51
|
HIV-1 Persistence and Chronic Induction of Innate Immune Responses in Macrophages. Viruses 2020; 12:v12070711. [PMID: 32630058 PMCID: PMC7412260 DOI: 10.3390/v12070711] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
A hallmark of HIV-1 infection is chronic inflammation, which plays a significant role in disease pathogenesis. Acute HIV infection induces robust inflammatory responses, which are insufficient to prevent or eliminate virus in mucosal tissues. While establishment of viral set-point is coincident with downregulation of acute innate responses, systemic inflammatory responses persist during the course of chronic HIV infection. Since the introduction of combination antiviral therapy (cART), most HIV-1+ individuals can suppress viremia under detection levels for decades. However, chronic immune activation persists and has been postulated to cause HIV associated non-AIDS complications (HANA). Importantly, inflammatory cytokines and activation markers associated with macrophages are strongly and selectively correlated with the incidence of HIV-associated neurocognitive disorder (HAND), cardiovascular dysfunctions (CVD) and other HANA conditions. In this review, we discuss the roles of macrophages in facilitating viral persistence and contributing to generation of persistent inflammatory responses.
Collapse
|
52
|
McHugh D, Myburgh R, Caduff N, Spohn M, Kok YL, Keller CW, Murer A, Chatterjee B, Rühl J, Engelmann C, Chijioke O, Quast I, Shilaih M, Strouvelle VP, Neumann K, Menter T, Dirnhofer S, Lam JK, Hui KF, Bredl S, Schlaepfer E, Sorce S, Zbinden A, Capaul R, Lünemann JD, Aguzzi A, Chiang AK, Kempf W, Trkola A, Metzner KJ, Manz MG, Grundhoff A, Speck RF, Münz C. EBV renders B cells susceptible to HIV-1 in humanized mice. Life Sci Alliance 2020; 3:3/8/e202000640. [PMID: 32576602 PMCID: PMC7335381 DOI: 10.26508/lsa.202000640] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022] Open
Abstract
HIV and EBV are human pathogens that cause a considerable burden to worldwide health. In combination, these viruses are linked to AIDS-associated lymphomas. We found that EBV, which transforms B cells, renders them susceptible to HIV-1 infection in a CXCR4 and CD4-dependent manner in vitro and that CXCR4-tropic HIV-1 integrates into the genome of these B cells with the same molecular profile as in autologous CD4+ T cells. In addition, we established a humanized mouse model to investigate the in vivo interactions of EBV and HIV-1 upon coinfection. The respective mice that reconstitute human immune system components upon transplantation with CD34+ human hematopoietic progenitor cells could recapitulate aspects of EBV and HIV immunobiology observed in dual-infected patients. Upon coinfection of humanized mice, EBV/HIV dual-infected B cells could be detected, but were susceptible to CD8+ T-cell-mediated immune control.
Collapse
Affiliation(s)
- Donal McHugh
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Renier Myburgh
- Department of Medical Oncology and Hematology, University and University Hospital of Zürich, Zürich, Switzerland
| | - Nicole Caduff
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Michael Spohn
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Yik Lim Kok
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland.,Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Christian W Keller
- Neuroinflammation, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Anita Murer
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Bithi Chatterjee
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Julia Rühl
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Christine Engelmann
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Obinna Chijioke
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland.,Institute of Pathology and Medical Genetics, University Hospital of Basel, Basel, Switzerland
| | - Isaak Quast
- Neuroinflammation, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Mohaned Shilaih
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland
| | - Victoria P Strouvelle
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland.,Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Kathrin Neumann
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland
| | - Thomas Menter
- Institute of Pathology and Medical Genetics, University Hospital of Basel, Basel, Switzerland
| | - Stephan Dirnhofer
- Institute of Pathology and Medical Genetics, University Hospital of Basel, Basel, Switzerland
| | - Janice Kp Lam
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kwai F Hui
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Simon Bredl
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland
| | - Erika Schlaepfer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland
| | - Silvia Sorce
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Andrea Zbinden
- Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Riccarda Capaul
- Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Jan D Lünemann
- Neuroinflammation, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Alan Ks Chiang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Werner Kempf
- Kempf und Pfaltz Histologische Diagnostik AG, Zürich, Switzerland
| | - Alexandra Trkola
- Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Karin J Metzner
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland.,Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University and University Hospital of Zürich, Zürich, Switzerland
| | - Adam Grundhoff
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Roberto F Speck
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
53
|
Khan T, Mayuresh Patkar M, Momin M, Omri A. Macrophage targeted nanocarrier delivery systems in HIV therapeutics. Expert Opin Drug Deliv 2020; 17:903-918. [PMID: 32347124 DOI: 10.1080/17425247.2020.1762565] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Human immunodeficiency virus (HIV) targets and modulates the immune system increasing the risk of other associated infections. Highly active antiretroviral therapy (HAART) has significantly improved AIDS-associated morbidity, but has limitations of adverse effects, frequent dosing regimen leading to medical non-adherence. Drug delivery systems that target HIV reservoirs could potentially reduce dose-dependent toxicity and the duration of treatment. The major cellular HIV reservoirs are macrophages and CD4+ T cells with macrophages being responsible for carrying and spreading the virus. The crucial involvement of macrophages in the pathogenesis of HIV infection has led to development of macrophage targeted nanocarrier delivery systems. AREAS COVERED Eradication of viral reservoirs like HIV-infected macrophages has emerged to be a fundamental barrier and challenge for complete eradication of HIV from the immune system. Literature reports several macrophage targeted nanocarrier delivery systems developed as either functionalized or non-functionalized formulations such as liposomes, ethosomes, polymeric nanoparticles, dendrimers, and solid lipid nanoparticles showcasing superior efficacy over the conventional antiretroviral delivery systems. EXPERT OPINION The development of fixed dose combination of antiretroviral drugs into macrophage targeted delivery systems should factor in the inherent plasticity and heterogeneity of macrophages that is dependent on their microenvironment. A rational selection of nanocarriers will facilitate selectivity and enhanced efficacy of antiretroviral drugs accompanied by reduced dosing and toxicity. Such macrophage targeted delivery systems would positively impact the therapeutic outcomes in the management of HIV infection.
Collapse
Affiliation(s)
- Tabassum Khan
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai, Maharashtra, India
| | - Mayuresh Mayuresh Patkar
- Department of Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai, Maharashtra, India
| | - Munira Momin
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai, Maharashtra, India
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University , Sudbury, ON, Canada
| |
Collapse
|
54
|
Mascarau R, Bertrand F, Labrousse A, Gennero I, Poincloux R, Maridonneau-Parini I, Raynaud-Messina B, Vérollet C. HIV-1-Infected Human Macrophages, by Secreting RANK-L, Contribute to Enhanced Osteoclast Recruitment. Int J Mol Sci 2020; 21:ijms21093154. [PMID: 32365752 PMCID: PMC7246503 DOI: 10.3390/ijms21093154] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/16/2022] Open
Abstract
HIV-1 infection is frequently associated with low bone density, which can progress to osteoporosis leading to a high risk of fractures. Only a few mechanisms have been proposed to explain the enhanced osteolysis in the context of HIV-1 infection. As macrophages are involved in bone homeostasis and are critical host cells for HIV-1, we asked whether HIV-1-infected macrophages could participate in bone degradation. Upon infection, human macrophages acquired some osteoclast features: they became multinucleated, upregulated the osteoclast markers RhoE and β3 integrin, and organized their podosomes as ring superstructures resembling osteoclast sealing zones. However, HIV-1-infected macrophages were not fully differentiated in osteoclasts as they did not upregulate NFATc-1 transcription factor and were unable to degrade bone. Investigating whether infected macrophages participate indirectly to virus-induced osteolysis, we showed that they produce RANK-L, the key osteoclastogenic cytokine. RANK-L secreted by HIV-1-infected macrophages was not sufficient to stimulate multinucleation, but promoted the protease-dependent migration of osteoclast precursors. In conclusion, we propose that, by stimulating RANK-L secretion, HIV-1-infected macrophages contribute to create a microenvironment that favors the recruitment of osteoclasts, participating in bone disorders observed in HIV-1 infected patients.
Collapse
Affiliation(s)
- Rémi Mascarau
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS UMR 5089, Université Toulouse III Paul Sabatier, CEDEX 04, 31077 Toulouse, France; (R.M.); (F.B.); (A.L.); (R.P.); (I.M.-P.)
| | - Florent Bertrand
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS UMR 5089, Université Toulouse III Paul Sabatier, CEDEX 04, 31077 Toulouse, France; (R.M.); (F.B.); (A.L.); (R.P.); (I.M.-P.)
| | - Arnaud Labrousse
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS UMR 5089, Université Toulouse III Paul Sabatier, CEDEX 04, 31077 Toulouse, France; (R.M.); (F.B.); (A.L.); (R.P.); (I.M.-P.)
| | - Isabelle Gennero
- Centre de Physiopathologie de Toulouse-Purpan, INSERM-CNRS UMR 1043, Université Toulouse III Paul Sabatier, 31024 Toulouse, France;
- Institut Fédératif de Biologie, Centre Hospitalier Universitaire Toulouse, 31059 Toulouse, France
| | - Renaud Poincloux
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS UMR 5089, Université Toulouse III Paul Sabatier, CEDEX 04, 31077 Toulouse, France; (R.M.); (F.B.); (A.L.); (R.P.); (I.M.-P.)
- International Associated Laboratory (LIA) CNRS “IM-TB/HIV” (1167), 31077 Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM-TB/HIV” (1167), Buenos Aires C1425AUM, Argentina
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS UMR 5089, Université Toulouse III Paul Sabatier, CEDEX 04, 31077 Toulouse, France; (R.M.); (F.B.); (A.L.); (R.P.); (I.M.-P.)
- International Associated Laboratory (LIA) CNRS “IM-TB/HIV” (1167), 31077 Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM-TB/HIV” (1167), Buenos Aires C1425AUM, Argentina
| | - Brigitte Raynaud-Messina
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS UMR 5089, Université Toulouse III Paul Sabatier, CEDEX 04, 31077 Toulouse, France; (R.M.); (F.B.); (A.L.); (R.P.); (I.M.-P.)
- International Associated Laboratory (LIA) CNRS “IM-TB/HIV” (1167), 31077 Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM-TB/HIV” (1167), Buenos Aires C1425AUM, Argentina
- Correspondence: (B.R.-M.); (C.V.)
| | - Christel Vérollet
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS UMR 5089, Université Toulouse III Paul Sabatier, CEDEX 04, 31077 Toulouse, France; (R.M.); (F.B.); (A.L.); (R.P.); (I.M.-P.)
- International Associated Laboratory (LIA) CNRS “IM-TB/HIV” (1167), 31077 Toulouse, France
- International Associated Laboratory (LIA) CNRS “IM-TB/HIV” (1167), Buenos Aires C1425AUM, Argentina
- Correspondence: (B.R.-M.); (C.V.)
| |
Collapse
|
55
|
de Pablo-Maiso L, Echeverría I, Rius-Rocabert S, Luján L, Garcin D, de Andrés D, Nistal-Villán E, Reina R. Sendai Virus, a Strong Inducer of Anti-Lentiviral State in Ovine Cells. Vaccines (Basel) 2020; 8:vaccines8020206. [PMID: 32365702 PMCID: PMC7349755 DOI: 10.3390/vaccines8020206] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 11/16/2022] Open
Abstract
Small ruminant lentiviruses (SRLVs) are widely spread in the ovine and caprine populations, causing an incurable disease affecting animal health and production. Vaccine development is hindered owing to the high genetic heterogeneity of lentiviruses and the selection of T-cell and antibody escape mutants, requiring antigen delivery optimization. Sendai virus (SeV) is a respiratory paramyxovirus in mice that has been recognized as a potent inducer of innate immune responses in several species, including mouse and human. The aim of this study was to stimulate an innate antiviral response in ovine cells and evaluate the potential inhibitory effect upon small ruminant lentivirus (SRLV) infections. Ovine alveolar macrophages (AMs), blood-derived macrophages (BDMs), and skin fibroblasts (OSFs) were stimulated through infection with SeV encoding green fluorescent protein (GFP). SeV efficiently infected ovine cells, inducing an antiviral state in AM from SRLV naturally-infected animals, as well as in in vitro SRLV-infected BDM and OSF from non-infected animals. Supernatants from SeV-infected AM induced an antiviral state when transferred to fresh cells challenged with SRLV. Similar to SRLV, infectivity of an HIV-1-GFP lentiviral vector was also restricted in ovine cells infected with SeV. In myeloid cells, an M1-like proinflammatory polarization was observed together with an APOBEC3Z1 induction, among other lentiviral restriction factors. Our observations may boost new approximations in ameliorating the SRLV burden by stimulation of the innate immune response using SeV-based vaccine vectors.
