liver, lung, and kidney cancer. However, the molecular mechanisms underlying HIV-1-associated increase of epithelial malignancies are not fully understood. Here, we showed that HPV-16-immortalized anal AKC-2 and cervical CaSki epithelial cells that undergo prolonged exposure to cell-free HIV-1 virions or HIV-1 viral proteins gp120 and tat respond with the epithelial-mesenchymal transition (EMT) and increased invasiveness. Similar responses were observed in HPV-16-infected SCC-47 and HPV-16-negative HSC-3 oral epithelial cancer cells that were cultured with these viral proteins. EMT induced by gp120 and tat led to detachment of poorly adherent cells from the culture substratum; these cells remained capable of reattachment, upon which they coexpressed both E-cadherin and vimentin, indicative of an intermediate stage of EMT. The reattached cells also expressed stem cell markers CD133 and CD44, which may play a critical role in cancer cell invasion and metastasis. Inhibition of transforming growth factor (TGF)-β1 and MAPK signaling and vimentin expression, and restoration of E-cadherin expression reduced HIV-induced EMT and the invasive activity of HPV-16-immortalized anal and cervical epithelial cells. Collectively, our results suggest that these approaches along with HIV viral suppression with antiretroviral therapy (ART) might be useful to limit the role of HIV-1 infection in the acceleration of HPV-associated or HPV-independent epithelial neoplasia. IMPORTANCE HPV-16-immortalized genital and oral epithelial cells and HPV-negative oral cancer cells that undergo prolonged contact with cell-free HIV-1 virions or with viral proteins gp120 and tat respond by becoming more invasive. EMT cells induced by HIV-1 in cultures of HPV-16-immortalized anal and cervical epithelial cells express the stem cell markers CD133 and CD44. These results suggest that the interaction of HIV-1 with neoplastic epithelial cells may lead to their de-differentiation into cancer stem cells that are resistant to apoptosis and anti-cancer drugs. Thus, this pathway may play a critical role in the development of invasive cancer. Inhibition of TGF-β1 and MAPK signaling and vimentin expression, and restoration of E-cadherin expression reduced HIV-induced EMT and the invasiveness of HPV-16-immortalized anal and cervical epithelial cells. Taken together, these results suggest that these approaches might be exploited to limit the role of HIV-1 infection in the acceleration of HPV-associated or HPV-independent epithelial neoplasia.
Collapse
Affiliation(s)
- Kathy Lien
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Wasima Mayer
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Rossana Herrera
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Nicole T. Padilla
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Xiaodan Cai
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Vicky Lin
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | | | - Joel Palefsky
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Sharof M. Tugizov
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| |
Collapse
|
17
|
Zhu J, Quizon PM, Wang Y, Adeniran CA, Strauss MJ, Jiménez-Torres AC, Patel P, Cirino TJ, Eans SO, Hammond HR, Deliscar LS, O'Hara P, Saini SK, Ofori E, Vekariya RH, Zhang S, Moukha-Chafiq O, Nguyen TH, Ananthan S, Augelli-Szafran CE, Zhan CG, McLaughlin JP. SRI-32743, a novel allosteric modulator, attenuates HIV-1 Tat protein-induced inhibition of the dopamine transporter and alleviates the potentiation of cocaine reward in HIV-1 Tat transgenic mice. Neuropharmacology 2022; 220:109239. [PMID: 36126727 DOI: 10.1016/j.neuropharm.2022.109239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 08/09/2022] [Accepted: 09/01/2022] [Indexed: 11/21/2022]
Abstract
Cocaine abuse increases the incidence of HIV-1-associated neurocognitive disorders. We have demonstrated that HIV-1 transactivator of transcription (Tat) allosterically modulates dopamine (DA) reuptake through the human DA transporter (hDAT), potentially contributing to Tat-induced cognitive impairment and potentiation of cocaine conditioned place preference (CPP). This study determined the effects of a novel allosteric modulator of DAT, SRI-32743, on the interactions of HIV-1 Tat, DA, cocaine, and [3H]WIN35,428 with hDAT in vitro. SRI-32743 (50 nM) attenuated Tat-induced inhibition of [3H]DA uptake and decreased the cocaine-mediated dissociation of [3H]WIN35,428 binding in CHO cells expressing hDAT, suggesting a SRI-32743-mediated allosteric modulation of the Tat-DAT interaction. In further in vivo studies utilizing doxycycline-inducible Tat transgenic (iTat-tg) mice, 14 days of Tat expression significantly reduced the recognition index by 31.7% in the final phase of novel object recognition (NOR) and potentiated cocaine-CPP 2.7-fold compared to responses of vehicle-treated control iTat-tg mice. The Tat-induced NOR deficits and potentiation of cocaine-CPP were not observed in saline-treated iTat-tg or doxycycline-treated G-tg (Tat-null) mice. Systemic administration (i.p.) of SRI-32743 prior to behavioral testing ameliorated Tat-induced impairment of NOR (at a dose of 10 mg/kg) and the Tat-induced potentiation of cocaine-CPP (at doses of 1 or 10 mg/kg). These findings demonstrate that Tat and cocaine interactions with DAT may be regulated by compounds interacting at the DAT allosteric modulatory sites, suggesting a potential therapeutic intervention for HIV-infected patients with concurrent cocaine abuse.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA.
| | - Pamela M Quizon
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Yingying Wang
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Charles A Adeniran
- Molecular Modeling and Biopharmaceutical Center, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Matthew J Strauss
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Ana C Jiménez-Torres
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Palak Patel
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Thomas J Cirino
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Shainnel O Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Haylee R Hammond
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Laure S Deliscar
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Priscilla O'Hara
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Surendra K Saini
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Edward Ofori
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Rakesh H Vekariya
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Sixue Zhang
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Omar Moukha-Chafiq
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Theresa H Nguyen
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Subramaniam Ananthan
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | | | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
18
|
Periyasamy P, Thangaraj A, Kannan M, Oladapo A, Buch S. The Epigenetic Role of miR-124 in HIV-1 Tat- and Cocaine-Mediated Microglial Activation. Int J Mol Sci 2022; 23:ijms232315017. [PMID: 36499350 PMCID: PMC9738975 DOI: 10.3390/ijms232315017] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
HIV-1 and drug abuse have been indissolubly allied as entwined epidemics. It is well-known that drug abuse can hasten the progression of HIV-1 and its consequences, especially in the brain, causing neuroinflammation. This study reports the combined effects of HIV-1 Transactivator of Transcription (Tat) protein and cocaine on miR-124 promoter DNA methylation and its role in microglial activation and neuroinflammation. The exposure of mouse primary microglial cells to HIV-1 Tat (25 ng/mL) and/or cocaine (10 μM) resulted in the significantly decreased expression of primary (pri)-miR-124-1, pri-miR-124-2, and mature miR-124 with a concomitant upregulation in DNMT1 expression as well as global DNA methylation. Our bisulfite-converted genomic DNA sequencing also revealed significant promoter DNA methylation in the pri-miR-124-1 and pri-miR-124-2 in HIV-1 Tat- and cocaine-exposed mouse primary microglial cells. We also found the increased expression of proinflammatory cytokines such as IL1β, IL6 and TNF in the mouse primary microglia exposed to HIV-1 Tat and cocaine correlated with microglial activation. Overall, our findings demonstrate that the exposure of mouse primary microglia to both HIV-1 Tat and cocaine could result in intensified microglial activation via the promoter DNA hypermethylation of miR-124, leading to the exacerbated release of proinflammatory cytokines, ultimately culminating in neuroinflammation.
Collapse
|
19
|
Natarajaseenivasan K, Garcia A, Velusamy P, Shanmughapriya S, Langford D. Citrate shuttling in astrocytes is required for processing cocaine-induced neuron-derived excess peroxidated fatty acids. iScience 2022; 25:105407. [PMID: 36389000 PMCID: PMC9646946 DOI: 10.1016/j.isci.2022.105407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 08/25/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Disturbances in lipid metabolism in the CNS contribute to neurodegeneration and cognitive impairments. Through tight metabolic coupling, astrocytes provide energy to neurons by delivering lactate and cholesterol and by taking up and processing neuron-derived peroxidated fatty acids (pFA). Disruption of CNS lipid homeostasis is observed in people who use cocaine and in several neurodegenerative disorders, including HIV. The brain's main source of energy is aerobic glycolysis, but numerous studies report a switch to β-oxidation of FAs in response to cocaine. Unlike astrocytes, in response to cocaine, neurons cannot efficiently consume excess pFAs for energy. Accumulation of pFA in neurons induces autophagy and release of pFA. Astrocytes endocytose the pFA for oxidation as an energy source. Our data show that blocking mitochondrial/cytosolic citrate transport reduces the neurotrophic capacity of astrocytes, leading to decreased neuronal fitness.
Collapse
Affiliation(s)
- Kalimuthusamy Natarajaseenivasan
- Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
- Department of Microbiology, Bharathidasan University, Tiruchirapalli, India
| | - Alvaro Garcia
- Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Prema Velusamy
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Santhanam Shanmughapriya
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Dianne Langford
- Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
20
|
Valyaeva AA, Tikhomirova MA, Potashnikova DM, Bogomazova AN, Snigiryova GP, Penin AA, Logacheva MD, Arifulin EA, Shmakova AA, Germini D, Kachalova AI, Saidova AA, Zharikova AA, Musinova YR, Mironov AA, Vassetzky YS, Sheval EV. Ectopic expression of HIV-1 Tat modifies gene expression in cultured B cells: implications for the development of B-cell lymphomas in HIV-1-infected patients. PeerJ 2022; 10:e13986. [PMID: 36275462 PMCID: PMC9586123 DOI: 10.7717/peerj.13986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/11/2022] [Indexed: 01/19/2023] Open
Abstract
An increased frequency of B-cell lymphomas is observed in human immunodeficiency virus-1 (HIV-1)-infected patients, although HIV-1 does not infect B cells. Development of B-cell lymphomas may be potentially due to the action of the HIV-1 Tat protein, which is actively released from HIV-1-infected cells, on uninfected B cells. The exact mechanism of Tat-induced B-cell lymphomagenesis has not yet been precisely identified. Here, we ectopically expressed either Tat or its TatC22G mutant devoid of transactivation activity in the RPMI 8866 lymphoblastoid B cell line and performed a genome-wide analysis of host gene expression. Stable expression of both Tat and TatC22G led to substantial modifications of the host transcriptome, including pronounced changes in antiviral response and cell cycle pathways. We did not find any strong action of Tat on cell proliferation, but during prolonged culturing, Tat-expressing cells were displaced by non-expressing cells, indicating that Tat expression slightly inhibited cell growth. We also found an increased frequency of chromosome aberrations in cells expressing Tat. Thus, Tat can modify gene expression in cultured B cells, leading to subtle modifications in cellular growth and chromosome instability, which could promote lymphomagenesis over time.
Collapse
Affiliation(s)
- Anna A. Valyaeva
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia,Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Maria A. Tikhomirova
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia,Koltzov Institute of Developmental Biology, Moscow, Russia
| | - Daria M. Potashnikova
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alexandra N. Bogomazova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow, Russia,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | | | | | - Maria D. Logacheva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia,Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Eugene A. Arifulin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Anna A. Shmakova
- Koltzov Institute of Developmental Biology, Moscow, Russia,UMR9018 (CNRS – Institut Gustave Roussy – Université Paris Saclay), Centre National de Recherche Scientifique, Villejuif, France, France
| | - Diego Germini
- UMR9018 (CNRS – Institut Gustave Roussy – Université Paris Saclay), Centre National de Recherche Scientifique, Villejuif, France, France
| | - Anastasia I. Kachalova
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Aleena A. Saidova
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Moscow, Russia
| | - Anastasia A. Zharikova
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Yana R. Musinova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia,Koltzov Institute of Developmental Biology, Moscow, Russia
| | - Andrey A. Mironov
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia,Institute for Information Transmission Problems, Moscow, Russia
| | - Yegor S. Vassetzky
- Koltzov Institute of Developmental Biology, Moscow, Russia,UMR9018 (CNRS – Institut Gustave Roussy – Université Paris Saclay), Centre National de Recherche Scientifique, Villejuif, France, France
| | - Eugene V. Sheval
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia,Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
21
|
T-cell evasion and invasion during HIV-1 infection: The role of HIV-1 Tat protein. Cell Immunol 2022; 377:104554. [DOI: 10.1016/j.cellimm.2022.104554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 12/22/2022]
|
22
|
Halcrow PW, Kumar N, Quansah DNK, Baral A, Liang B, Geiger JD. Endolysosome Iron Chelation Inhibits HIV-1 Protein-Induced Endolysosome De-Acidification-Induced Increases in Mitochondrial Fragmentation, Mitophagy, and Cell Death. Cells 2022; 11:1811. [PMID: 35681506 PMCID: PMC9180803 DOI: 10.3390/cells11111811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
People with human immunodeficiency virus-1 (PLWH) experience high rates of HIV-1-associated neurocognitive disorders (HANDs); clinical symptoms range from being asymptomatic to experiencing HIV-associated dementia. Antiretroviral therapies have effectively prolonged the life expectancy related to PLWH; however, the prevalence of HANDs has increased. Implicated in the pathogenesis of HANDs are two HIV-1 proteins, transactivator of transcription (Tat) and gp120; both are neurotoxic and damage mitochondria. The thread-like morphological features of functional mitochondria become fragmented when levels of reactive oxygen species (ROS) increase, and ROS can be generated via Fenton-like chemistry in the presence of ferrous iron (Fe2+). Endolysosomes are central to iron trafficking in cells and contain readily releasable Fe2+ stores. However, it is unclear whether the endolysosome store is sufficient to account for insult-induced increases in levels of ROS, mitochondrial fragmentation, autophagy, and cell death. Using U87MG astrocytoma and SH-SY5Y neuroblastoma cells, we determined that chloroquine (CQ), Tat, and gp120 all (1) de-acidified endolysosomes, (2) decreased endolysosome numbers and increased endolysosome sizes, (3) increased mitochondrial numbers (fragmentation), (4) increased autophagosome numbers, (5) increased autolysosome numbers, (6) increased mitochondrial fragments within endolysosomes, and (7) increased cell death. These effects were all blocked by the endolysosome-specific iron chelator deferoxamine (DFO). Thus, the endolysosome de-acidification-induced release of endolysosome Fe2+ is sufficient to account for inter-organellar signaling events and cell biology consequences of HIV-1 proteins, including mitochondrial fragmentation, autophagy, and cell death.
Collapse
Affiliation(s)
| | | | | | | | | | - Jonathan D. Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 504 Hamline Street, Room 110, Grand Forks, ND 58203, USA; (P.W.H.); (N.K.); (D.N.K.Q.); (A.B.); (B.L.)
| |
Collapse
|
23
|
Zhao X, Zhang F, Kandel SR, Brau F, He JJ. HIV Tat and cocaine interactively alter genome-wide DNA methylation and gene expression and exacerbate learning and memory impairments. Cell Rep 2022; 39:110765. [PMID: 35508123 PMCID: PMC9615417 DOI: 10.1016/j.celrep.2022.110765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/16/2022] [Accepted: 04/07/2022] [Indexed: 11/03/2022] Open
Abstract
Cocaine use is a major comorbidity of HIV-associated neurocognitive disorder (HAND). In this study, we show that cocaine exposure worsens the learning and memory of doxycycline-inducible and brain-specific HIV Tat transgenic mice (iTat) and results in 14,838 hypermethylated CpG-related differentially methylated regions (DMRs) and 15,800 hypomethylated CpG-related DMRs, which are linked to 52 down- and 127 upregulated genes, respectively, in the hippocampus of iTat mice. These genes are mostly enriched at the neuronal function-, cell morphology-, and synapse formation-related extracellular matrix (ECM) receptor-ligand interaction pathway and mostly impacted in microglia. The accompanying neuropathological changes include swollen dendritic spines, increased synaptophysin expression, and diminished glial activation. We also find that sex (female) and age additively worsen the behavioral and pathological changes. These findings together indicate that chronic cocaine and long-term Tat expression interactively contribute to HAND, likely involving changes of DNA methylation and ECM receptor-ligand interactions.
Collapse
Affiliation(s)
- Xiaojie Zhao
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA; Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA; School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Fan Zhang
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Suresh R Kandel
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA; Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Frédéric Brau
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis 06560, France
| | - Johnny J He
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA; Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA; School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL 60064, USA.
| |
Collapse
|
24
|
Kovacs L, Kress TC, Belin de Chantemèle EJ. HIV, Combination Antiretroviral Therapy, and Vascular Diseases in Men and Women. JACC Basic Transl Sci 2022; 7:410-421. [PMID: 35540101 PMCID: PMC9079796 DOI: 10.1016/j.jacbts.2021.10.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/01/2022]
Abstract
Thanks to the advent of combination antiretroviral therapy (cART), people living with human immunodeficiency virus (HIV) (PLWH) experienced a marked increase in life expectancy but are now at higher risk for cardiovascular disease (CVD), the current leading cause of death in PLWH on cART. Although HIV preponderantly affects men over women, manifestations of HIV-related CVD differ by sex with women experiencing greater risks than men. Despite extensive investigation, the etiopathology of CVD, notably the respective contribution of viral infection and cART, remain ill-defined. However, both viral infection and cART have been reported to contribute to endothelial dysfunction, the precursor and major cause of atherosclerosis-associated CVD, through mechanisms involving endothelial cell activation, inflammation, and oxidative stress, all leading to reduced nitric oxide bioavailability. Therefore, preserving endothelial function in PLWH on cART should be a main target to reduce CVD morbidity and mortality, notably in females.
Collapse
Key Words
- CVD, cardiovascular disease
- FMD, flow-mediated dilatation
- HF, heart failure
- HIV
- HIV, human immunodeficiency virus
- MI, myocardial infarction
- NO, nitric oxide
- PAD, peripheral artery disease
- PH, pulmonary hypertension
- PLWH, people living with HIV
- cART, combination antiretroviral therapy
- cIMT, carotid intima-media thickness
- combination antiretroviral therapy
- endothelial dysfunction
- sex differences
Collapse
Affiliation(s)
- Laszlo Kovacs
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Taylor C Kress
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA.,Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta Georgia, USA
| |
Collapse
|
25
|
Khan N, Halcrow PW, Afghah Z, Baral A, Geiger J, Chen X. HIV-1 Tat endocytosis and retention in endolysosomes affects HIV-1 Tat-induced LTR transactivation in astrocytes. FASEB J 2022; 36:e22184. [PMID: 35113458 PMCID: PMC9627655 DOI: 10.1096/fj.202101722r] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/13/2022] [Accepted: 01/18/2022] [Indexed: 11/11/2022]
Abstract
The presence of latent HIV-1 reservoirs in the periphery and brain represents a major obstacle to curing HIV-1 infection. As an essential protein for HIV-1 viral replication, HIV-1 Tat, mostly intracellular, has been implicated in latent HIV-1 infection. From HIV-1 infected cells, HIV-1 Tat is actively secreted and bystander cells uptake the released Tat whereupon it is endocytosed and internalized into endolysosomes. However, to activate the HIV-1 LTR promoter and increase HIV-1 replication, HIV-1 Tat must first escape from the endolysosomes and then enter the nucleus. Here, we tested the hypothesis that HIV-1 Tat can accumulate in endolysosomes and contribute to the activation of latent HIV-1 in astrocytes. Using U87MG astrocytoma cells expressing HIV-1 LTR-driven luciferase and primary human astrocytes we found that exogenous HIV-1 Tat enters endolysosomes, resides in endolysosomes for extended periods of time, and induces endolysosome de-acidification as well as enlargement. The weak base chloroquine promoted the release of HIV-1 Tat from endolysosomes and induced HIV-1 LTR transactivation. Similar results were observed by activating endolysosome Toll-like receptor 3 (TLR3) and TLR7/8. Conversely, pharmacological block of TLRs and knocking down expression levels of TLR3 and TLR7, but not TLR8, prevented endolysosome leakage and attenuated HIV-1 Tat-mediated HIV-1 LTR transactivation. Our findings suggest that HIV-1 Tat accumulation in endolysosomes may play an important role in controlling HIV-1 transactivation.