Collapse
Affiliation(s)
- Lorena de Pablo-Maiso
- Department of Animal Health, Institute of Agrobiotechnology (CSIC-Government of Navarra), 31192 Mutilva, Navarra, Spain; (L.d.P.-M.); (I.E.); (D.d.A.)
| | - Irache Echeverría
- Department of Animal Health, Institute of Agrobiotechnology (CSIC-Government of Navarra), 31192 Mutilva, Navarra, Spain; (L.d.P.-M.); (I.E.); (D.d.A.)
| | - Sergio Rius-Rocabert
- Microbiology Section, Departamento Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, CEU Universities, Boadilla del Monte, 28668 Madrid, Spain; (S.R.-R.); (E.N.-V.)
- CEMBIO (Centre for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad CEU San Pablo, CEU Universities, Boadilla del Monte, 28668 Madrid, Spain
| | - Lluís Luján
- Department of Animal Pathology, University of Zaragoza, 50013 Zaragoza, Spain;
| | - Dominique Garcin
- Department of Microbiology and Molecular Medicine, University of Geneva, 1211 Geneva, Switzerland;
| | - Damián de Andrés
- Department of Animal Health, Institute of Agrobiotechnology (CSIC-Government of Navarra), 31192 Mutilva, Navarra, Spain; (L.d.P.-M.); (I.E.); (D.d.A.)
| | - Estanislao Nistal-Villán
- Microbiology Section, Departamento Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, CEU Universities, Boadilla del Monte, 28668 Madrid, Spain; (S.R.-R.); (E.N.-V.)
- Instituto de Medicina Molecular Aplicada (IMMA), Universidad CEU San Pablo, Pablo-CEU, CEU Universities, Boadilla del Monte, 28003 Madrid, Spain
| | - Ramsés Reina
- Department of Animal Health, Institute of Agrobiotechnology (CSIC-Government of Navarra), 31192 Mutilva, Navarra, Spain; (L.d.P.-M.); (I.E.); (D.d.A.)
- Correspondence:
| |
Collapse
|
56
|
Dupont M, Sattentau QJ. Macrophage Cell-Cell Interactions Promoting HIV-1 Infection. Viruses 2020; 12:E492. [PMID: 32354203 PMCID: PMC7290394 DOI: 10.3390/v12050492] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023] Open
Abstract
Many pathogens infect macrophages as part of their intracellular life cycle. This is particularly true for viruses, of which HIV-1 is one of the best studied. HIV-1 infection of macrophages has important consequences for viral persistence and pathogenesis, but the mechanisms of macrophage infection remain to be fully elucidated. Despite expressing viral entry receptors, macrophages are inefficiently infected by cell-free HIV-1 virions, whereas direct cell-cell spread is more efficient. Different modes of cell-cell spread have been described, including the uptake by macrophages of infected T cells and the fusion of infected T cells with macrophages, both leading to macrophage infection. Cell-cell spread can also transmit HIV-1 between macrophages and from macrophages to T cells. Here, we describe the current state of the field concerning the cell-cell spread of HIV-1 to and from macrophages, discuss mechanisms, and highlight potential in vivo relevance.
Collapse
Affiliation(s)
- Maeva Dupont
- The Sir William Dunn School of Pathology, The University of Oxford, Oxford OX13RE, UK
| | | |
Collapse
|
57
|
Khan N, Chen X, Geiger JD. Role of Divalent Cations in HIV-1 Replication and Pathogenicity. Viruses 2020; 12:E471. [PMID: 32326317 PMCID: PMC7232465 DOI: 10.3390/v12040471] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/09/2020] [Accepted: 04/18/2020] [Indexed: 12/22/2022] Open
Abstract
Divalent cations are essential for life and are fundamentally important coordinators of cellular metabolism, cell growth, host-pathogen interactions, and cell death. Specifically, for human immunodeficiency virus type-1 (HIV-1), divalent cations are required for interactions between viral and host factors that govern HIV-1 replication and pathogenicity. Homeostatic regulation of divalent cations' levels and actions appear to change as HIV-1 infection progresses and as changes occur between HIV-1 and the host. In people living with HIV-1, dietary supplementation with divalent cations may increase HIV-1 replication, whereas cation chelation may suppress HIV-1 replication and decrease disease progression. Here, we review literature on the roles of zinc (Zn2+), iron (Fe2+), manganese (Mn2+), magnesium (Mg2+), selenium (Se2+), and copper (Cu2+) in HIV-1 replication and pathogenicity, as well as evidence that divalent cation levels and actions may be targeted therapeutically in people living with HIV-1.
Collapse
Affiliation(s)
| | | | - Jonathan D. Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA; (N.K.); (X.C.)
| |
Collapse
|
58
|
Abstract
PURPOSE OF REVIEW HIV-1 infection is incurable due to the existence of latent reservoirs that persist in the face of cART. In this review, we describe the existence of multiple HIV-1 reservoirs, the mechanisms that support their persistence, and the potential use of tyrosine kinase inhibitors (TKIs) to block several pathogenic processes secondary to HIV-1 infection. RECENT FINDINGS Dasatinib interferes in vitro with HIV-1 persistence by two independent mechanisms. First, dasatinib blocks infection and potential expansion of the latent reservoir by interfering with the inactivating phosphorylation of SAMHD1. Secondly, dasatinib inhibits the homeostatic proliferation induced by γc-cytokines. Since homeostatic proliferation is thought to be the main mechanism behind the maintenance of the latent reservoir, we propose that blocking this process will gradually reduce the size of the reservoir. TKIs together with cART will interfere with HIV-1 latent reservoir persistence, favoring the prospect for viral eradication.
Collapse
|
59
|
Dupont M, Souriant S, Balboa L, Vu Manh TP, Pingris K, Rousset S, Cougoule C, Rombouts Y, Poincloux R, Ben Neji M, Allers C, Kaushal D, Kuroda MJ, Benet S, Martinez-Picado J, Izquierdo-Useros N, Sasiain MDC, Maridonneau-Parini I, Neyrolles O, Vérollet C, Lugo-Villarino G. Tuberculosis-associated IFN-I induces Siglec-1 on tunneling nanotubes and favors HIV-1 spread in macrophages. eLife 2020; 9:52535. [PMID: 32223897 PMCID: PMC7173963 DOI: 10.7554/elife.52535] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/30/2020] [Indexed: 12/20/2022] Open
Abstract
While tuberculosis (TB) is a risk factor in HIV-1-infected individuals, the mechanisms by which Mycobacterium tuberculosis (Mtb) worsens HIV-1 pathogenesis remain scarce. We showed that HIV-1 infection is exacerbated in macrophages exposed to TB-associated microenvironments due to tunneling nanotube (TNT) formation. To identify molecular factors associated with TNT function, we performed a transcriptomic analysis in these macrophages, and revealed the up-regulation of Siglec-1 receptor. Siglec-1 expression depends on Mtb-induced production of type I interferon (IFN-I). In co-infected non-human primates, Siglec-1 is highly expressed by alveolar macrophages, whose abundance correlates with pathology and activation of IFN-I/STAT1 pathway. Siglec-1 localizes mainly on microtubule-containing TNT that are long and carry HIV-1 cargo. Siglec-1 depletion decreases TNT length, diminishes HIV-1 capture and cell-to-cell transfer, and abrogates the exacerbation of HIV-1 infection induced by Mtb. Altogether, we uncover a deleterious role for Siglec-1 in TB-HIV-1 co-infection and open new avenues to understand TNT biology.
Collapse
Affiliation(s)
- Maeva Dupont
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.,International associated laboratory (LIA) CNRS 'IM-TB/HIV', Toulouse, France
| | - Shanti Souriant
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.,International associated laboratory (LIA) CNRS 'IM-TB/HIV', Toulouse, France
| | - Luciana Balboa
- International associated laboratory (LIA) CNRS 'IM-TB/HIV', Toulouse, France.,Institute of Experimental Medicine-CONICET, National Academy of Medicine, Buenos Aires, Argentina
| | | | - Karine Pingris
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Stella Rousset
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Céline Cougoule
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.,International associated laboratory (LIA) CNRS 'IM-TB/HIV', Toulouse, France
| | - Yoann Rombouts
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Renaud Poincloux
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Myriam Ben Neji
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Carolina Allers
- Tulane National Primate Research Center, Department of Microbiology and Immunology, School of Medicine, Tulane University, Covington, United States
| | - Deepak Kaushal
- Tulane National Primate Research Center, Department of Microbiology and Immunology, School of Medicine, Tulane University, Covington, United States
| | - Marcelo J Kuroda
- Tulane National Primate Research Center, Department of Microbiology and Immunology, School of Medicine, Tulane University, Covington, United States
| | - Susana Benet
- IrsiCaixa AIDS Research Institute, Department of Retrovirology, Badalona, Spain.,Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, Department of Retrovirology, Badalona, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Department of Retrovirology, Badalona, Spain.,Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Maria Del Carmen Sasiain
- International associated laboratory (LIA) CNRS 'IM-TB/HIV', Toulouse, France.,Institute of Experimental Medicine-CONICET, National Academy of Medicine, Buenos Aires, Argentina
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.,International associated laboratory (LIA) CNRS 'IM-TB/HIV', Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.,International associated laboratory (LIA) CNRS 'IM-TB/HIV', Toulouse, France
| | - Christel Vérollet
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.,International associated laboratory (LIA) CNRS 'IM-TB/HIV', Toulouse, France
| | - Geanncarlo Lugo-Villarino
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.,International associated laboratory (LIA) CNRS 'IM-TB/HIV', Toulouse, France
| |
Collapse
|
60
|
Moar P, Sushmita K, Kateriya S, Tandon R. Transcriptional profiling indicates cAMP-driven reversal of HIV latency in monocytes occurs via transcription factor SP-1. Virology 2020; 542:40-53. [PMID: 32056667 DOI: 10.1016/j.virol.2020.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/11/2020] [Accepted: 01/14/2020] [Indexed: 01/16/2023]
Abstract
Latent HIV reservoir is a major barrier to absolute HIV cure. Studies on latency reversal agents (LRA) have by far focused mainly on CD4+ T-lymphocytes, while myeloid reservoirs remain under-represented despite their persistence and key contribution to HIV pathogenesis. cAMP has been shown to increase HIV-1 transcription in latently-infected monocytes/macrophages. In this communication, we explored the potential of commercially available pharmacological drugs and phosphodiesterase inhibitors to reactivate HIV in latently-infected monocytic cell-line, U1. We showed that increased levels of intracellular cAMP reverse HIV latency in vitro, which is specific to cells of the myeloid lineage. High throughput RNA-seq analysis revealed that cAMP modulates transcriptional profile of latently HIV-infected cells and provides favourable cellular environment for HIV to produce viral proteins. This reactivation of latent HIV was inhibited by Mithramycin A, a selective Sp1 inhibitor, indicating that the reversal of HIV latency in monocytes is driven by transcription factor Sp1.
Collapse
Affiliation(s)
- Preeti Moar
- Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Kumari Sushmita
- Laboratory of Optobiology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Suneel Kateriya
- Laboratory of Optobiology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Ravi Tandon
- Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
61
|
Waters R, Ndengane M, Abrahams MR, Diedrich CR, Wilkinson RJ, Coussens AK. The Mtb-HIV syndemic interaction: why treating M. tuberculosis infection may be crucial for HIV-1 eradication. Future Virol 2020; 15:101-125. [PMID: 32273900 PMCID: PMC7132588 DOI: 10.2217/fvl-2019-0069] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Accelerated tuberculosis and AIDS progression seen in HIV-1 and Mycobacterium tuberculosis (Mtb)-coinfected individuals indicates the important interaction between these syndemic pathogens. The immunological interaction between HIV-1 and Mtb has been largely defined by how the virus exacerbates tuberculosis disease pathogenesis. Understanding of the mechanisms by which pre-existing or subsequent Mtb infection may favor the replication, persistence and progression of HIV, is less characterized. We present a rationale for the critical consideration of ‘latent’ Mtb infection in HIV-1 prevention and cure strategies. In support of this position, we review evidence of the effect of Mtb infection on HIV-1 acquisition, replication and persistence. We propose that ‘latent’ Mtb infection may have considerable impact on HIV-1 pathogenesis and the continuing HIV-1 epidemic in sub-Saharan Africa.
Collapse
Affiliation(s)
- Robyn Waters
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Medicine, University of Cape Town, Observatory 7925, WC, South Africa
| | - Mthawelanga Ndengane
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Pathology, University of Cape Town, Observatory 7925, WC, South Africa
| | - Melissa-Rose Abrahams
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Pathology, University of Cape Town, Observatory 7925, WC, South Africa
| | - Collin R Diedrich
- Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Robert J Wilkinson
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Infectious Diseases, Imperial College London, London W2 1PG, United Kingdom.,The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Anna K Coussens
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Observatory 7925, WC, South Africa.,Department of Pathology, University of Cape Town, Observatory 7925, WC, South Africa.,Infectious Diseases and Immune Defence Division, The Walter & Eliza Hall Institute of Medical Research, Parkville 3279, VIC, Australia.,Division of Medical Biology, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville 3279, VIC, Australia
| |
Collapse
|
62
|
RifeMagalis B, Strickland SL, Shank SD, Autissier P, Schuetz A, Sithinamsuwan P, Lerdlum S, Fletcher JLK, de Souza M, Ananworanich J, Valcour V, Williams KC, Kosakovsky Pond SL, RattoKim S, Salemi M. Phyloanatomic characterization of the distinct T cell and monocyte contributions to the peripheral blood HIV population within the host. Virus Evol 2020; 6:veaa005. [PMID: 32355568 PMCID: PMC7185683 DOI: 10.1093/ve/veaa005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Human immunodeficiency virus (HIV) is a rapidly evolving virus, allowing its genetic sequence to act as a fingerprint for epidemiological processes among, as well as within, individual infected hosts. Though primarily infecting the CD4+ T-cell population, HIV can also be found in monocytes, an immune cell population that differs in several aspects from the canonical T-cell viral target. Using single genome viral sequencing and statistical phylogenetic inference, we investigated the viral RNA diversity and relative contribution of each of these immune cell types to the viral population within the peripheral blood. Results provide evidence of an increased prevalence of circulating monocytes harboring virus in individuals with high viral load in the absence of suppressive antiretroviral therapy. Bayesian phyloanatomic analysis of three of these individuals demonstrated a measurable role for these cells, but not the circulating T-cell population, as a source of cell-free virus in the plasma, supporting the hypothesis that these cells can act as an additional conduit of virus spread.
Collapse
Affiliation(s)
- Brittany RifeMagalis
- Department of Pathology, Immunology, and Laboratory Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL 32601, USA
| | - Samantha L Strickland
- Department of Pathology, Immunology, and Laboratory Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL 32601, USA
| | - Stephen D Shank
- Department of Biology, Temple University, Philadelphia, PA 19122, USA
| | | | - Alexandra Schuetz
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences - United States Component, Bangkok 10400, Thailand
- SEARCH, Thai Red Cross AIDS Research Center, Bangkok 10330, Thailand
| | - Pasiri Sithinamsuwan
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Rockville, MD 20850, USA
| | - Sukalaya Lerdlum
- Division of Neurology, Department of Medicine, Phramongkutklao Hospital, Bangkok 10400, Thailand
| | - James L K Fletcher
- Faculty of Medicine, Department of Radiology, Chulalongkorn University, Bangkok 10330, Thailand
| | - Mark de Souza
- Faculty of Medicine, Department of Radiology, Chulalongkorn University, Bangkok 10330, Thailand
| | - Jintanat Ananworanich
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences - United States Component, Bangkok 10400, Thailand
- SEARCH, Thai Red Cross AIDS Research Center, Bangkok 10330, Thailand
- Faculty of Medicine, Department of Radiology, Chulalongkorn University, Bangkok 10330, Thailand
| | - Victor Valcour
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | | | | | | | - Silvia RattoKim
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences - United States Component, Bangkok 10400, Thailand
- SEARCH, Thai Red Cross AIDS Research Center, Bangkok 10330, Thailand
| | - Marco Salemi
- Department of Pathology, Immunology, and Laboratory Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL 32601, USA
| |
Collapse
|
63
|
Abstract
: Given the challenges of life-long adherence to suppressive HIV antiretroviral therapy (ART) and possibilities of comorbidities, such as HIV association neurocognitive disorder, HIV remission and eradication are desirable goals for people living with HIV. In some individuals, there is evidence that HIV persists and replicates in the CNS, impacting the success of HIV remission interventions. This article addresses the role of HIV CNS latency on HIV eradication, examines the effects of early ART, latency-modifying agents, antibody-based and T-cell enhancing therapies on the CNS as well as ART interruption in remission studies. We propose the integration of CNS monitoring into such studies in order to clarify the short-term and long-term neurological safety of experimental agents and treatment interruption, and to better characterize their effects on HIV CNS persistence.
Collapse
|
64
|
Trimeric heptad repeat synthetic peptides HR1 and HR2 efficiently inhibit HIV-1 entry. Biosci Rep 2019; 39:BSR20192196. [PMID: 31477581 PMCID: PMC6757187 DOI: 10.1042/bsr20192196] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/30/2019] [Accepted: 08/06/2019] [Indexed: 12/16/2022] Open
Abstract
The trimeric heptad repeat domains HR1 and HR2 of the human immunodeficiency virus 1 (HIV-1) gp41 play a key role in HIV-1-entry by membrane fusion. To develop efficient inhibitors against this step, the corresponding trimeric-N36 and C34 peptides were designed and synthesized. Analysis by circular dichroism of monomeric and trimeric N36 and C34 peptides showed their capacities to adopt α-helical structures and to establish physical interactions. At the virological level, while trimeric-C34 conserves the same high anti-fusion activity as monomeric-C34, trimerization of N36-peptide induced a significant increase, reaching 500-times higher in anti-fusion activity, against R5-tropic virus-mediated fusion. This result was associated with increased stability of the N36 trimer peptide with respect to the monomeric form, as demonstrated by the comparative kinetics of their antiviral activities during 6-day incubation in a physiological medium. Collectively, our findings demonstrate that while the trimerization of C34 peptide had no beneficial effect on its stability and antiviral activity, the trimerization of N36 peptide strengthened both stability and antiviral activity. This approach, promotes trimers as new promising HIV-1 inhibitors and point to future development aimed toward innovative peptide fusion inhibitors, microbicides or as immunogens.
Collapse
|
65
|
Abreu CM, Veenhuis RT, Avalos CR, Graham S, Parrilla DR, Ferreira EA, Queen SE, Shirk EN, Bullock BT, Li M, Metcalf Pate KA, Beck SE, Mangus LM, Mankowski JL, Mac Gabhann F, O'Connor SL, Gama L, Clements JE. Myeloid and CD4 T Cells Comprise the Latent Reservoir in Antiretroviral Therapy-Suppressed SIVmac251-Infected Macaques. mBio 2019; 10:e01659-19. [PMID: 31431552 PMCID: PMC6703426 DOI: 10.1128/mbio.01659-19] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022] Open
Abstract
Human immunodeficiency virus (HIV) eradication or long-term suppression in the absence of antiretroviral therapy (ART) requires an understanding of all viral reservoirs that could contribute to viral rebound after ART interruption. CD4 T cells (CD4s) are recognized as the predominant reservoir in HIV type 1 (HIV-1)-infected individuals. However, macrophages are also infected by HIV-1 and simian immunodeficiency virus (SIV) during acute infection and may persist throughout ART, contributing to the size of the latent reservoir. We sought to determine whether tissue macrophages contribute to the SIVmac251 reservoir in suppressed macaques. Using cell-specific quantitative viral outgrowth assays (CD4-QVOA and MΦ-QVOA), we measured functional latent reservoirs in CD4s and macrophages in ART-suppressed SIVmac251-infected macaques. Spleen, lung, and brain in all suppressed animals contained latently infected macrophages, undetectable or low-level SIV RNA, and detectable SIV DNA. Silent viral genomes with potential for reactivation and viral spread were also identified in blood monocytes, although these cells might not be considered reservoirs due to their short life span. Additionally, virus produced in the MΦ-QVOA was capable of infecting healthy activated CD4s. Our results strongly suggest that functional latent reservoirs in CD4s and macrophages can contribute to viral rebound and reestablishment of productive infection after ART interruption. These findings should be considered in the design and implementation of future HIV cure strategies.IMPORTANCE This study provides further evidence that the latent reservoir is comprised of both CD4+ T cells and myeloid cells. The data presented here suggest that CD4+ T cells and macrophages found throughout tissues in the body can contain replication-competent SIV and contribute to rebound of the virus after treatment interruption. Additionally, we have shown that monocytes in blood contain latent virus and, though not considered a reservoir themselves due to their short life span, could contribute to the size of the latent reservoir upon entering the tissue and differentiating into long-lived macrophages. These new insights into the size and location of the SIV reservoir using a model that is heavily studied in the HIV field could have great implications for HIV-infected individuals and should be taken into consideration with the development of future HIV cure strategies.
Collapse
Affiliation(s)
- Celina M Abreu
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Rebecca T Veenhuis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Claudia R Avalos
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Shelby Graham
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Daymond R Parrilla
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Edna A Ferreira
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Suzanne E Queen
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Erin N Shirk
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Brandon T Bullock
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ming Li
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Kelly A Metcalf Pate
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Sarah E Beck
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Lisa M Mangus
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Joseph L Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Feilim Mac Gabhann
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shelby L O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lucio Gama
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Janice E Clements
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
66
|
Wang Q, Su L. Vpr Enhances HIV-1 Env Processing and Virion Infectivity in Macrophages by Modulating TET2-Dependent IFITM3 Expression. mBio 2019; 10:e01344-19. [PMID: 31431548 PMCID: PMC6703422 DOI: 10.1128/mbio.01344-19] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/24/2019] [Indexed: 12/28/2022] Open
Abstract
HIV-1 Vpr enhances viral replication in human macrophages via multiple mechanisms that are not clearly defined. It does not affect HIV-1 virion production during the first round of infection. We have recently discovered that Vpr targets the DNA demethylase TET2 for degradation, which leads to sustained interleukin-6 (IL-6) expression and elevated HIV-1 replication. We report here that Vpr enhanced Env processing in infected macrophages, associated with increased Env incorporation into virions with higher infectivity. Interestingly, IFITM3 was constitutively expressed in macrophages in a TET2-dependent fashion. We showed that Vpr-enhanced Env processing depended genetically on TET2 and IFITM3. We further showed that Vpr reduced IFITM3 expression by reducing demethylation of the IFITM3 promoter in macrophages, associated with degradation of TET2 and reduced TET2 binding to the IFITIM3 promoter. Our findings indicate that the Vpr-TET2 axis enhances HIV-1 replication in macrophages via two independent mechanisms: reduced IFTIM3 expression to enhance Env processing and virion infectivity and sustained IL-6 expression to increase HIV-1 replication. The Vpr-TET2 axis may provide a novel target to develop therapeutics to inhibit HIV-1 infection and pathogenesis.IMPORTANCE How Vpr enhances HIV-1 replication in macrophages is still unclear. We report here that Vpr enhanced HIV-1 Env processing during the first round of HIV-1 replication, resulting in virions with higher Env incorporation and viral infectivity. These higher-quality viral particles contributed to elevated infection during the second round and spreading infection in macrophages and other HIV-1 target cells. We have recently discovered that TET2 is a novel host factor degraded by Vpr, which leads to sustained IL-6 expression in macrophages. Interestingly, Vpr-enhanced HIV-1 Env processing depended on both the IFITIM3 and TET2 genes. The constitutive expression of IFITIM3 expression in macrophages was maintained by TET2, which demethylated the IFITIM3 promoter. We conclude that the Vpr degrades TET2 to enhance HIV-1 replication in macrophages by reducing IFITIM3 expression to increase viral Env processing, virion incorporation, and infectivity and by sustaining IL-6 expression to increase HIV-1 gene expression. The Vpr-TET2 axis may serve as a novel target to develop anti-HIV drugs to inhibit HIV-1 infection and pathogenesis.