Collapse
Affiliation(s)
- Nabab Khan
- Department of Biomedical Sciences University of North Dakota School of Medicine and Health Sciences Grand Forks North Dakota USA
| | - Peter W. Halcrow
- Department of Biomedical Sciences University of North Dakota School of Medicine and Health Sciences Grand Forks North Dakota USA
| | - Zahra Afghah
- Department of Biomedical Sciences University of North Dakota School of Medicine and Health Sciences Grand Forks North Dakota USA
| | - Aparajita Baral
- Department of Biomedical Sciences University of North Dakota School of Medicine and Health Sciences Grand Forks North Dakota USA
| | - Jonathan D. Geiger
- Department of Biomedical Sciences University of North Dakota School of Medicine and Health Sciences Grand Forks North Dakota USA
| | - Xuesong Chen
- Department of Biomedical Sciences University of North Dakota School of Medicine and Health Sciences Grand Forks North Dakota USA
| |
Collapse
|
26
|
Davis S, Zhu J. Substance abuse and neurotransmission. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 93:403-441. [PMID: 35341573 PMCID: PMC9759822 DOI: 10.1016/bs.apha.2021.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The number of people who suffer from a substance abuse disorder has continued to rise over the last decade; particularly, the number of drug-related overdose deaths has sharply increased during the COVID-19 pandemic. Converging lines of clinical observations, supported by imaging and neuropsychological performance testing, have demonstrated that substance abuse-induced dysregulation of neurotransmissions in the brain is critical for development and expression of the addictive properties of abused substances. Recent scientific advances have allowed for better understanding of the neurobiological processes that mediates drugs of abuse and addiction. This chapter presents the past classic concepts and the recent advances in our knowledge about how cocaine, amphetamines, opioids, alcohol, and nicotine alter multiple neurotransmitter systems, which contribute to the behaviors associated with each drug. Additionally, we discuss the interactive effects of HIV-1 or COVID-19 and substance abuse on neurotransmission and neurobiological pathways. Finally, we introduce therapeutic strategies for development of pharmacotherapies for substance abuse disorders.
Collapse
Affiliation(s)
- Sarah Davis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States.
| |
Collapse
|
27
|
Wu S, Frank I, Derby N, Martinelli E, Cheng CY. HIV-1 Establishes a Sanctuary Site in the Testis by Permeating the BTB Through Changes in Cytoskeletal Organization. Endocrinology 2021; 162:6338140. [PMID: 34343260 PMCID: PMC8407494 DOI: 10.1210/endocr/bqab156] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Indexed: 11/19/2022]
Abstract
Studies suggest that HIV-1 invades the testis through initial permeation of the blood-testis barrier (BTB). The selectivity of the BTB to antiretroviral drugs makes this site a sanctuary for the virus. Little is known about how HIV-1 crosses the BTB and invades the testis. Herein, we used 2 approaches to examine the underlying mechanism(s) by which HIV-1 permeates the BTB and gains entry into the seminiferous epithelium. First, we examined if recombinant Tat protein was capable of perturbing the BTB and making the barrier leaky, using the primary rat Sertoli cell in vitro model that mimics the BTB in vivo. Second, we used HIV-1-infected Sup-T1 cells to investigate the activity of HIV-1 infection on cocultured Sertoli cells. Using both approaches, we found that the Sertoli cell tight junction permeability barrier was considerably perturbed and that HIV-1 effectively permeates the BTB by inducing actin-, microtubule-, vimentin-, and septin-based cytoskeletal changes in Sertoli cells. These studies suggest that HIV-1 directly perturbs BTB function, potentially through the activity of the Tat protein.
Collapse
Affiliation(s)
- Siwen Wu
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Ines Frank
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Nina Derby
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Elena Martinelli
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - C Yan Cheng
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| |
Collapse
|
28
|
Wang H, Huang W, Wang Y, Li W, Liu Q, Yin B, Liang L, Wang D, Guan X, Wang L. Enzyme Hinders HIV-1 Tat Viral Transport and Real-Time Measured with Nanopores. ACS Sens 2021; 6:3781-3788. [PMID: 34528798 DOI: 10.1021/acssensors.1c01717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
HIV-1 Tat protein, an intercellular transporter with a determinant function of delivering "information-rich" molecules in viral multiplication, was tryptic-hydrolyzed and real-time single molecule-monitored in a transmembrane pore. The electrokinetic studies revealed the catalytic and inhibitory effects on enzymatic digestion associated with Ca2+ and Cu2+ ions, respectively, in response to binding interactions with trypsin. Our strategy permits accurate and distinguishable sensing of Ca2+ and Cu2+ via an enzyme assay. In addition, considering the closer mimic of the real situation of HIV spread, measurements in the serum and on cells were also investigated. Transmembrane current measurements together with fluorescence microscopy imaging indicated the potential to perturb the Tat transport in the serum environment and on cells. Because the involved Tat proteolysis should prevent the occurrence of viral delivery, the presented method probably enables efficient hindrance to HIV-1 infection, in complementary to current traditional treatments.
Collapse
Affiliation(s)
- Han Wang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing 401147, China
| | - Wenli Huang
- Biotechnology and Nuclear Technology Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, China
| | - Yunjiao Wang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
| | - Wei Li
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
| | - Qianshan Liu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
| | - Bohua Yin
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Liyuan Liang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
| | - Deqiang Wang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
| | - Xiyun Guan
- Department of Chemistry, Illinois Institute of Technology, Chicago, Illinois 60616, United States
| | - Liang Wang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing 401147, China
| |
Collapse
|
29
|
Chronic Exposure to HIV-Derived Protein Tat Impairs Endothelial Function via Indirect Alteration in Fat Mass and Nox1-Mediated Mechanisms in Mice. Int J Mol Sci 2021; 22:ijms222010977. [PMID: 34681637 PMCID: PMC8540571 DOI: 10.3390/ijms222010977] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/03/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022] Open
Abstract
People living with human immunodeficiency virus (HIV) (PLWH) have increased risk for atherosclerosis-related cardiovascular disease (CVD), the main cause of death in this population. Notwithstanding, the mechanisms of HIV-associated vascular pathogenesis are not fully elucidated. Therefore, we sought to determine whether HIV-regulatory protein Tat mediates HIV-induced endothelial dysfunction via NADPH oxidase 1 (Nox1)-dependent mechanisms. Body weight, fat mass, leptin levels, expression of reactive oxygen species (ROS)-producing enzymes and vascular function were assessed in C57BL/6 male mice treated with Tat for 3 days and 4 weeks. Aortic rings and human endothelial cells were also treated with Tat for 2–24 h in ex vivo and in vitro settings. Chronic (4 weeks) but not acute (3 days and 2–24 h) treatment with Tat decreased body weight, fat mass, and leptin levels and increased the expression of Nox1 and its coactivator NADPH oxidase Activator 1 (NoxA1). This was associated with impaired endothelium-dependent vasorelaxation. Importantly, specific inhibition of Nox1 with GKT771 and chronic leptin infusion restored endothelial function in Tat-treated mice. These data rule out direct effects of HIV-Tat on endothelial function and imply the contribution of reductions in adipose mass and leptin production which likely explain upregulated expression of Nox1 and NoxA1. The Nox1 and leptin system may provide potential targets to improve vascular function in HIV infection-associated CVD.
Collapse
|
30
|
Niu F, Liao K, Hu G, Moidunny S, Roy S, Buch S. HIV Tat-Mediated Induction of Monocyte Transmigration Across the Blood-Brain Barrier: Role of Chemokine Receptor CXCR3. Front Cell Dev Biol 2021; 9:724970. [PMID: 34527676 PMCID: PMC8435688 DOI: 10.3389/fcell.2021.724970] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/10/2021] [Indexed: 01/17/2023] Open
Abstract
HIV trans-activator of transcription (Tat), one of the cytotoxic proteins secreted from HIV-infected cells, is also known to facilitate chemokine-mediated transmigration of monocytes into the brain leading, in turn, to neuroinflammation and thereby contributing to the development of HIV-associated neurocognitive disorders (HAND). The mechanism(s) underlying HIV Tat-mediated enhancement of monocyte transmigration, however, remain largely unknown. CXC chemokine receptor 3 (CXCR3) that is expressed by the peripheral monocytes is known to play a role in the monocyte influx and accumulation. In the present study, we demonstrate for the first time that exposure of human monocytes to HIV Tat protein resulted in upregulated expression of CXCR3 leading, in turn, to increased monocyte transmigration across the blood–brain barrier (BBB) both in the in vitro and in vivo model systems. This process involved activation of toll-like receptor 4 (TLR4), with downstream phosphorylation and activation of TANK-binding kinase 1 (TBK1), and subsequent phosphorylation and nuclear translocation of interferon regulatory factor 3 (IRF3), ultimately leading to enhanced expression of CXCR3 in human monocytes. These findings imply a novel molecular mechanism underlying HIV Tat-mediated increase of monocyte transmigration across the BBB, while also implicating a novel role of CXCR3-dependent monocyte transmigration in HIV Tat-mediated neuroinflammation.