Collapse
Affiliation(s)
- Qi Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology & Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lishan Su
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology & Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
67
|
Ibrahim IM, Bade AN, Lin Z, Soni D, Wojtkiewicz M, Dyavar Shetty BL, Gautam N, McMillan JM, Alnouti Y, Edagwa BJ, Gendelman HE. Synthesis and characterization of a long-acting emtricitabine prodrug nanoformulation. Int J Nanomedicine 2019; 14:6231-6247. [PMID: 31496683 PMCID: PMC6689761 DOI: 10.2147/ijn.s215447] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 07/10/2019] [Indexed: 12/16/2022] Open
Abstract
Purpose A palmitoylated prodrug of emtricitabine (FTC) was synthesized to extend the drug’s half-life, antiretroviral activities and biodistribution. Methods A modified FTC prodrug (MFTC) was synthesized by palmitoyl chloride esterification. MFTC’s chemical structure was evaluated by nuclear magnetic resonance. The created hydrophobic prodrug nanocrystals were encased into a poloxamer surfactant and the pharmacokinetics (PK), biodistribution and antiretroviral activities of the nanoformulation (NMFTC) were assessed. The conversion of MFTC to FTC triphosphates was evaluated. Results MFTC coated with poloxamer formed stable nanocrystals (NMFTC). NMFTC demonstrated an average particle size, polydispersity index and zeta potential of 350 nm, 0.24 and −20 mV, respectively. Drug encapsulation efficiency was 90%. NMFTC was readily taken up by human monocyte-derived macrophages yielding readily detected intracellular FTC triphosphates and an extended PK profile. Conclusion NMFTC shows improved antiretroviral activities over native FTC. This is coordinate with its extended apparent half-life. The work represents an incremental advance in the development of a long-acting FTC formulation.
Collapse
Affiliation(s)
- Ibrahim M Ibrahim
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Pharmacology, College of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Aditya N Bade
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Zhiyi Lin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dhruvkumar Soni
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Melinda Wojtkiewicz
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bhagya Laxmi Dyavar Shetty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nagsen Gautam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - JoEllyn M McMillan
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yazen Alnouti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benson J Edagwa
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
68
|
Mu Y, Patters BJ, Midde NM, He H, Kumar S, Cory TJ. Tobacco and Antiretrovirals Modulate Transporter, Metabolic Enzyme, and Antioxidant Enzyme Expression and Function in Polarized Macrophages. Curr HIV Res 2019; 16:354-363. [PMID: 30706821 PMCID: PMC6446460 DOI: 10.2174/1570162x17666190130114531] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/24/2019] [Accepted: 01/27/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cigarette smoking increases systemic oxidative stress, inflammation, and viral replication in individuals with HIV. Macrophages are infected during HIV infection and serve as an important reservoir throughout the process. Macrophages exist in two phenotypes, the classically activated M1 macrophage and alternatively activated M2 macrophage. The expression of drug efflux transporters and metabolic enzymes, which have direct effects on intracellular drug concentrations, differ between the pro-inflammatory M1 macrophage and the anti-inflammatory M2 macrophage. OBJECTIVE To further explain the role of tobacco use in worsened outcomes in the HIV + population receiving antiretroviral therapy. METHODS Western blotting was used to examine macrophage polarization and expression of drug efflux transporters, CYP enzymes, and antioxidant enzymes. The arginase assay was used to measure arginase activity. Cytokine production was measured using the human multiplex inflammatory cytokine assay kit. The 8-OHdG DNA Damage Quantification Direct Kit was used to quantify DNA damage. Viral replication under the influence of tobacco and antiretroviral drug use was measured by p24 Elisa. RESULTS We observed phenotypic shifts from M1 to M2 with both individual and combination treatments with cigarette smoke condensate and the protease inhibitor antiretroviral drug lopinavir. These shifts lead to changes in cytokine production, the expression of CYP enzymes, anti-oxidant enzymes, and drug efflux transporters, as well as changes in viral replication. CONCLUSION This data suggest a mechanism by which tobacco use impairs HIV antiretroviral therapy to increase intracellular drug concentrations in this important cellular reservoir.
Collapse
Affiliation(s)
- Ying Mu
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| | - Benjamin J Patters
- Department of Pharmaceutical Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| | - Narasimha M Midde
- Department of Pharmaceutical Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| | - Hui He
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| | - Santosh Kumar
- Department of Pharmaceutical Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| | - Theodore J Cory
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, United States
| |
Collapse
|
69
|
Cattin A, Wiche Salinas TR, Gosselin A, Planas D, Shacklett B, Cohen EA, Ghali MP, Routy JP, Ancuta P. HIV-1 is rarely detected in blood and colon myeloid cells during viral-suppressive antiretroviral therapy. AIDS 2019; 33:1293-1306. [PMID: 30870200 PMCID: PMC6686847 DOI: 10.1097/qad.0000000000002195] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The aim of this study was to explore the contribution of blood and colon myeloid cells to HIV persistence during antiretroviral therapy (ART). DESIGN Leukapheresis was collected from HIV-infected individuals with undetectable plasma viral load during ART (HIV + ART; n = 15) and viremics untreated (HIV+; n = 6). Rectal sigmoid biopsies were collected from n = 8 HIV+ART. METHODS Myeloid cells (total monocytes (Mo), CD16/CD16 Mo, CD1c dendritic cells) and CD4 T cells were isolated by magnetic-activated cell sorting (MACS) and/or fluorescence-activated cell sorting (FACS) from peripheral blood. Matched myeloid and CCR6CD4 T cells were isolated from blood and rectal biopsies by FACS. Levels of early (RU5 primers), late (Gag primers) and/or integrated HIV-DNA (Alu/HIV primers) were quantified by nested real-time PCR. Replication-competent HIV was amplified by co-culturing cells from HIV-positive individuals with CD3/CD28-activated CD4 T cells from uninfected donors. RESULTS Early/late but not integrated HIV reverse transcripts were detected in blood myeloid subsets of four out of 10 HIV+ART; in contrast, integrated HIV-DNA was exclusively detected in CD4 T cells. In rectal biopsies, late HIV reverse transcripts were detected in myeloid cells and CCR6CD4 T cells from one out of eight and seven out of eight HIV+ART individuals, respectively. Replication-competent HIV was outgrown from CD4 T cells but not from myeloid of untreated/ART-treated HIV-positive individuals. CONCLUSION In contrast to CD4 T cells, blood and colon myeloid cells carry detectable HIV only in a small fraction of HIV+ART individuals. This is consistent with the documented resistance of Mo to HIV infection and the rapid turnover of Mo-derived macrophages in the colon. Future assessment of multiple lymphoid and nonlymphoid tissues is required to include/exclude myeloid cells as relevant HIV reservoirs during ART.
Collapse
Affiliation(s)
- Amélie Cattin
- CHUM-Research Centre, Montréal, Qc, Canada
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Qc, Canada
| | - Tomas Raul Wiche Salinas
- CHUM-Research Centre, Montréal, Qc, Canada
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Qc, Canada
| | | | - Delphine Planas
- CHUM-Research Centre, Montréal, Qc, Canada
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Qc, Canada
| | | | - Eric A. Cohen
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Qc, Canada
- Institut de Recherche Clinique de Montréal, Montréal, Qc, Canada
| | - Maged P. Ghali
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Canada
| | - Jean-Pierre Routy
- Division of Hematology, McGill University Health Centre, Montreal, QC, Canada
- Chronic Viral Illness Service and Research Institute, McGill University Health Centre, Montreal, QC, Canada
| | - Petronela Ancuta
- CHUM-Research Centre, Montréal, Qc, Canada
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Qc, Canada
| |
Collapse
|
70
|
Balasubramaniam M, Pandhare J, Dash C. Immune Control of HIV. JOURNAL OF LIFE SCIENCES (WESTLAKE VILLAGE, CALIF.) 2019; 1:4-37. [PMID: 31468033 PMCID: PMC6714987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The human immunodeficiency virus (HIV) infection of the immune cells expressing the cluster of differentiation 4 cell surface glycoprotein (CD4+ cells) causes progressive decline of the immune system and leads to the acquired immunodeficiency syndrome (AIDS). The ongoing global HIV/AIDS pandemic has already claimed over 35 million lives. Even after 37 years into the epidemic, neither a cure is available for the 37 million people living with HIV (PLHIV) nor is a vaccine discovered to avert the millions of new HIV infections that continue to occur each year. If left untreated, HIV infection typically progresses to AIDS and, ultimately, causes death in a majority of PLHIV. The recommended combination antiretroviral therapy (cART) suppresses virus replication and viremia, prevents or delays progression to AIDS, reduces transmission rates, and lowers HIV-associated mortality and morbidity. However, because cART does not eliminate HIV, and an enduring pool of infected resting memory CD4+ T cells (latent HIV reservoir) is established early on, any interruption to cART leads to a relapse of viremia and disease progression. Hence, strict adherence to a life-long cART regimen is mandatory for managing HIV infection in PLHIV. The HIV-1-specific cytotoxic T cells expressing the CD8 glycoprotein (CD8+ CTL) limit the virus replication in vivo by recognizing the viral antigens presented by human leukocyte antigen (HLA) class I molecules on the infected cell surface and killing those cells. Nevertheless, CTLs fail to durably control HIV-1 replication and disease progression in the absence of cART. Intriguingly, <1% of cART-naive HIV-infected individuals called elite controllers/HIV controllers (HCs) exhibit the core features that define a HIV-1 "functional cure" outcome in the absence of cART: durable viral suppression to below the limit of detection, long-term non-progression to AIDS, and absence of viral transmission. Robust HIV-1-specific CTL responses and prevalence of protective HLA alleles associated with enduring HIV-1 control have been linked to the HC phenotype. An understanding of the molecular mechanisms underlying the CTL-mediated suppression of HIV-1 replication and disease progression in HCs carrying specific protective HLA alleles may yield promising insights towards advancing the research on HIV cure and prophylactic HIV vaccine.
Collapse
Affiliation(s)
- Muthukumar Balasubramaniam
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN – 37208. USA
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN – 37208. USA
| | - Jui Pandhare
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN – 37208. USA
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN – 37208. USA
| | - Chandravanu Dash
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN – 37208. USA
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN – 37208. USA
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN – 37208. USA
| |
Collapse
|
71
|
Ikeda T, Molan AM, Jarvis MC, Carpenter MA, Salamango DJ, Brown WL, Harris RS. HIV-1 restriction by endogenous APOBEC3G in the myeloid cell line THP-1. J Gen Virol 2019; 100:1140-1152. [PMID: 31145054 DOI: 10.1099/jgv.0.001276] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
HIV-1 replication in CD4-positive T lymphocytes requires counteraction of multiple different innate antiviral mechanisms. Macrophage cells are also thought to provide a reservoir for HIV-1 replication but less is known in this cell type about virus restriction and counteraction mechanisms. Many studies have combined to demonstrate roles for APOBEC3D, APOBEC3F, APOBEC3G and APOBEC3H in HIV-1 restriction and mutation in CD4-positive T lymphocytes, whereas the APOBEC enzymes involved in HIV-1 restriction in macrophages have yet to be delineated fully. We show that multiple APOBEC3 genes including APOBEC3G are expressed in myeloid cell lines such as THP-1. Vif-deficient HIV-1 produced from THP-1 is less infectious than Vif-proficient virus, and proviral DNA resulting from such Vif-deficient infections shows strong G to A mutation biases in the dinucleotide motif preferred by APOBEC3G. Moreover, Vif mutant viruses with selective sensitivity to APOBEC3G show Vif null-like infectivity levels and similarly strong APOBEC3G-biased mutation spectra. Importantly, APOBEC3G-null THP-1 cells yield Vif-deficient particles with significantly improved infectivities and proviral DNA with background levels of G to A hypermutation. These studies combine to indicate that APOBEC3G is the main HIV-1 restricting APOBEC3 family member in THP-1 cells.
Collapse
Affiliation(s)
- Terumasa Ikeda
- 2 Institute for Molecular Virology, Minneapolis, MN 55455, USA.,3 Center for Genome Engineering, Minneapolis, MN 55455, USA.,5 Howard Hughes Medical Institute, University of Minnesota, Minneapolis, MN 55455, USA.,1 Department of Biochemistry, Molecular Biology, and Biophysics, Minneapolis, MN 55455, USA.,4 Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Amy M Molan
- 4 Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.,2 Institute for Molecular Virology, Minneapolis, MN 55455, USA.,3 Center for Genome Engineering, Minneapolis, MN 55455, USA.,1 Department of Biochemistry, Molecular Biology, and Biophysics, Minneapolis, MN 55455, USA
| | - Matthew C Jarvis
- 3 Center for Genome Engineering, Minneapolis, MN 55455, USA.,1 Department of Biochemistry, Molecular Biology, and Biophysics, Minneapolis, MN 55455, USA.,4 Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.,2 Institute for Molecular Virology, Minneapolis, MN 55455, USA
| | - Michael A Carpenter
- 3 Center for Genome Engineering, Minneapolis, MN 55455, USA.,1 Department of Biochemistry, Molecular Biology, and Biophysics, Minneapolis, MN 55455, USA.,4 Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.,2 Institute for Molecular Virology, Minneapolis, MN 55455, USA.,5 Howard Hughes Medical Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel J Salamango
- 1 Department of Biochemistry, Molecular Biology, and Biophysics, Minneapolis, MN 55455, USA.,4 Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.,2 Institute for Molecular Virology, Minneapolis, MN 55455, USA.,3 Center for Genome Engineering, Minneapolis, MN 55455, USA
| | - William L Brown
- 4 Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.,3 Center for Genome Engineering, Minneapolis, MN 55455, USA.,2 Institute for Molecular Virology, Minneapolis, MN 55455, USA.,1 Department of Biochemistry, Molecular Biology, and Biophysics, Minneapolis, MN 55455, USA
| | - Reuben S Harris
- 3 Center for Genome Engineering, Minneapolis, MN 55455, USA.,1 Department of Biochemistry, Molecular Biology, and Biophysics, Minneapolis, MN 55455, USA.,4 Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.,2 Institute for Molecular Virology, Minneapolis, MN 55455, USA.,5 Howard Hughes Medical Institute, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
72
|
Martín-Moreno A, Sepúlveda-Crespo D, Serramía-Lobera MJ, Perisé-Barrios AJ, Muñoz-Fernández MA. G2-S16 dendrimer microbicide does not interfere with the vaginal immune system. J Nanobiotechnology 2019; 17:65. [PMID: 31092246 PMCID: PMC6518660 DOI: 10.1186/s12951-019-0496-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/04/2019] [Indexed: 12/19/2022] Open
Abstract
It is essential that prophylactic drugs do not interfere with the normal function of the immune system. The use of nanoparticles as vaginal microbicides is a promising prevention strategy against sexually transmitted infections. With that aim, our group is working with the G2-S16, a second generation carbosilane dendrimer with sulfonate groups in the periphery, which has been previously shown to be effective against HIV-1 and HSV-2 infection, and it is now on the road to clinical trials. Our objective in this new study is to assess the effects of G2-S16 on the immune barrier of the female reproductive tract. The expression of differentiation, maturation and activation markers was measured in epithelial cells, dendritic cells, M and GM macrophages, and T cells using RT-qPCR and flow cytometry. The results demonstrate that G2-S16 does not alter the natural immunity of the vagina, strongly supporting the biosafety of this dendrimer for clinical use.![]()
Collapse
Affiliation(s)
- Alba Martín-Moreno
- Sección Inmunología, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), and Spanish HIV-HGM BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Daniel Sepúlveda-Crespo
- Sección Inmunología, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), and Spanish HIV-HGM BioBank, Madrid, Spain
| | - Mª Jesús Serramía-Lobera
- Sección Inmunología, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), and Spanish HIV-HGM BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Ana Judith Perisé-Barrios
- Sección Inmunología, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), and Spanish HIV-HGM BioBank, Madrid, Spain
| | - Mª Angeles Muñoz-Fernández
- Sección Inmunología, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), and Spanish HIV-HGM BioBank, Madrid, Spain. .,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
73
|
Rodrigues V, Benaroch P. Macrophages hide HIV in the urethra. Nat Microbiol 2019; 4:556-557. [PMID: 30899108 DOI: 10.1038/s41564-019-0418-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Vasco Rodrigues
- Institut Curie, PSL Research University, INSERM U932, Paris, France.
| | - Philippe Benaroch
- Institut Curie, PSL Research University, INSERM U932, Paris, France.
| |
Collapse
|
74
|
Takahashi N, Sugimoto C, Allers C, Alvarez X, Kim WK, Didier ES, Kuroda MJ. Shifting Dynamics of Intestinal Macrophages during Simian Immunodeficiency Virus Infection in Adult Rhesus Macaques. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:2682-2689. [PMID: 30926643 PMCID: PMC6478513 DOI: 10.4049/jimmunol.1801457] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/04/2019] [Indexed: 02/07/2023]
Abstract
The intestinal tract is a primary barrier to invading pathogens and contains immune cells, including lymphocytes and macrophages. We previously reported that CD163+CD206- (single-positive [SP]) interstitial macrophages of the lung are short-lived and succumb early to SIV infection. Conversely, CD163+CD206+ (double-positive [DP]) alveolar macrophages are long-lived, survive after SIV infection, and may contribute to the virus reservoir. This report characterizes analogous populations of macrophages in the intestinal tract of rhesus macaques (Macaca mulatta) with SIV/AIDS. By flow cytometry analysis, immunofluorescence staining, and confocal microscopy, CD163+CD206+ DP macrophages predominated in the lamina propria of uninfected animals, compared with CD163+CD206- SP macrophages, which predominated in the lamina propria in animals with SIV infection that were exhibiting AIDS. In submucosal areas, CD163+CD206+ DP macrophages predominated in both SIV-infected and uninfected macaques. Furthermore, BrdU-labeled CD163+CD206+ DP and CD163+CD206- SP macrophages recently arriving in the colon, which are both presumed to be shorter-lived, were observed to localize only in the lamina propria. Conversely, longer-lived CD163+CD206+ DP macrophages that retained dextran at least 2 mo after in vivo administration localized exclusively in the submucosa. This suggests that CD163+CD206+ DP intestinal macrophages of the lamina propria were destroyed after SIV infection and replaced by immature CD163+CD206- SP macrophages, whereas longer-lived CD163+CD206+ DP macrophages remained in the submucosa, supporting their potential role as an SIV/HIV tissue reservoir. Moreover, the DP macrophages in the submucosa, which differ from lamina propria DP macrophages, may be missed from pinch biopsy sampling, which may preclude detecting virus reservoirs for monitoring HIV cure.
Collapse
Affiliation(s)
- Naofumi Takahashi
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433
| | - Chie Sugimoto
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433
| | - Carolina Allers
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433
| | - Xavier Alvarez
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507; and
| | - Elizabeth S Didier
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433
| | - Marcelo J Kuroda
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433;
| |
Collapse
|
75
|
|
76
|
Increased Plasma Levels of the TH2 chemokine CCL18 associated with low CD4+ T cell counts in HIV-1-infected Patients with a Suppressed Viral Load. Sci Rep 2019; 9:5963. [PMID: 30979916 PMCID: PMC6461658 DOI: 10.1038/s41598-019-41588-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 02/25/2019] [Indexed: 12/15/2022] Open
Abstract
The chemokine (C-C motif) chemokine ligand 18 (CCL18) is a structural homolog of CCL3 primarily produced by monocyte-derived cells with an M2 phenotype. Elevated levels of CCL18 have been observed in several diseases associated with malignancies and chronic inflammation. The role of CCL18 in Human Immunodeficiency Virus (HIV-1) infection remains unknown. We analyzed expression levels of T helper cell-mediated (TH2) chemokines CCL18, CCL17, and CCL22 by ELISA in plasma collected from HIV-1-infected and healthy donors. In HIV-1-infected individuals, plasma viral loads were monitored by NucliSense HIV-1 QT assay and T cell counts and expression of the activation marker CD38 were determined by flow cytometry. Our data showed a significant increase in plasma levels of CCL18 in HIV-1-infected individuals compared to uninfected controls (p < 0.001) and a significant correlation between CCL18 levels and viral load in untreated patients. No significant difference of CCL18 levels was detected among the HIV-1-infected patients treated with combined antiretroviral therapy (cART) and HIV-1-untreated patients.CCL18 values are negatively correlated with CD4+CD38+ cell numbers and total CD4+ T cell counts in patients with a suppressed viral load. Notably, plasma levels of the TH2 chemokines CCL17 and CCL22 are also elevated during HIV-1 infection. However, no correlation of CCL17 and CCL22 production with CD4+ T cell counts was detected. Presented data shows that the chemokines, CCL17, CCL18, and CCL22 are increased during HIV-1 infection. However, only increased levels of CCL18, a marker of M2 macrophages, correlate with low CD4+ T cell counts in patients with suppressed viral load, raising the possibility that CCL18 and/or CCL18-producing cells may interfere with their reconstitution in HIV-1-infected patients on cART.
Collapse
|
77
|
Souriant S, Balboa L, Dupont M, Pingris K, Kviatcovsky D, Cougoule C, Lastrucci C, Bah A, Gasser R, Poincloux R, Raynaud-Messina B, Al Saati T, Inwentarz S, Poggi S, Moraña EJ, González-Montaner P, Corti M, Lagane B, Vergne I, Allers C, Kaushal D, Kuroda MJ, Sasiain MDC, Neyrolles O, Maridonneau-Parini I, Lugo-Villarino G, Vérollet C. Tuberculosis Exacerbates HIV-1 Infection through IL-10/STAT3-Dependent Tunneling Nanotube Formation in Macrophages. Cell Rep 2019; 26:3586-3599.e7. [PMID: 30917314 PMCID: PMC6733268 DOI: 10.1016/j.celrep.2019.02.091] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/08/2018] [Accepted: 02/21/2019] [Indexed: 12/13/2022] Open
Abstract
The tuberculosis (TB) bacillus, Mycobacterium tuberculosis (Mtb), and HIV-1 act synergistically; however, the mechanisms by which Mtb exacerbates HIV-1 pathogenesis are not well known. Using in vitro and ex vivo cell culture systems, we show that human M(IL-10) anti-inflammatory macrophages, present in TB-associated microenvironment, produce high levels of HIV-1. In vivo, M(IL-10) macrophages are expanded in lungs of co-infected non-human primates, which correlates with disease severity. Furthermore, HIV-1/Mtb co-infected patients display an accumulation of M(IL-10) macrophage markers (soluble CD163 and MerTK). These M(IL-10) macrophages form direct cell-to-cell bridges, which we identified as tunneling nanotubes (TNTs) involved in viral transfer. TNT formation requires the IL-10/STAT3 signaling pathway, and targeted inhibition of TNTs substantially reduces the enhancement of HIV-1 cell-to-cell transfer and overproduction in M(IL-10) macrophages. Our study reveals that TNTs facilitate viral transfer and amplification, thereby promoting TNT formation as a mechanism to be explored in TB/AIDS potential therapeutics.