Collapse
Affiliation(s)
- Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States.,Division of Clinical Research and Evaluative Sciences, Department of Medicine, Creighton University, Omaha, NE, United States
| | - Ke Liao
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shamsudheen Moidunny
- Department of Surgery, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Sabita Roy
- Department of Surgery, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
31
|
Human Immunodeficiency Virus (HIV) and Human Cytomegalovirus (HCMV) Coinfection of Infant Tonsil Epithelium May Synergistically Promote both HIV-1 and HCMV Spread and Infection. J Virol 2021; 95:e0092121. [PMID: 34232730 DOI: 10.1128/jvi.00921-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mother-to-child transmission (MTCT) of human immunodeficiency virus type 1 (HIV-1) and human cytomegalovirus (HCMV) may occur during pregnancy, labor, or breastfeeding. These viruses from amniotic fluid, cervicovaginal secretions, and breast milk may simultaneously interact with oropharyngeal and tonsil epithelia; however, the molecular mechanism of HIV-1 and HCMV cotransmission through the oral mucosa and its role in MTCT are poorly understood. To study the molecular mechanism of HIV-1 and HCMV MTCT via oral epithelium, we established polarized infant tonsil epithelial cells and polarized-oriented ex vivo tonsil tissue explants. Using these models, we showed that cell-free HIV-1 and its proteins gp120 and tat induce the disruption of tonsil epithelial tight junctions and increase paracellular permeability, which facilitates HCMV spread within the tonsil mucosa. Inhibition of HIV-1 gp120-induced upregulation of mitogen-activated protein kinase (MAPK) and NF-κB signaling in tonsil epithelial cells, reduces HCMV infection, indicating that HIV-1-activated MAPK and NF-κB signaling may play a critical role in HCMV infection of tonsil epithelium. HCMV infection of tonsil epithelial cells also leads to the disruption of tight junctions and increases paracellular permeability, facilitating HIV-1 paracellular spread into tonsil mucosa. HCMV-promoted paracellular spread of HIV-1 increases its accessibility to tonsil CD4 T lymphocytes, macrophages, and dendritic cells. HIV-1-enhanced HCMV paracellular spread and infection of epithelial cells subsequently leads to the spread of HCMV to tonsil macrophages and dendritic cells. Our findings revealed that HIV-1- and HCMV-induced disruption of infant tonsil epithelial tight junctions promotes MTCT of these viruses through tonsil mucosal epithelium, and therapeutic intervention for both HIV-1 and HCMV infection may substantially reduce their MTCT. IMPORTANCE Most HIV-1 and HCMV MTCT occurs in infancy, and the cotransmission of these viruses may occur via infant oropharyngeal and tonsil epithelia, which are the first biological barriers for viral pathogens. We have shown that HIV-1 and HCMV disrupt epithelial junctions, reducing the barrier functions of epithelia and thus allowing paracellular penetration of both viruses via mucosal epithelia. Subsequently, HCMV infects epithelial cells, macrophages, and dendritic cells, and HIV-1 infects CD4+ lymphocytes, macrophages, and dendritic cells. Infection of these cells in HCMV- and HIV-1-coinfected tonsil tissues is much higher than that by HCMV or HIV-1 infection alone, promoting their MTCT at its initial stages via infant oropharyngeal and tonsil epithelia.
Collapse
|
32
|
Ensoli B, Moretti S, Borsetti A, Maggiorella MT, Buttò S, Picconi O, Tripiciano A, Sgadari C, Monini P, Cafaro A. New insights into pathogenesis point to HIV-1 Tat as a key vaccine target. Arch Virol 2021; 166:2955-2974. [PMID: 34390393 PMCID: PMC8363864 DOI: 10.1007/s00705-021-05158-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023]
Abstract
Despite over 30 years of enormous effort and progress in the field, no preventative and/or therapeutic vaccines against human immunodeficiency virus (HIV) are available. Here, we briefly summarize the vaccine strategies and vaccine candidates that in recent years advanced to efficacy trials with mostly unsatisfactory results. Next, we discuss a novel and somewhat contrarian approach based on biological and epidemiological evidence, which led us to choose the HIV protein Tat for the development of preventive and therapeutic HIV vaccines. Toward this goal, we review here the role of Tat in the virus life cycle as well as experimental and epidemiological evidence supporting its key role in the natural history of HIV infection and comorbidities. We then discuss the preclinical and clinical development of a Tat therapeutic vaccine, which, by improving the functionality and homeostasis of the immune system and by reducing the viral reservoir in virologically suppressed vaccinees, helps to establish key determinants for intensification of combination antiretroviral therapy (cART) and a functional cure. Future developments and potential applications of the Tat therapeutic vaccine are also discussed, as well as the rationale for its use in preventative strategies. We hope this contribution will lead to a reconsideration of the current paradigms for the development of HIV/AIDS vaccines, with a focus on targeting of viral proteins with key roles in HIV pathogenesis.
Collapse
Affiliation(s)
- Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessandra Borsetti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Maria Teresa Maggiorella
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Stefano Buttò
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Orietta Picconi
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Antonella Tripiciano
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Cecilia Sgadari
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Paolo Monini
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Aurelio Cafaro
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
33
|
Blanco A, Mahajan T, Coronado RA, Ma K, Demma DR, Dar RD. Synergistic Chromatin-Modifying Treatments Reactivate Latent HIV and Decrease Migration of Multiple Host-Cell Types. Viruses 2021; 13:v13061097. [PMID: 34201394 PMCID: PMC8228244 DOI: 10.3390/v13061097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/29/2021] [Accepted: 06/02/2021] [Indexed: 11/29/2022] Open
Abstract
Upon infection of its host cell, human immunodeficiency virus (HIV) establishes a quiescent and non-productive state capable of spontaneous reactivation. Diverse cell types harboring the provirus form a latent reservoir, constituting a major obstacle to curing HIV. Here, we investigate the effects of latency reversal agents (LRAs) in an HIV-infected THP-1 monocyte cell line in vitro. We demonstrate that leading drug treatments synergize activation of the HIV long terminal repeat (LTR) promoter. We establish a latency model in THP-1 monocytes using a replication incompetent HIV reporter vector with functional Tat, and show that chromatin modifiers synergize with a potent transcriptional activator to enhance HIV reactivation, similar to T-cells. Furthermore, leading reactivation cocktails are shown to differentially affect latency reactivation and surface expression of chemokine receptor type 4 (CXCR4), leading to altered host cell migration. This study investigates the effect of chromatin-modifying LRA treatments on HIV latent reactivation and cell migration in monocytes. As previously reported in T-cells, epigenetic mechanisms in monocytes contribute to controlling the relationship between latent reactivation and cell migration. Ultimately, advanced “Shock and Kill” therapy needs to successfully target and account for all host cell types represented in a complex and composite latency milieu.
Collapse
Affiliation(s)
- Alexandra Blanco
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
| | - Tarun Mahajan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
| | - Robert A. Coronado
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
| | - Kelly Ma
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
| | - Dominic R. Demma
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
| | - Roy D. Dar
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (A.B.); (T.M.); (R.A.C.); (K.M.); (D.R.D.)
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Correspondence: ; Tel.: +1-(217)-265-0708
| |
Collapse
|
34
|
Yandrapally S, Mohareer K, Arekuti G, Vadankula GR, Banerjee S. HIV co-receptor-tropism: cellular and molecular events behind the enigmatic co-receptor switching. Crit Rev Microbiol 2021; 47:499-516. [PMID: 33900141 DOI: 10.1080/1040841x.2021.1902941] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recognition of cell-surface receptors and co-receptors is a crucial molecular event towards the establishment of HIV infection. HIV exists as several variants that differentially recognize the principal co-receptors, CCR5 and CXCR4, in different cell types, known as HIV co-receptor-tropism. The relative levels of these variants dynamically adjust to the changing host selection pressures to infect a vast repertoire of cells in a stage-specific manner. HIV infection sets in through immune cells such as dendritic cells, macrophages, and T-lymphocytes in the acute stage, while a wide range of other cells, including astrocytes, glial cells, B-lymphocytes, and epithelial cells, are infected during chronic stages. A change in tropism occurs during the transition from acute to a chronic phase, termed as co-receptor switching marked by a change in disease severity. The cellular and molecular events leading to co-receptor switching are poorly understood. This review aims to collate our present understanding of the dynamics of HIV co-receptor-tropism vis-à-vis host and viral factors, highlighting the cellular and molecular events involved therein. We present the possible correlations between virus entry, cell tropism, and co-receptor switching, speculating its consequences on disease progression, and proposing new scientific pursuits to help in an in-depth understanding of HIV biology.
Collapse
Affiliation(s)
| | | | - Geethika Arekuti
- Department of Biochemistry, University of Hyderabad, Hyderabad, India
| | | | | |
Collapse
|
35
|
Cirino TJ, McLaughlin JP. Mini review: Promotion of substance abuse in HIV patients: Biological mediation by HIV-1 Tat protein. Neurosci Lett 2021; 753:135877. [PMID: 33838257 DOI: 10.1016/j.neulet.2021.135877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 11/29/2022]
Abstract
Despite successful viral suppression by combinatorial anti-retroviral therapy, HIV infection continues to negatively impact the quality of life of patients by promoting neuropathy and HIV-Associated Neurocognitive Disorders (HAND), where substance use disorder (SUD) is highly comorbid and known to worsen health outcomes. While substance abuse exacerbates the progression of HIV, emerging evidence also suggests the virus may potentiate the rewarding effect of abused substances. As HIV does not infect neurons, these effects are theorized to be mediated by viral proteins. Key among these proteins are HIV-1 Tat, which can continue to be produced under viral suppression in patients. This review will recap the behavioral evidence for HIV-1 Tat mediation of a potentiation of cocaine, opioid and alcohol reward, and explore the neurochemical dysfunction associated by Tat as potential mechanisms underlying changes in reward. Targeting rampant oxidative stress, inflammation and excitotoxicity associated with HIV and Tat protein exposure may prove useful in combating persistent substance abuse comorbid with HIV in the clinic.
Collapse
Affiliation(s)
- Thomas J Cirino
- Department of Neurology, School of Medicine, University of California at San Francisco, San Francisco, CA, 94158, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
36
|
Tripiciano A, Picconi O, Moretti S, Sgadari C, Cafaro A, Francavilla V, Arancio A, Paniccia G, Campagna M, Pavone-Cossut MR, Sighinolfi L, Latini A, Mercurio VS, Pietro MD, Castelli F, Saracino A, Mussini C, Perri GD, Galli M, Nozza S, Ensoli F, Monini P, Ensoli B. Anti-Tat immunity defines CD4 + T-cell dynamics in people living with HIV on long-term cART. EBioMedicine 2021; 66:103306. [PMID: 33839064 PMCID: PMC8105504 DOI: 10.1016/j.ebiom.2021.103306] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Low-level HIV viremia originating from virus reactivation in HIV reservoirs is often present in cART treated individuals and represents a persisting source of immune stimulation associated with sub-optimal recovery of CD4+ T cells. The HIV-1 Tat protein is released in the extracellular milieu and activates immune cells and latent HIV, leading to virus production and release. However, the relation of anti-Tat immunity with residual viremia, persistent immune activation and CD4+ T-cell dynamics has not yet been defined. METHODS Volunteers enrolled in a 3-year longitudinal observational study were stratified by residual viremia, Tat serostatus and frequency of anti-Tat cellular immune responses. The impact of anti-Tat immunity on low-level viremia, persistent immune activation and CD4+ T-cell recovery was investigated by test for partitions, longitudinal regression analysis for repeated measures and generalized estimating equations. FINDINGS Anti-Tat immunity is significantly associated with higher nadir CD4+ T-cell numbers, control of low-level viremia and long-lasting CD4+ T-cell recovery, but not with decreased immune activation. In adjusted analysis, the extent of CD4+ T-cell restoration reflects the interplay among Tat immunity, residual viremia and immunological determinants including CD8+ T cells and B cells. Anti-Env immunity was not related to CD4+ T-cell recovery. INTERPRETATION Therapeutic approaches aiming at reinforcing anti-Tat immunity should be investigated to improve immune reconstitution in people living with HIV on long-term cART. TRIAL REGISTRATION ISS OBS T-002 ClinicalTrials.gov identifier: NCT01024556 FUNDING: Italian Ministry of Health, special project on the Development of a vaccine against HIV based on the Tat protein and Ricerca Corrente 2019/2020.
Collapse
Affiliation(s)
- Antonella Tripiciano
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Orietta Picconi
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Cecilia Sgadari
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Aurelio Cafaro
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Vittorio Francavilla
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Angela Arancio
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Giovanni Paniccia
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Massimo Campagna
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | | | - Laura Sighinolfi
- Unit of Infectious Diseases, University Hospital of Ferrara, Ferrara, Italy
| | - Alessandra Latini
- Unit of Dermatology and Sexually Transmitted Diseases, San Gallicano Institute - Istituti Fisioterapici Ospitalieri (IFO) IRCCS, Rome, Italy
| | - Vito S Mercurio
- Department of Infectious Diseases, S. Maria Goretti Hospital, Latina, Italy
| | - Massimo Di Pietro
- Unit of Infectious Diseases, S.M. Annunziata Hospital, Florence, Italy
| | - Francesco Castelli
- University Division of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili, Brescia, Italy
| | - Annalisa Saracino
- Division of Infectious Diseases, Policlinic Hospital, University of Bari, Bari, Italy
| | - Cristina Mussini
- Division of Infectious Diseases, University Policlinic of Modena, Modena, Italy
| | - Giovanni Di Perri
- Clinic of Infectious Diseases, Amedeo di Savoia University Hospital, Turin, Italy
| | - Massimo Galli
- Institute of Tropical and Infectious Diseases, L. Sacco University Hospital, Milan, Italy
| | - Silvia Nozza
- Division of Infectious Diseases, S. Raffaele University Hospital IRCCS, Milan, Italy
| | - Fabrizio Ensoli
- Pathology and Microbiology, San Gallicano Institute - (IFO) IRCCS, Rome, Italy
| | - Paolo Monini
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy.
| |
Collapse
|
37
|
Chen Q, Wu Y, Yu Y, Wei J, Huang W. Rho-kinase inhibitor hydroxyfasudil protects against HIV-1 Tat-induced dysfunction of tight junction and neprilysin/Aβ transfer receptor expression in mouse brain microvessels. Mol Cell Biochem 2021; 476:2159-2170. [PMID: 33548010 PMCID: PMC8057965 DOI: 10.1007/s11010-021-04056-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 01/11/2021] [Indexed: 11/28/2022]
Abstract
HIV-1 transactivator protein (Tat) induces tight junction (TJ) dysfunction and amyloid-beta (Aβ) clearance dysfunction, contributing to the development and progression of HIV-1-associated neurocognitive disorder (HAND). The Rho/ROCK signaling pathway has protective effects on neurodegenerative disease. However, the underlying mechanisms of whether Rho/ROCK protects against HIV-1 Tat-caused dysfunction of TJ and neprilysin (NEP)/Aβ transfer receptor expression have not been elucidated. C57BL/6 mice were administered sterile saline (i.p., 100 μL) or Rho-kinase inhibitor hydroxyfasudil (HF) (i.p., 10 mg/kg) or HIV-1 Tat (i.v., 100 μg/kg) or HF 30 min before being exposed to HIV-1 Tat once a day for seven consecutive days. Evans Blue (EB) leakage was detected via spectrophotometer and brain slides in mouse brains. The protein and mRNA levels of zonula occludens-1 (ZO-1), occludin, NEP, receptor for advanced glycation end products (RAGE), and low-density lipoprotein receptor-related protein 1 (LRP1) in mouse brain microvessels were, respectively, analyzed by Western blotting and quantitative real-time polymerase chain reaction (qRT-PCR) analyses. Exposure of the mice to HIV-1 Tat increased the amount of EB leakage, EB fluorescence intensity, blood–brain barrier (BBB) permeability, as well as the RAGE protein and mRNA levels, and decreased the protein and mRNA levels of ZO-1, occludin, NEP, and LRP1 in mouse brain microvessels. However, these effects were weakened by Rho-kinase inhibitor HF. Taken together, these results provide information that the Rho/ROCK signaling pathway is involved in HIV-1 Tat-induced dysfunction of TJ and NEP/Aβ transfer receptor expression in the C57BL/6 mouse brain. These findings shed some light on potentiality of inhibiting Rho/Rock signaling pathway in handling HAND.
Collapse
Affiliation(s)
- Qiangtang Chen
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China.,Department of Neurology, The First People's Hospital of Qinzhou, Qinzhou, 535099, Guangxi, China
| | - Yu Wu
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Yachun Yu
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Junxiang Wei
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Wen Huang
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
38
|
Akbay B, Germini D, Bissenbaev AK, Musinova YR, Sheval EV, Vassetzky Y, Dokudovskaya S. HIV-1 Tat Activates Akt/mTORC1 Pathway and AICDA Expression by Downregulating Its Transcriptional Inhibitors in B Cells. Int J Mol Sci 2021; 22:ijms22041588. [PMID: 33557396 PMCID: PMC7915967 DOI: 10.3390/ijms22041588] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/25/2021] [Accepted: 02/01/2021] [Indexed: 01/26/2023] Open
Abstract
HIV-1 infects T cells, but the most frequent AIDS-related lymphomas are of B-cell origin. Molecular mechanisms of HIV-1-induced oncogenic transformation of B cells remain largely unknown. HIV-1 Tat protein may participate in this process by penetrating and regulating gene expression in B cells. Both immune and cancer cells can reprogram communications between extracellular signals and intracellular signaling pathways via the Akt/mTORC1 pathway, which plays a key role in the cellular response to various stimuli including viral infection. Here, we investigated the role of HIV-1 Tat on the modulation of the Akt/mTORC1 pathway in B cells. We found that HIV-1 Tat activated the Akt/mTORC1 signaling pathway; this leads to aberrant activation of activation-induced cytidine deaminase (AICDA) due to inhibition of the AICDA transcriptional repressors c-Myb and E2F8. These perturbations may ultimately lead to an increased genomic instability and proliferation that might cause B cell malignancies.
Collapse
Affiliation(s)
- Burkitkan Akbay
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; (B.A.); (D.G.); (Y.V.)
- Department of Molecular Biology and Genetics, Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan;
| | - Diego Germini
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; (B.A.); (D.G.); (Y.V.)
| | - Amangeldy K. Bissenbaev
- Department of Molecular Biology and Genetics, Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan;
- Scientific Research Institute of Biology and Biotechnology Problems, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | - Yana R. Musinova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 119991 Moscow, Russia;
- Belozersky Institute of Physicochemical Biology, Moscow State University, 119899 Moscow, Russia;
| | - Evgeny V. Sheval
- Belozersky Institute of Physicochemical Biology, Moscow State University, 119899 Moscow, Russia;
| | - Yegor Vassetzky
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; (B.A.); (D.G.); (Y.V.)
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Svetlana Dokudovskaya
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; (B.A.); (D.G.); (Y.V.)