Collapse
Affiliation(s)
- Shanti Souriant
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France, and Buenos Aires, Argentina
| | - Luciana Balboa
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France, and Buenos Aires, Argentina; Institute of Experimental Medicine-CONICET, National Academy of Medicine, Buenos Aires, Argentina
| | - Maeva Dupont
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France, and Buenos Aires, Argentina
| | - Karine Pingris
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Denise Kviatcovsky
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France, and Buenos Aires, Argentina; Institute of Experimental Medicine-CONICET, National Academy of Medicine, Buenos Aires, Argentina
| | - Céline Cougoule
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France, and Buenos Aires, Argentina
| | - Claire Lastrucci
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; Centre for Genomic Regulation, Barcelona, Spain
| | - Aicha Bah
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Romain Gasser
- Centre de Physiopathologie de Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Renaud Poincloux
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Brigitte Raynaud-Messina
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Talal Al Saati
- INSERM/UPS/ENVT-US006/CREFRE, Service d'Histopathologie, CHU Purpan, 31024 Toulouse, France
| | - Sandra Inwentarz
- Instituto de Tisioneumonologia "Raúl F. Vaccarezza," Universitad de Buenos Aires, Argentina
| | - Susana Poggi
- Instituto de Tisioneumonologia "Raúl F. Vaccarezza," Universitad de Buenos Aires, Argentina
| | - Eduardo Jose Moraña
- Instituto de Tisioneumonologia "Raúl F. Vaccarezza," Universitad de Buenos Aires, Argentina
| | | | - Marcelo Corti
- Division de SIDA, Hospital de Infecciosas Dr. F.J. Muñiz, Buenos Aires, Argentina
| | - Bernard Lagane
- Centre de Physiopathologie de Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Isabelle Vergne
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Carolina Allers
- Tulane National Primate Research Center, Covington, LA 70433, USA; Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Deepak Kaushal
- Tulane National Primate Research Center, Covington, LA 70433, USA; Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Marcelo J Kuroda
- Tulane National Primate Research Center, Covington, LA 70433, USA; Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Maria Del Carmen Sasiain
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France, and Buenos Aires, Argentina; Institute of Experimental Medicine-CONICET, National Academy of Medicine, Buenos Aires, Argentina
| | - Olivier Neyrolles
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France, and Buenos Aires, Argentina
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France, and Buenos Aires, Argentina
| | - Geanncarlo Lugo-Villarino
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France, and Buenos Aires, Argentina.
| | - Christel Vérollet
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France, and Buenos Aires, Argentina.
| |
Collapse
|
78
|
Expression of MDM2 in Macrophages Promotes the Early Postentry Steps of HIV-1 Infection through Inhibition of p53. J Virol 2019; 93:JVI.01871-18. [PMID: 30674627 DOI: 10.1128/jvi.01871-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/13/2019] [Indexed: 12/19/2022] Open
Abstract
The molecular basis for HIV-1 susceptibility in primary human monocyte-derived macrophages (MDMs) was previously evaluated by comparing the transcriptome of infected and bystander populations. Careful analysis of the data suggested that the ubiquitin ligase MDM2 acted as a positive regulator of HIV-1 replication in MDMs. In this study, MDM2 silencing through transcript-specific small interfering RNAs in MDMs induced a reduction in HIV-1 reverse transcription and integration along with an increase in the expression of p53-induced genes, including CDKN1A Experiments with Nutlin-3, a pharmacological inhibitor of MDM2 p53-binding activity, showed a similar effect on HIV-1 infection, suggesting that the observed restriction in HIV-1 production results from the release/activation of p53 and not the absence of MDM2 per se Knockdown and inhibition of MDM2 also both correlate with a decrease in the Thr592-phosphorylated inactive form of SAMHD1. The expression level of MDM2 and the p53 activation status are therefore important factors in the overall susceptibility of macrophages to HIV-1 infection, bringing a new understanding of signaling events controlling the process of virus replication in this cell type.IMPORTANCE Macrophages, with their long life span in vivo and their resistance to HIV-1-mediated cytopathic effect, might serve as viral reservoirs, contributing to virus persistence in an infected individual. Identification of host factors that increase the overall susceptibility of macrophages to HIV-1 might provide new therapeutic targets for the efficient control of viral replication in these cells and limit the formation of reservoirs in exposed individuals. In this study, we demonstrate the importance of p53 regulation by MDM2, which creates a cellular environment more favorable to the early steps of HIV-1 replication. Moreover, we show that p53 stabilization reduces virus infection in human macrophages, highlighting the important role of p53 in antiviral immunity.
Collapse
|
79
|
Szaniawski MA, Spivak AM, Bosque A, Planelles V. Sex Influences SAMHD1 Activity and Susceptibility to Human Immunodeficiency Virus-1 in Primary Human Macrophages. J Infect Dis 2019; 219:777-785. [PMID: 30299483 PMCID: PMC6376916 DOI: 10.1093/infdis/jiy583] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 10/04/2018] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Macrophages are major targets for HIV-1, contribute to viral propagation in vivo, and are instrumental in the pathogenesis of HAND. While it is known that host sex affects HIV-1 viremia and influences the severity of HIV-1-associated neurocognitive disease, a cellular or molecular basis for these findings remains elusive. METHODS We explored whether sex affects HIV-1 infectivity of primary human macrophages and CD4+ T cells in vitro. RESULTS Macrophages derived from female donors were less susceptible to HIV-1 infection than those derived from males. This sex-dependent difference in macrophage infectivity was independent of the requirement for CD4/CCR5-mediated virus entry and was not observed in CD4+ T cells. Investigations into the mechanism governing these sex-dependent differences revealed that the host restriction factor SAMHD1 exists in a hyperphosphorylated, less active state in male-derived macrophages. In addition, the major kinase responsible for SAMHD1 phosphorylation, CDK1, exhibited lower levels of expression in female-derived macrophages in all tested donor pairs. The sex-dependent differences in viral restriction imposed by SAMHD1 were abrogated upon its depletion. CONCLUSIONS We conclude that SAMHD1 is an essential modulator of infectivity in a sex-dependent manner in macrophages, constituting a novel component of sex differences in innate immune control of HIV-1.
Collapse
Affiliation(s)
- Matthew A Szaniawski
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine
| | - Adam M Spivak
- Department of Medicine, University of Utah School of Medicine, Salt Lake City
| | - Alberto Bosque
- Department of Microbiology Immunology and Tropical Medicine, George Washington University, Washington, District of Columbia
| | - Vicente Planelles
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine
| |
Collapse
|
80
|
Rao S, Amorim R, Niu M, Breton Y, Tremblay MJ, Mouland AJ. Host mRNA decay proteins influence HIV-1 replication and viral gene expression in primary monocyte-derived macrophages. Retrovirology 2019; 16:3. [PMID: 30732620 PMCID: PMC6367771 DOI: 10.1186/s12977-019-0465-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 01/29/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Mammalian cells harbour RNA quality control and degradative machineries such as nonsense-mediated mRNA decay that target cellular mRNAs for clearance from the cell to avoid aberrant gene expression. The role of the host mRNA decay pathways in macrophages in the context of human immunodeficiency virus type 1 (HIV-1) infection is yet to be elucidated. Macrophages are directly infected by HIV-1, mediate the dissemination of the virus and contribute to the chronic activation of the inflammatory response observed in infected individuals. Therefore, we characterized the effects of four host mRNA decay proteins, i.e., UPF1, UPF2, SMG6 and Staufen1, on viral replication in HIV-1-infected primary monocyte-derived macrophages (MDMs). RESULTS Steady-state expression levels of these host mRNA decay proteins were significantly downregulated in HIV-1-infected MDMs. Moreover, UPF2 and SMG6 inhibited HIV-1 gene expression in macrophages to a similar level achieved by SAMHD1, by directly influencing viral genomic RNA levels. Staufen1, a host protein also involved in UPF1-dependent mRNA decay and that acts at several HIV-1 replication steps, enhanced HIV-1 gene expression in MDMs. CONCLUSIONS These results provide new evidence for roles of host mRNA decay proteins in regulating HIV-1 replication in infected macrophages and can serve as potential targets for broad-spectrum antiviral therapeutics.
Collapse
Affiliation(s)
- Shringar Rao
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, Canada.,Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | - Raquel Amorim
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, Canada.,Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Meijuan Niu
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, Canada
| | - Yann Breton
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec, Québec, Canada
| | - Michel J Tremblay
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec, Québec, Canada.,Département de Microbiologie-Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Andrew J Mouland
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, Canada. .,Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada. .,Department of Medicine, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
81
|
An P, Kirk GD, Limou S, Binns-Roemer E, Kopp JB, Winkler CA. Impact of APOL1 Genetic Variants on HIV-1 Infection and Disease Progression. Front Immunol 2019; 10:53. [PMID: 30733721 PMCID: PMC6353846 DOI: 10.3389/fimmu.2019.00053] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/09/2019] [Indexed: 01/03/2023] Open
Abstract
Apolipoprotein L1 (APOL1) has broad innate immune functions and has been shown to restrict HIV replication in vitro by multiple mechanisms. Coding variants in APOL1 are strongly associated with HIV-associated nephropathy (HIVAN) in persons with untreated HIV infection; however, the mechanism by which APOL1 variant protein potentiates renal injury in the presence of high viral load is not resolved. Little is known about the association of APOL1 genotypes with HIV viral load, HIV acquisition, or progression to AIDS. We assessed the role of APOL1 coding variants on HIV-1 acquisition using the conditional logistic regression test, on viral load using the t-test or ANOVA, and on progression to AIDS using Cox proportional hazards models among African Americans enrolled in the ALIVE HIV natural history cohort (n = 775). APOL1 variants were not associated with susceptibility to HIV-1 acquisition by comparing genotype frequencies between HIV-1 positive and exposed or at-risk HIV-1 uninfected groups (recessive model, 12.8 vs. 12.5%, respectively; OR 1.02, 95% CI 0.62-1.70). Similar null results were observed for dominant and additive models. APOL1 variants were not associated with HIV-1 viral load or with risk of progression to AIDS [Relative hazards (RH) 1.33, 95% CI 0.30-5.89 and 0.96, 95% CI 0.49-1.88, for recessive and additive models, respectively]. In summary, we found no evidence that APOL1 variants are associated with host susceptibility to HIV-1 acquisition, set-point HIV-1 viral load or time to incident AIDS. These results suggest that APOL1 variants are unlikely to influence HIV infection or progression among individuals of African ancestry.
Collapse
Affiliation(s)
- Ping An
- Molecular Genetic Epidemiology Section, Basic Science Program, Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Gregory D Kirk
- Departments of Epidemiology and Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Sophie Limou
- Molecular Genetic Epidemiology Section, Basic Science Program, Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States.,CRTI UMR1064, Inserm, Université de Nantes & ITUN, CHU Nantes, Nantes, France.,Ecole Centrale de Nantes, Nantes, France
| | - Elizabeth Binns-Roemer
- Molecular Genetic Epidemiology Section, Basic Science Program, Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, United States
| | - Cheryl A Winkler
- Molecular Genetic Epidemiology Section, Basic Science Program, Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States
| |
Collapse
|
82
|
Plitnik T, Sharkey ME, Mahboubi B, Kim B, Stevenson M. Incomplete Suppression of HIV-1 by SAMHD1 Permits Efficient Macrophage Infection. Pathog Immun 2018; 3:197-223. [PMID: 30656243 PMCID: PMC6333473 DOI: 10.20411/pai.v3i2.263] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background: Sterile alpha motif and histidine/aspartic acid domain-containing protein (SAMHD1) is a dNTP triphosphorylase that reduces cellular dNTP levels in non-dividing cells, such as macrophages. Since dNTPs are required for reverse transcription, HIV-2 and most SIVs encode a Vpx protein that promotes proteasomal degradation of SAMHD1. It is unclear how HIV-1, which does not appear to harbor a SAMHD1 escape mechanism, is able to infect macrophages in the face of SAMHD1 restriction. Methods: To assess whether HIV-1 had a mechanism to negate SAMHD1 activity, we compared SAMHD1 and dNTP levels in macrophages infected by HIV-1 and SIV. We examined whether macrophages infected by HIV-1 still harbored antiviral levels of SAMHD1 by assessing their susceptibility to superinfection by vpx-deleted SIV. Finally, to assess whether HIV-1 reverse transcriptase (RT) has adapted to a low dNTP environment, we evaluated SAMHD1 sensitivity of chimeric HIV-1 and SIV variants in which the RT regions were functionally exchanged. Results: Here, we demonstrate that HIV-1 efficiently infects macrophages without modulating SAMHD1 activity or cellular dNTP levels, and that macrophages permissive to HIV-1 infection remained refractory to superinfection by vpx-deleted SIV. Furthermore, through the use of chimeric HIV/SIV, we demonstrate that the differential sensitivity of HIV-1 and SIV to SAMHD1 restriction is not dictated by RT. Conclusions: Our study reveals fundamental differences between HIV-1 and SIV in the strategy used to evade restriction by SAMHD1 and suggests a degree of resistance of HIV-1 to the antiviral environment created by SAMHD1. Understanding how these cellular restrictions antagonize viral replication will be important for the design of novel antiviral strategies.