- Correspondence:
| |
Collapse
|
39
|
Li J, Huang J, He Y, Wang W, Leung CK, Zhang D, Zhang R, Wang S, Li Y, Liu L, Zeng X, Li Z. The protective effect of gastrodin against the synergistic effect of HIV-Tat protein and METH on the blood-brain barrier via glucose transporter 1 and glucose transporter 3. Toxicol Res (Camb) 2021; 10:91-101. [PMID: 33613977 DOI: 10.1093/toxres/tfaa102] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/04/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
Many individuals infected with human immunodeficiency virus (HIV) are also afflicted with HIV-associated neurocognitive disorders (HANDs). Methamphetamine (METH) abuse puts HIV-1 patients at risk for HANDs because METH and HIV-1 proteins, such as trans-activator of transcription (Tat), can synergistically damage the blood-brain barrier (BBB). However, the underlying mechanism of METH- and HIV-Tat-induced BBB damage remains unclear. In this study, male adult tree shrews and human brain capillary endothelial cells were treated with HIV-Tat, METH, and gastrodin. We used western blotting to examine the expressions of glucose transporters (GLUT1 and GLUT3), tight junctions, and junctional adhesion molecule A (JAMA) and to evaluate the damage and detect Evans blue (EB) and fluorescein sodium in the brain to assess BBB permeability to study the effect of METH and the HIV-1 Tat protein on BBB function in vitro and in vivo. The results showed that the group treated with Tat and METH experienced a significant change at the ultrastructural level of the tree shrew cerebral cortex, decreased protein levels of occluding, claudin-5, Zonula occludens 1 (ZO1), and JAMA in vitro and in vivo, and increased levels of EB and fluorescein sodium in the tree shrew cerebral cortex. The protein levels of GLUT1 and GLUT3 was downregulated in patients with Tat- and METH-induced BBB damage. Pretreatment with gastrodin significantly increased the levels of EB and fluorescein sodium in the tree shrew cerebral cortex and increased the expressions of occluding, ZO1, JAMA, and GLUT1 and GLUT. These results indicate that gastrodin may offer a potential therapeutic option for patients with HANDs.
Collapse
Affiliation(s)
- Juan Li
- School of Basic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China.,Center of Tree Shrew Germplasm Resources, Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Yunnan Innovation Team of Standardization and Application Research in Tree Shrew, Chinese Academy of Medical Science and Peking Union Medical College, 935 Jiaoling road,Yunnan 650531, China
| | - Jian Huang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China.,School of Forensic Medicine, Southern Medical University, 1838 Guangzhou Dadao Bei, Baiyun District, Guangzhou 510515, Guangdong, China
| | - Yongwang He
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Wenguang Wang
- Center of Tree Shrew Germplasm Resources, Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Yunnan Innovation Team of Standardization and Application Research in Tree Shrew, Chinese Academy of Medical Science and Peking Union Medical College, 935 Jiaoling road,Yunnan 650531, China
| | - Chi-Kwan Leung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,CUHK-SDU Joint Laboratory of Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Dongxian Zhang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Ruilin Zhang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Shangwen Wang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Yuanyuan Li
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Liu Liu
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Xiaofeng Zeng
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| | - Zhen Li
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong, Kunming, Yunnan 650500, China
| |
Collapse
|
40
|
HIV-1 Tat Protein Enters Dysfunctional Endothelial Cells via Integrins and Renders Them Permissive to Virus Replication. Int J Mol Sci 2020; 22:ijms22010317. [PMID: 33396807 PMCID: PMC7796023 DOI: 10.3390/ijms22010317] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/21/2020] [Accepted: 12/26/2020] [Indexed: 12/21/2022] Open
Abstract
Previous work has shown that the Tat protein of Human Immunodeficiency Virus (HIV)-1 is released by acutely infected cells in a biologically active form and enters dendritic cells upon the binding of its arginine-glycine-aspartic acid (RGD) domain to the α5β1, αvβ3, and αvβ5 integrins. The up-regulation/activation of these integrins occurs in endothelial cells exposed to inflammatory cytokines that are increased in HIV-infected individuals, leading to endothelial cell dysfunction. Here, we show that inflammatory cytokine-activated endothelial cells selectively bind and rapidly take up nano-micromolar concentrations of Tat, as determined by flow cytometry. Protein oxidation and low temperatures reduce Tat entry, suggesting a conformation- and energy-dependent process. Consistently, Tat entry is competed out by RGD-Tat peptides or integrin natural ligands, and it is blocked by anti-α5β1, -αvβ3, and -αvβ5 antibodies. Moreover, modelling–docking calculations identify a low-energy Tat-αvβ3 integrin complex in which Tat makes contacts with both the αv and β3 chains. It is noteworthy that internalized Tat induces HIV replication in inflammatory cytokine-treated, but not untreated, endothelial cells. Thus, endothelial cell dysfunction driven by inflammatory cytokines renders the vascular system a target of Tat, which makes endothelial cells permissive to HIV replication, adding a further layer of complexity to functionally cure and/or eradicate HIV infection.
Collapse
|
41
|
Human Immunodeficiency Virus Type 1 and Methamphetamine-Mediated Mitochondrial Damage and Neuronal Degeneration in Human Neurons. J Virol 2020; 94:JVI.00924-20. [PMID: 32796068 DOI: 10.1128/jvi.00924-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022] Open
Abstract
Methamphetamine, a potent psychostimulant, is a highly addictive drug commonly used by persons living with HIV (PLWH), and its use can result in cognitive impairment and memory deficits long after its use is discontinued. Although the mechanism(s) involved with persistent neurological deficits is not fully known, mitochondrial dysfunction is a key component in methamphetamine neuropathology. Specific mitochondrial autophagy (mitophagy) and mitochondrial fusion and fission are protective quality control mechanisms that can be dysregulated in HIV infection, and the use of methamphetamine can further negatively affect these protective cellular mechanisms. Here, we observed that treatment of human primary neurons (HPNs) with methamphetamine and HIV gp120 and Tat increase dynamin-related protein 1 (DRP1)-dependent mitochondrial fragmentation and neuronal degeneration. Methamphetamine and HIV proteins increased microtubule-associated protein 1 light chain 3 beta-II (LC3B-II) lipidation and induced sequestosome 1 (SQSTM1, p62) translocation to damaged mitochondria. Additionally, the combination inhibited autophagic flux, increased reactive oxygen species (ROS) production and mitochondrial damage, and reduced microtubule-associated protein 2 (MAP2) dendrites in human neurons. N-Acetylcysteine (NAC), a strong antioxidant and ROS scavenger, abrogated DRP1-dependent mitochondrial fragmentation and neurite degeneration. Thus, we show that methamphetamine combined with HIV proteins inhibits mitophagy and induces neuronal damage, and NAC reverses these deleterious effects on mitochondrial function.IMPORTANCE Human and animal studies show that HIV infection, combined with the long-term use of psychostimulants, increases neuronal stress and the occurrence of HIV-associated neurocognitive disorders (HAND). On the cellular level, mitochondrial function is critical for neuronal health. In this study, we show that in human primary neurons, the combination of HIV proteins and methamphetamine increases oxidative stress, DRP1-mediated mitochondrial fragmentation, and neuronal injury manifested by a reduction in neuronal network and connectivity. The use of NAC, a potent antioxidant, reversed the neurotoxic effects of HIV and methamphetamine, suggesting a novel approach to ameliorate the effects of HIV- and methamphetamine-associated cognitive deficits.
Collapse
|
42
|
Weber EA, Singh MV, Singh VB, Jackson JW, Ture SK, Suwunnakorn S, Morrell CN, Maggirwar SB. Novel Mechanism of Microvesicle Regulation by the Antiviral Protein Tetherin During HIV Infection. J Am Heart Assoc 2020; 9:e015998. [PMID: 32819189 PMCID: PMC7660781 DOI: 10.1161/jaha.120.015998] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023]
Abstract
Background Microvesicles are cell membrane-derived vesicles that have been shown to augment inflammation. Specifically, monocyte-derived microvesicles (MDMVs), which can express the coagulation protein tissue factor, contribute to thrombus formation and cardiovascular disease. People living with HIV experience higher prevalence of cardiovascular disease and also exhibit increased levels of plasma microvesicles. The process of microvesicle release has striking similarity to budding of enveloped viruses. The surface protein tetherin inhibits viral budding by physically tethering budding virus particles to cells. Hence, we investigated the role of tetherin in regulating the release of MDMVs during HIV infection. Methods and Results The plasma of aviremic HIV-infected individuals had increased levels of tissue factor + MDMVs, as measured by flow cytometry, and correlated to reduced tetherin expression on monocytes. Superresolution confocal and electron microscopy showed that tetherin localized at the site of budding MDMVs. Mechanistic studies revealed that the exposure of monocytes to HIV-encoded Tat triggered tetherin loss and subsequent rise in MDMV production. Overexpression of tetherin in monocytes led to morphologic changes in the pseudopodia directly underneath the MDMVs. Further, tetherin knockout mice demonstrated a higher number of circulating MDMVs and less time to bleeding cessation. Conclusions Our studies define a novel regulatory mechanism of MDMV release through tetherin and explore its contribution to the procoagulatory state that is frequently observed in people with HIV. Such insights could lead to improved therapies for individuals infected with HIV and also for those with cardiovascular disease.
Collapse
Affiliation(s)
- Emily A. Weber
- Department of Microbiology & ImmunologyUniversity of Rochester Medical CenterRochesterNY
| | - Meera V. Singh
- Department of Microbiology & ImmunologyUniversity of Rochester Medical CenterRochesterNY
| | - Vir B. Singh
- Department of Basic and Clinical SciencesAlbany College of Pharmacy and Health SciencesRochesterNY
| | - Joseph W. Jackson
- Department of Microbiology & ImmunologyUniversity of Rochester Medical CenterRochesterNY
| | - Sara K. Ture
- Aab Cardiovascular Research InstituteUniversity of Rochester Medical CenterRochesterNY
| | - Sumanun Suwunnakorn
- Department of Microbiology & ImmunologyUniversity of Rochester Medical CenterRochesterNY
| | - Craig N. Morrell
- Aab Cardiovascular Research InstituteUniversity of Rochester Medical CenterRochesterNY
| | - Sanjay B. Maggirwar
- Department of Microbiology & ImmunologyUniversity of Rochester Medical CenterRochesterNY
| |
Collapse
|
43
|
Gorska AM, Eugenin EA. The Glutamate System as a Crucial Regulator of CNS Toxicity and Survival of HIV Reservoirs. Front Cell Infect Microbiol 2020; 10:261. [PMID: 32670889 PMCID: PMC7326772 DOI: 10.3389/fcimb.2020.00261] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
Glutamate (Glu) is the most abundant excitatory neurotransmitter in the central nervous system (CNS). HIV-1 and viral proteins compromise glutamate synaptic transmission, resulting in poor cell-to-cell signaling and bystander toxicity. In this study, we identified that myeloid HIV-1-brain reservoirs survive in Glu and glutamine (Gln) as a major source of energy. Thus, we found a link between synaptic compromise, metabolomics of viral reservoirs, and viral persistence. In the current manuscript we will discuss all these interactions and the potential to achieve eradication and cure using this unique metabolic profile.
Collapse
Affiliation(s)
- Anna Maria Gorska
- Department of Neuroscience, Cell Biology, and Anatomy, The University of Texas Medical Branch, Galveston, TX, United States
| | - Eliseo A Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, The University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
44
|
Wu MM, Thayer SA. HIV Tat Protein Selectively Impairs CB 1 Receptor-Mediated Presynaptic Inhibition at Excitatory But Not Inhibitory Synapses. eNeuro 2020; 7:ENEURO.0119-20.2020. [PMID: 32471847 PMCID: PMC7307634 DOI: 10.1523/eneuro.0119-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/17/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the success of antiretroviral therapy in suppressing viral load, nearly half of the 37 million people infected with HIV experience cognitive and motor impairments, collectively classified as HIV-associated neurocognitive disorders (HAND). In the CNS, HIV-infected microglia release neurotoxic agents that act indirectly to elicit excitotoxic synaptic injury. HIV trans-activator of transcription (Tat) protein is one such neurotoxin that is thought to play a major role in the neuropathogenesis of HAND. The endocannabinoid (eCB) system provides on-demand neuroprotection against excitotoxicity, and exogenous cannabinoids attenuate neurotoxicity in animal models of HAND. Whether this neuroprotective system is altered in the presence of HIV is unknown. Here, we examined the effects of Tat on the eCB system in rat primary hippocampal cultures. Using whole-cell patch-clamp electrophysiology, we measured changes in retrograde eCB signaling following exposure to Tat. Treatment with Tat significantly reduced the magnitude of depolarization-induced suppression of excitation (DSE) in a graded manner over the course of 48 h. Interestingly, Tat did not alter this form of short-term synaptic plasticity at inhibitory terminals. The Tat-induced decrease in eCB signaling resulted from impaired CB1 receptor (CB1R)-mediated presynaptic inhibition of glutamate release. This novel loss-of-function was particularly dramatic for low-efficacy agonists such as the eCB 2-arachidonoylglycerol (2-AG) and Δ9-tetrahydrocannabinol (Δ9-THC), the main psychoactive ingredient in marijuana. Our observation that HIV Tat decreases CB1R function in vitro suggests that eCB-mediated neuroprotection may be reduced in vivo; this effect of Tat may contribute to synaptodendritic injury in HAND.
Collapse
Affiliation(s)
- Mariah M Wu
- Graduate Program in Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Stanley A Thayer
- Graduate Program in Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455
| |
Collapse
|
45
|
Aksenova M, Sybrandt J, Cui B, Sikirzhytski V, Ji H, Odhiambo D, Lucius MD, Turner JR, Broude E, Peña E, Lizarraga S, Zhu J, Safro I, Wyatt MD, Shtutman M. Inhibition of the Dead Box RNA Helicase 3 Prevents HIV-1 Tat and Cocaine-Induced Neurotoxicity by Targeting Microglia Activation. J Neuroimmune Pharmacol 2020; 15:209-223. [PMID: 31802418 PMCID: PMC8048136 DOI: 10.1007/s11481-019-09885-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 10/01/2019] [Indexed: 01/09/2023]
Abstract
HIV-1 Associated Neurocognitive Disorder (HAND) is a common and clinically detrimental complication of HIV infection. Viral proteins, including Tat, released from infected cells, cause neuronal toxicity. Substance abuse in HIV-infected patients greatly influences the severity of neuronal damage. To repurpose small molecule inhibitors for anti-HAND therapy, we employed MOLIERE, an AI-based literature mining system that we developed. All human genes were analyzed and prioritized by MOLIERE to find previously unknown targets connected to HAND. From the identified high priority genes, we narrowed the list to those with known small molecule ligands developed for other applications and lacking systemic toxicity in animal models. To validate the AI-based process, the selective small molecule inhibitor of DDX3 helicase activity, RK-33, was chosen and tested for neuroprotective activity. The compound, previously developed for cancer treatment, was tested for the prevention of combined neurotoxicity of HIV Tat and cocaine. Rodent cortical cultures were treated with 6 or 60 ng/ml of HIV Tat and 10 or 25 μM of cocaine, which caused substantial toxicity. RK-33 at doses as low as 1 μM greatly reduced the neurotoxicity of Tat and cocaine. Transcriptome analysis showed that most Tat-activated transcripts are microglia-specific genes and that RK-33 blocks their activation. Treatment with RK-33 inhibits the Tat and cocaine-dependent increase in the number and size of microglia and the proinflammatory cytokines IL-6, TNF-α, MCP-1/CCL2, MIP-2, IL-1α and IL-1β. These findings reveal that inhibition of DDX3 may have the potential to treat not only HAND but other neurodegenerative diseases. Graphical Abstract RK-33, selective inhibitor of Dead Box RNA helicase 3 (DDX3) protects neurons from combined Tat and cocaine neurotoxicity by inhibition of microglia activation and production of proinflammatory cytokines.
Collapse
Affiliation(s)
- Marina Aksenova
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter st, Columbia, SC, 29208, USA
| | - Justin Sybrandt
- School of Computing, Clemson University, 228 McAdams Hall, Clemson, SC, USA
| | - Biyun Cui
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter st, Columbia, SC, 29208, USA
| | - Vitali Sikirzhytski
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter st, Columbia, SC, 29208, USA
| | - Hao Ji
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter st, Columbia, SC, 29208, USA
| | - Diana Odhiambo
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter st, Columbia, SC, 29208, USA
| | - Matthew D Lucius
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter st, Columbia, SC, 29208, USA
| | - Jill R Turner
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter st, Columbia, SC, 29208, USA
- School of Computing, Clemson University, 228 McAdams Hall, Clemson, SC, USA
| | - Eugenia Broude
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter st, Columbia, SC, 29208, USA
| | - Edsel Peña
- Department of Statistics, College of Arts and Sciences, University of South Carolina, Columbia, SC, USA
| | - Sofia Lizarraga
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC, USA
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter st, Columbia, SC, 29208, USA
| | - Ilya Safro
- School of Computing, Clemson University, 228 McAdams Hall, Clemson, SC, USA.
| | - Michael D Wyatt
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter st, Columbia, SC, 29208, USA
| | - Michael Shtutman
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter st, Columbia, SC, 29208, USA.
| |
Collapse
|
46
|
Strauss M, O'Donovan B, Ma Y, Xiao Z, Lin S, Bardo MT, Ortinski PI, McLaughlin JP, Zhu J. [ 3H]Dopamine Uptake through the Dopamine and Norepinephrine Transporters is Decreased in the Prefrontal Cortex of Transgenic Mice Expressing HIV-1 Transactivator of Transcription Protein. J Pharmacol Exp Ther 2020; 374:241-251. [PMID: 32461322 DOI: 10.1124/jpet.120.266023] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/21/2020] [Indexed: 01/16/2023] Open
Abstract
Dysregulation of dopamine neurotransmission has been linked to the development of human immunodeficiency virus (HIV)-associated neurocognitive disorder (HAND). To investigate the mechanisms underlying this phenomenon, this study used an inducible HIV-1 transactivator of transcription (Tat) transgenic (iTat-tg) mouse model, which demonstrates brain-specific Tat expression induced by administration of doxycycline. We found that induction of Tat expression in the iTat-tg mice for either 7 or 14 days resulted in a decrease (∼30%) in the V max of [3H]dopamine uptake via both the dopamine transporter (DAT) and norepinephrine transporter (NET) in the prefrontal cortex (PFC), which was comparable to the magnitude (∼35%) of the decrease in B max for [3H]WIN 35,428 and [3H]nisoxetine binding to DAT and NET, respectively. The decreased V max was not accompanied by a reduction of total or plasma membrane expression of DAT and NET. Consistent with the decreased V max for DAT and NET in the PFC, the current study also found an increase in the tissue content of DA and dihydroxyphenylacetic acid in the PFC of iTat-tg mice after 7 days' administration of doxycycline. Electrophysiological recordings in layer V pyramidal neurons of the prelimbic cortex from iTat-tg mice found a significant reduction in action potential firing, which was not sensitive to selective inhibitors for DAT and NET, respectively. These findings provide a molecular basis for using the iTat-tg mouse model in the studies of NeuroHIV. Determining the mechanistic basis underlying the interaction between Tat and DAT/NET may reveal novel therapeutic possibilities for preventing the increase in comorbid conditions as well as HAND. SIGNIFICANCE STATEMENT: Human immunodeficiency virus (HIV)-1 infection disrupts dopaminergic neurotransmission, leading to HIV-associated neurocognitive disorders (HANDs). Based on our in vitro and in vivo studies, dopamine uptake via both dopamine and norepinephrine transporters is decreased in the prefrontal cortex of HIV-1 Tat transgenic mice, which is consistent with the increased dopamine and dihydroxyphenylacetic acid contents in this brain region. Thus, these plasma membrane transporters are an important potential target for therapeutic intervention for patients with HAND.