Collapse
Affiliation(s)
- Timothy Plitnik
- Department of Microbiology & Immunology; Miller School of Medicine, University of Miami; Miami, Florida
| | - Mark E Sharkey
- Department of Medicine; Miller School of Medicine, University of Miami; Miami, Florida
| | - Bijan Mahboubi
- Department of Pediatrics, Emory University; Atlanta, Georgia.,Center for Drug Discovery, Children's Healthcare of Atlanta; Atlanta, Georgia
| | - Baek Kim
- Department of Pediatrics, Emory University; Atlanta, Georgia.,Center for Drug Discovery, Children's Healthcare of Atlanta; Atlanta, Georgia.,Department of Pharmacy, Kyung-Hee University; Seoul; South Korea
| | - Mario Stevenson
- Department of Microbiology & Immunology; Miller School of Medicine, University of Miami; Miami, Florida.,Department of Medicine; Miller School of Medicine, University of Miami; Miami, Florida
| |
Collapse
|
83
|
Carrillo J, Clotet B, Blanco J. Antibodies and Antibody Derivatives: New Partners in HIV Eradication Strategies. Front Immunol 2018; 9:2429. [PMID: 30405624 PMCID: PMC6205993 DOI: 10.3389/fimmu.2018.02429] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/02/2018] [Indexed: 12/25/2022] Open
Abstract
Promptly after primoinfection, HIV generates a pool of infected cells carrying transcriptionally silent integrated proviral DNA, the HIV-1 reservoir. These cells are not cleared by combined antiretroviral therapy (cART), and persist lifelong in treated HIV-infected individuals. Defining clinical strategies to eradicate the HIV reservoir and cure HIV-infected individuals is a major research field that requires a deep understanding of the mechanisms of seeding, maintenance and destruction of latently infected cells. Although CTL responses have been classically associated with the control of HIV replication, and hence with the size of HIV reservoir, broadly neutralizing antibodies (bNAbs) have emerged as new players in HIV cure strategies. Several reasons support this potential role: (i) over the last years a number of bNAbs with high potency and ability to cope with the extreme variability of HIV have been identified; (ii) antibodies not only block HIV replication but mediate effector functions that may contribute to the removal of infected cells and to boost immune responses against HIV; (iii) a series of new technologies have allowed for the in vitro design of improved antibodies with increased antiviral and effector functions. Recent studies in non-human primate models and in HIV-infected individuals have shown that treatment with recombinant bNAbs isolated from HIV-infected individuals is safe and may have a beneficial effect both on the seeding of the HIV reservoir and on the inhibition of HIV replication. These promising data and the development of antibody technology have paved the way for treating HIV infection with engineered monoclonal antibodies with high potency of neutralization, wide coverage of HIV diversity, extended plasma half-life in vivo and improved effector functions. The exciting effects of these newly designed antibodies in vivo, either alone or in combination with other cure strategies (latency reversing agents or therapeutic vaccines), open a new hope in HIV eradication.
Collapse
Affiliation(s)
- Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Institut de Recerca Germans Trias i Pujol, Badalona, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Institut de Recerca Germans Trias i Pujol, Badalona, Spain.,Chair in AIDS and Related Illnesses, Centre for Health and Social Care Research (CEES), Faculty of Medicine, Universitat de Vic - Universitat Central de Catalunya, Vic, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Institut de Recerca Germans Trias i Pujol, Badalona, Spain.,Chair in AIDS and Related Illnesses, Centre for Health and Social Care Research (CEES), Faculty of Medicine, Universitat de Vic - Universitat Central de Catalunya, Vic, Spain
| |
Collapse
|
84
|
Chandra PK, Gerlach SL, Wu C, Khurana N, Swientoniewski LT, Abdel-Mageed AB, Li J, Braun SE, Mondal D. Mesenchymal stem cells are attracted to latent HIV-1-infected cells and enable virus reactivation via a non-canonical PI3K-NFκB signaling pathway. Sci Rep 2018; 8:14702. [PMID: 30279437 PMCID: PMC6168583 DOI: 10.1038/s41598-018-32657-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 09/11/2018] [Indexed: 02/08/2023] Open
Abstract
Persistence of latent HIV-1 in macrophages (MACs) and T-helper lymphocytes (THLs) remain a major therapeutic challenge. Currently available latency reversing agents (LRAs) are not very effective in vivo. Therefore, understanding of physiologic mechanisms that dictate HIV-1 latency/reactivation in reservoirs is clearly needed. Mesenchymal stromal/stem cells (MSCs) regulate the function of immune cells; however, their role in regulating virus production from latently-infected MACs & THLs is not known. We documented that exposure to MSCs or their conditioned media (MSC-CM) rapidly increased HIV-1 p24 production from the latently-infected U1 (MAC) & ACH2 (THL) cell lines. Exposure to MSCs also increased HIV-1 long terminal repeat (LTR) directed gene expression in the MAC and THL reporter lines, U937-VRX and J-Lat (9.2), respectively. MSCs exposed to CM from U1 cells (U1-CM) showed enhanced migratory ability towards latently-infected cells and retained their latency-reactivation potential. Molecular studies showed that MSC-mediated latency-reactivation was dependent upon both the phosphatidyl inositol-3-kinase (PI3K) and nuclear factor-κB (NFκB) signaling pathways. The pre-clinically tested inhibitors of PI3K (PX-866) and NFκB (CDDO-Me) suppressed MSC-mediated HIV-1 reactivation. Furthermore, coexposure to MSC-CM enhanced the latency-reactivation efficacy of the approved LRAs, vorinostat and panobinostat. Our findings on MSC-mediated latency-reactivation may provide novel strategies against persistent HIV-1 reservoirs.
Collapse
Affiliation(s)
- Partha K Chandra
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Samantha L Gerlach
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Chengxiang Wu
- Tulane National Primate Research Center, Covington, LA, USA
| | - Namrata Khurana
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA.,Department of Urology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Asim B Abdel-Mageed
- Department of Urology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jian Li
- Tulane School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Stephen E Braun
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA.,Tulane National Primate Research Center, Covington, LA, USA
| | - Debasis Mondal
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
85
|
Kandathil AJ, Sugawara S, Goyal A, Durand CM, Quinn J, Sachithanandham J, Cameron AM, Bailey JR, Perelson AS, Balagopal A. No recovery of replication-competent HIV-1 from human liver macrophages. J Clin Invest 2018; 128:4501-4509. [PMID: 30198905 PMCID: PMC6159970 DOI: 10.1172/jci121678] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022] Open
Abstract
Long-lived HIV-1 reservoirs that persist despite antiretroviral therapy (ART) are a major impediment to a cure for HIV-1. We examined whether human liver macrophages (LMs), the largest tissue macrophage population, comprise an HIV-1 reservoir. We purified LMs from liver explants and included treatment with a T cell immunotoxin to reduce T cells to 1% or less. LMs were purified from 9 HIV-1-infected persons, 8 of whom were on ART (range 8-140 months). Purified LMs were stimulated ex vivo and supernatants from 6 of 8 LMs from persons on ART transmitted infection. However, HIV-1 propagation from LMs was not sustained except in LMs from 1 person taking ART for less than 1 year. Bulk liver sequences matched LM-derived HIV-1 in 5 individuals. Additional in vitro experiments undertaken to quantify the decay of HIV-1-infected LMs from 3 healthy controls showed evidence of infection and viral release for prolonged durations (>170 days). Released HIV-1 propagated robustly in target cells, demonstrating that viral outgrowth was observable using our methods. The t1/2 of HIV-1-infected LMs ranged from 3.8-55 days. These findings suggest that while HIV-1 persists in LMs during ART, it does so in forms that are inert, suggesting that they are defective or restricted with regard to propagation.
Collapse
Affiliation(s)
| | - Sho Sugawara
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ashish Goyal
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | | | - Jeffrey Quinn
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Andrew M. Cameron
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Justin R. Bailey
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alan S. Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Ashwin Balagopal
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
86
|
Poultsidi A, Dimopoulos Y, He TF, Chavakis T, Saloustros E, Lee PP, Petrovas C. Lymph Node Cellular Dynamics in Cancer and HIV: What Can We Learn for the Follicular CD4 (Tfh) Cells? Front Immunol 2018; 9:2233. [PMID: 30319664 PMCID: PMC6170630 DOI: 10.3389/fimmu.2018.02233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 09/07/2018] [Indexed: 12/17/2022] Open
Abstract
Lymph nodes (LNs) are central in the generation of adaptive immune responses. Follicular helper CD4 T (Tfh) cells, a highly differentiated CD4 population, provide critical help for the development of antigen-specific B cell responses within the germinal center. Throughout the past decade, numerous studies have revealed the important role of Tfh cells in Human Immunodeficiency Virus (HIV) pathogenesis as well as in the development of neutralizing antibodies post-infection and post-vaccination. It has also been established that tumors influence various immune cell subsets not only in their proximity, but also in draining lymph nodes. The role of local or tumor associated lymph node Tfh cells in disease progression is emerging. Comparative studies of Tfh cells in chronic infections and cancer could therefore provide novel information with regards to their differentiation plasticity and to the mechanisms regulating their development.
Collapse
Affiliation(s)
- Antigoni Poultsidi
- Department of Surgery, Medical School, University of Thessaly, Larissa, Greece
| | - Yiannis Dimopoulos
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, United States
| | - Ting-Fang He
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Emmanouil Saloustros
- Department of Internal Medicine, Medical School, University of Thessaly, Larissa, Greece
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, United States
| |
Collapse
|
87
|
Graziano F, Aimola G, Forlani G, Turrini F, Accolla RS, Vicenzi E, Poli G. Reversible Human Immunodeficiency Virus Type-1 Latency in Primary Human Monocyte-Derived Macrophages Induced by Sustained M1 Polarization. Sci Rep 2018; 8:14249. [PMID: 30250078 PMCID: PMC6155284 DOI: 10.1038/s41598-018-32451-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 08/13/2018] [Indexed: 01/08/2023] Open
Abstract
We have reported that short-term stimulation of primary human monocyte-derived macrophages (MDM) with interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α), i.e. M1 polarization, leads to a significant containment of virus replication. Here we show that M1-MDM restimulation with these cytokines 7 days after infection (M12 MDM) promoted an increased restriction of HIV-1 replication characterized by very low levels of virus production near to undetectable levels. In comparison to control and M1-MDM that were not restimulated, M12 MDM showed a stronger reduction of both total and integrated HIV DNA as well as of viral mRNA expression. M12 MDM were characterized by an upregulated expression of restriction factors acting at the level of reverse transcription (RT), including apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3A (APOBEC3A) and APOBEC3G, but not SAM domain and HD domain-containing protein 1 (SAMHD1). M12 MDM also showed an increased expression of Class II Transactivator (CIITA) and Tripartite Motif22 (TRIM22), two negative regulators of proviral transcription, whereas expression and phosphorylation of transcriptional inducers of HIV-1, such as nuclear factor kB (NF-kB) and signal transducer and activator of transcription 1 (STAT1), were not impaired in these cells. The almost quiescent state of the infection in M12 MDM was promptly reversed by coculture with mitogen-stimulated leukocytes or cell incubation with their filtered culture supernatant. M12 MDM harbored replication-competent HIV-1 as virus spreading following cell stimulation was fully prevented by the RT inhibitor lamivudine/3TC. Selective reactivation of proviral expression in M12 MDM, but not in control or in M1-MDM that were not restimulated, was confirmed in cells infected with single round Vesicular Stomatitis Virus-G-pseudotyped HIV-1. Thus, M12 MDM represent an in vitro model of reversible, almost quiescent HIV-1 infection of primary human macrophages that could be further exploited for “Cure” related investigations.