Collapse
Affiliation(s)
- Matthew Strauss
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Bernadette O'Donovan
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Yizhi Ma
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Ziyu Xiao
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Steven Lin
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Michael T Bardo
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Pavel I Ortinski
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Jay P McLaughlin
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (M.S., Y.M., Z.X., S.L., J.Z.) and Department of Physiology, Pharmacology and Neuroscience, School of Medicine (B.O.), University of South Carolina, Columbia, South Carolina; Departments of Psychology (M.B.) and Neuroscience (P.O.), University of Kentucky, Lexington, Kentucky; and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.)
| |
Collapse
|
47
|
Sil S, Hu G, Liao K, Niu F, Callen S, Periyasamy P, Fox HS, Buch S. HIV-1 Tat-mediated astrocytic amyloidosis involves the HIF-1α/lncRNA BACE1-AS axis. PLoS Biol 2020; 18:e3000660. [PMID: 32453744 PMCID: PMC7274476 DOI: 10.1371/journal.pbio.3000660] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 06/05/2020] [Accepted: 04/28/2020] [Indexed: 12/22/2022] Open
Abstract
Increased life expectancy of patients diagnosed with HIV in the current era of antiretroviral therapy is unfortunately accompanied with the prevalence of HIV-associated neurocognitive disorders (HANDs) and risk of comorbidities such as Alzheimer-like pathology. HIV-1 transactivator of transcription (Tat) protein has been shown to induce the production of toxic neuronal amyloid protein and also enhance neurotoxicity. The contribution of astrocytes in Tat-mediated amyloidosis remains an enigma. We report here, in simian immunodeficiency virus (SIV)+ rhesus macaques and patients diagnosed with HIV, brain region-specific up-regulation of amyloid precursor protein (APP) and Aβ (40 and 42) in astrocytes. In addition, we find increased expression of β-site cleaving enzyme (BACE1), APP, and Aβ in human primary astrocytes (HPAs) exposed to Tat. Mechanisms involved up-regulation of hypoxia-inducible factor (HIF-1α), its translocation and binding to the long noncoding RNA (lncRNA) BACE1-antisense transcript (BACE1-AS), resulting, in turn, in the formation of the BACE1-AS/BACE1 RNA complex, subsequently leading to increased BACE1 protein, and activity and generation of Aβ-42. Gene silencing approaches confirmed the regulatory role of HIF-1α in BACE1-AS/BACE1 in Tat-mediated amyloidosis. This is the first report implicating the role of the HIF-1α/lncRNABACE1-AS/BACE1 axis in Tat-mediated induction of astrocytic amyloidosis, which could be targeted as adjunctive therapies for HAND-associated Alzheimer-like comorbidity.
Collapse
Affiliation(s)
- Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ke Liao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Shannon Callen
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Howard S. Fox
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
48
|
Bahraoui E, Serrero M, Planès R. HIV-1 Tat - TLR4/MD2 interaction drives the expression of IDO-1 in monocytes derived dendritic cells through NF-κB dependent pathway. Sci Rep 2020; 10:8177. [PMID: 32424165 PMCID: PMC7235218 DOI: 10.1038/s41598-020-64847-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 02/06/2020] [Indexed: 12/31/2022] Open
Abstract
In the present study we showed that HIV-1 Tat protein stimulated the expression of Indoleamine 2,3 dioxygenase (IDO) -1 in human monocytes derived dendritic cells (MoDC) but not IDO-2 by acting directly at the cell membrane level. This induction of IDO-1 is dependent on the secondary structure of Tat protein, since stimulation with a chemically oxidized Tat protein loses its capacity to induce the production of IDO-1. Among the variety of candidate receptors described for Tat, we demonstrated that Tat protein interacted physically with TLR4/MD2 complex. Strikingly, blockade of Tat-TLR4 interaction by anti-TLR4 antibodies (clone HTA125), LPS-RS, a known TLR4 antagonist, or by soluble recombinant TLR4/MD2 complex inhibited strongly or totally the capacity of Tat to induce IDO-1 in MoDC while such treatments had no effect on IFN-γ-induced IDO-1. Furthermore, we showed that the activation of the transcription factor NF-κB by Tat is essential for the production of IDO-1 by human MoDC. Indeed, Tat activated NF-κB pathway in MoDC as demonstrated by the phosphorylation of p65 in Tat-treated MoDC. Further, we demonstrate that the stimulation of IDO-1 by Tat or by IFN-γ was totally or partially inhibited in the presence of NF-κB inhibitor respectively. These results suggest that Tat and IFN-γ act probably by two distinct mechanisms to induce the production of IDO-1. Our results clearly demonstrated that, although TLR4 pathway is necessary for Tat-induced IDO-1 in MoDC, it seems not to be sufficient since stable transfection of a functional TLR4/MD2 pathway in HEK or HeLa cell lines which are endogenously defectives for TLR4, did not restore the capacity of Tat to induce IDO-1 while IFN-γ treatment induces IDO-1 in HeLa cells independently of TLR4 pathway. These results suggest the involvement of additional stimuli in addition to TLR4 pathway which remain to be identified. Altogether our results demonstrated that, in human MoDC, HIV-1 Tat protein induced IDO-1 expression and activity in a NF-κB dependent-manner by recruiting TLR4 pathway.
Collapse
Affiliation(s)
- Elmostafa Bahraoui
- INSERM, U1043, CPTP, CHU purpan, Toulouse, France. .,CNRS, U5282 CPTP, CHU purpan, Toulouse, France. .,Université Paul Sabatier, CPTP, CHU purpan, Toulouse, France.
| | - Manutea Serrero
- INSERM, U1043, CPTP, CHU purpan, Toulouse, France.,CNRS, U5282 CPTP, CHU purpan, Toulouse, France.,Université Paul Sabatier, CPTP, CHU purpan, Toulouse, France
| | - Rémi Planès
- INSERM, U1043, CPTP, CHU purpan, Toulouse, France. .,CNRS, U5282 CPTP, CHU purpan, Toulouse, France. .,Université Paul Sabatier, CPTP, CHU purpan, Toulouse, France.
| |
Collapse
|
49
|
Jin H, Li D, Lin MH, Li L, Harrich D. Tat-Based Therapies as an Adjuvant for an HIV-1 Functional Cure. Viruses 2020; 12:v12040415. [PMID: 32276443 PMCID: PMC7232260 DOI: 10.3390/v12040415] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/31/2020] [Accepted: 04/04/2020] [Indexed: 12/18/2022] Open
Abstract
The human immunodeficiency virus type 1 (HIV) establishes a chronic infection that can be well controlled, but not cured, by combined antiretroviral therapy (cART). Interventions have been explored to accomplish a functional cure, meaning that a patient remains infected but HIV is undetectable in the blood, with the aim of allowing patients to live without cART. Tat, the viral transactivator of transcription protein, plays a critical role in controlling HIV transcription, latency, and viral rebound following the interruption of cART treatment. Therefore, a logical approach for controlling HIV would be to block Tat. Tackling Tat with inhibitors has been a difficult task, but some recent discoveries hold promise. Two anti-HIV proteins, Nullbasic (a mutant of Tat) and HT1 (a fusion of HEXIM1 and Tat functional domains) inhibit viral transcription by interfering with the interaction of Tat and cellular factors. Two small molecules, didehydro-cortistatin A (dCA) and triptolide, inhibit Tat by different mechanisms: dCA through direct binding and triptolide through enhanced proteasomal degradation. Finally, two Tat-based vaccines under development elicit Tat-neutralizing antibodies. These vaccines have increased the levels of CD4+ cells and reduced viral loads in HIV-infected people, suggesting that the new vaccines are therapeutic. This review summarizes recent developments of anti-Tat agents and how they could contribute to a functional cure for HIV.
Collapse
Affiliation(s)
- Hongping Jin
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (H.J.); (D.L.); (M.-H.L.)
| | - Dongsheng Li
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (H.J.); (D.L.); (M.-H.L.)
| | - Min-Hsuan Lin
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (H.J.); (D.L.); (M.-H.L.)
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia;
| | - David Harrich
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (H.J.); (D.L.); (M.-H.L.)
- Correspondence: ; Tel.: +617-3845-3679
| |
Collapse
|
50
|
Ajasin D, Eugenin EA. HIV-1 Tat: Role in Bystander Toxicity. Front Cell Infect Microbiol 2020; 10:61. [PMID: 32158701 PMCID: PMC7052126 DOI: 10.3389/fcimb.2020.00061] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/06/2020] [Indexed: 12/21/2022] Open
Abstract
HIV Tat protein is a critical protein that plays multiple roles in HIV pathogenesis. While its role as the transactivator of HIV transcription is well-established, other non-viral replication-associated functions have been described in several HIV-comorbidities even in the current antiretroviral therapy (ART) era. HIV Tat protein is produced and released into the extracellular space from cells with active HIV replication or from latently HIV-infected cells into neighboring uninfected cells even in the absence of active HIV replication and viral production due to effective ART. Neighboring uninfected and HIV-infected cells can take up the released Tat resulting in the upregulation of inflammatory genes and activation of pathways that leads to cytotoxicity observed in several comorbidities such as HIV associated neurocognitive disorder (HAND), HIV associated cardiovascular impairment, and accelerated aging. Thus, understanding how Tat modulates host and viral response is important in designing novel therapeutic approaches to target the chronic inflammatory effects of soluble viral proteins in HIV infection.
Collapse
Affiliation(s)
- David Ajasin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, United States
| | - Eliseo A Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|