Collapse
Affiliation(s)
- Francesca Graziano
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milano, Italy.,Institute Curie Laboratoire Immunité et Cancer - INSERM U932, 26 rue d'Ulm, 75248, Paris cedex 05, Paris, France
| | - Giulia Aimola
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milano, Italy
| | - Greta Forlani
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Filippo Turrini
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milano, Italy
| | - Roberto S Accolla
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Elisa Vicenzi
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milano, Italy
| | - Guido Poli
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milano, Italy. .,Vita-Salute San Raffaele University School of Medicine, Milano, Italy.
| |
Collapse
|
88
|
Lidofsky A, Holmes JA, Feeney ER, Kruger AJ, Salloum S, Zheng H, Seguin IS, Altinbas A, Masia R, Corey KE, Gustafson JL, Schaefer EA, Hunt PW, Deeks S, Somsouk M, Chew KW, Chung RT, Alatrakchi N. Macrophage Activation Marker Soluble CD163 Is a Dynamic Marker of Liver Fibrogenesis in Human Immunodeficiency Virus/Hepatitis C Virus Coinfection. J Infect Dis 2018; 218:1394-1403. [PMID: 29868909 PMCID: PMC6151081 DOI: 10.1093/infdis/jiy331] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
Background Coinfection with human immunodeficiency virus (HIV) accelerates hepatitis C virus (HCV)-related liver fibrosis. Macrophages are triggered during both viral infections and are critical in liver inflammation/fibrogenesis. Liver fibrosis strongly associates with serum soluble CD163 (sCD163, a macrophage activation marker); comprehensive evaluation in HIV/HCV coinfection is lacking. Methods We retrospectively analyzed sCD163 (enzyme-linked immunosorbent assay) and hepatic CD163 (immunofluorescent CD163/CD68 costaining) in patients infected with HIV/HCV, HCV, or HIV, pre- and post-antiviral therapy. Results sCD163 was significantly higher in HIV/HCV compared to either monoinfection, and decreased following successful antiviral therapy, although did not fully normalize. In HIV/HCV, sCD163 was associated with necroinflammation, Ishak fibrosis scores, and noninvasive fibrosis scores. We observed a novel trend whereby sCD163 levels progressively increase with increasing Ishak fibrosis score, peaking at stage 4, above which levels plateaued. Periportal CD163+ macrophage frequency was also higher with increasing fibrosis score. When stratified by fibrosis stage, sCD163 levels were higher in HIV/HCV than HCV but only in individuals with mild to moderate fibrosis. Conclusions In HIV/HCV, increasing sCD163 levels accompanied periportal CD163+ macrophage enrichment in mild to moderate fibrosis, but not in established cirrhosis, suggesting that sCD163 is a dynamic biomarker of fibrogenesis rather than accumulated fibrosis. Our findings implicate HIV-related macrophage activation in accelerated fibrosis progression in HIV/HCV coinfection.
Collapse
Affiliation(s)
- Anna Lidofsky
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jacinta A Holmes
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Eoin R Feeney
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- HIV Molecular Research Group, University College of Dublin, Ireland
| | - Annie J Kruger
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Shadi Salloum
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hui Zheng
- Biostatistics Center, Massachusetts General Hospital, Boston
| | - Isabel S Seguin
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Akif Altinbas
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ricard Masia
- Department of Pathology, Massachusetts General Hospital, Boston
| | - Kathleen E Corey
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jenna L Gustafson
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Esperance A Schaefer
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Peter W Hunt
- Department of Medicine, University of California, San Francisco
| | - Steven Deeks
- Department of Medicine, University of California, San Francisco
| | - Ma Somsouk
- Department of Medicine, University of California, San Francisco
| | - Kara W Chew
- Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles
| | - Raymond T Chung
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nadia Alatrakchi
- Liver Center, Gastroenterology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
89
|
Benzo(a)pyrene in Cigarette Smoke Enhances HIV-1 Replication through NF-κB Activation via CYP-Mediated Oxidative Stress Pathway. Sci Rep 2018; 8:10394. [PMID: 29991690 PMCID: PMC6039513 DOI: 10.1038/s41598-018-28500-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023] Open
Abstract
Smoking aggravates HIV-1 pathogenesis and leads to decreased responses to antiretroviral therapy. In this study, we aim to find a molecular mechanism that would explain smoking-induced HIV-1 replication. Benzo(a)pyrene (BaP), a major carcinogen in cigarette, requires metabolic activation through cytochrome P450s (CYPs) to exert its toxic effects. We hypothesized that CYP-mediated BaP metabolism generates reactive oxygen species (ROS), and the resultant oxidative stress aggravates HIV-1 replication. As expected, we observed ~3 to 4-fold increase in HIV-1 replication in U1 cells and human primary macrophages after chronic BaP exposure. We also observed ~30-fold increase in the expression of CYP1A1 at mRNA level, ~2.5-fold increase in its enzymatic activity as well as elevated ROS and cytotoxicity in U1 cells. The knock-down of the CYP1A1 gene using siRNA and treatment with selective CYP inhibitors and antioxidants significantly reduced HIV-1 replication. Further, we observed a nuclear translocation of NF-κB subunits (p50 and p65) after chronic BaP exposure, which was reduced by treatment with siRNA and antioxidants/CYP inhibitors. Suppression of NF-κB pathway using specific NF-κB inhibitors also significantly reduced HIV-1 replication. Altogether, our results suggest that BaP enhances HIV-1 replication in macrophages by a CYP-mediated oxidative stress pathway followed by the NF-κB pathway.
Collapse
|
90
|
Trifone C, Salido J, Ruiz MJ, Leng L, Quiroga MF, Salomón H, Bucala R, Ghiglione Y, Turk G. Interaction Between Macrophage Migration Inhibitory Factor and CD74 in Human Immunodeficiency Virus Type I Infected Primary Monocyte-Derived Macrophages Triggers the Production of Proinflammatory Mediators and Enhances Infection of Unactivated CD4 + T Cells. Front Immunol 2018; 9:1494. [PMID: 29997630 PMCID: PMC6030361 DOI: 10.3389/fimmu.2018.01494] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 06/15/2018] [Indexed: 11/13/2022] Open
Abstract
Understanding the mechanisms of human immunodeficiency virus type I (HIV-1) pathogenesis would facilitate the identification of new therapeutic targets to control the infection in face of current antiretroviral therapy limitations. CD74 membrane expression is upregulated in HIV-1-infected cells and the magnitude of its modulation correlates with immune hyperactivation in HIV-infected individuals. In addition, plasma level of the CD74 activating ligand macrophage migration inhibitory factor (MIF) is increased in infected subjects. However, the role played by MIF/CD74 interaction in HIV pathogenesis remains unexplored. Here, we studied the effect of MIF/CD74 interaction on primary HIV-infected monocyte-derived macrophages (MDMs) and its implications for HIV immunopathogenesis. Confocal immunofluorescence analysis of CD74 and CD44 (the MIF signal transduction co-receptor) expression indicated that both molecules colocalized at the plasma membrane specifically in wild-type HIV-infected MDMs. Treatment of infected MDMs with MIF resulted in an MIF-dependent increase in TLR4 expression. Similarly, there was a dose-dependent increase in the production of IL-6, IL-8, TNFα, IL-1β, and sICAM compared to the no-MIF condition, specifically from infected MDMs. Importantly, the effect observed on IL-6, IL-8, TNFα, and IL-1β was abrogated by impeding MIF interaction with CD74. Moreover, the use of a neutralizing αMIF antibody or an MIF antagonist reverted these effects, supporting the specificity of the results. Treatment of unactivated CD4+ T-cells with MIF-treated HIV-infected MDM-derived culture supernatants led to enhanced permissiveness to HIV-1 infection. This effect was lost when CD4+ T-cells were treated with supernatants derived from infected MDMs in which CD74/MIF interaction had been blocked. Moreover, the enhanced permissiveness of unactivated CD4+ T-cells was recapitulated by exogenous addition of IL-6, IL-8, IL-1β, and TNFα, or abrogated by neutralizing its biological activity using specific antibodies. Results obtained with BAL and NL4-3 HIV laboratory strains were reproduced using transmitted/founder primary isolates. This evidence indicated that MIF/CD74 interaction resulted in a higher production of proinflammatory cytokines from HIV-infected MDMs. This caused the generation of an inflammatory microenvironment which predisposed unactivated CD4+ T-cells to HIV-1 infection, which might contribute to viral spreading and reservoir seeding. Overall, these results support a novel role of the MIF/CD74 axis in HIV pathogenesis that deserves further investigation.
Collapse
Affiliation(s)
- César Trifone
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Jimena Salido
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - María Julia Ruiz
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Lin Leng
- Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - María Florencia Quiroga
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Horacio Salomón
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Richard Bucala
- Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Yanina Ghiglione
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Gabriela Turk
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| |
Collapse
|
91
|
Temerozo JR, de Azevedo SSD, Insuela DBR, Vieira RC, Ferreira PLC, Carvalho VF, Bello G, Bou-Habib DC. The Neuropeptides Vasoactive Intestinal Peptide and Pituitary Adenylate Cyclase-Activating Polypeptide Control HIV-1 Infection in Macrophages Through Activation of Protein Kinases A and C. Front Immunol 2018; 9:1336. [PMID: 29951068 PMCID: PMC6008521 DOI: 10.3389/fimmu.2018.01336] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/29/2018] [Indexed: 12/19/2022] Open
Abstract
Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are highly similar neuropeptides present in several tissues, endowed with immunoregulatory functions and other systemic effects. We previously reported that both neuropeptides reduce viral production in HIV-1-infected primary macrophages, with the participation of β-chemokines and IL-10, and now we describe molecular mechanisms engaged in this activity. Macrophages exposed to VIP or PACAP before HIV-1 infection showed resistance to viral replication, comparable to that observed when the cells were treated after infection. Also, multiple treatments with a suboptimal dose of VIP or PACAP after macrophage infection resulted in a decline of virus production similar to the inhibition promoted by a single exposure to the optimal inhibitory concentration. Cellular signaling pathways involving cAMP production and activation of protein kinases A and C were critical components of the VIP and PACAP anti-HIV-1 effects. Analysis of the transcription factors and the transcriptional/cell cycle regulators showed that VIP and PACAP induced cAMP response element-binding protein activation, inhibited NF-kB, and reduced Cyclin D1 levels in HIV-1-infected cells. Remarkably, VIP and PACAP promoted G-to-A mutations in the HIV-1 provirus, matching those derived from the activity of the APOBEC family of viral restriction factors, and reduced viral infectivity. In conclusion, our findings strengthen the antiretroviral potential of VIP and PACAP and point to new therapeutic approaches to control the progression of HIV-1 infection.
Collapse
Affiliation(s)
- Jairo R Temerozo
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Suwellen S D de Azevedo
- Laboratory of AIDS and Molecular Immunology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Daniella B R Insuela
- Laboratory of Inflammation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Rhaíssa C Vieira
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Pedro L C Ferreira
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Vinícius F Carvalho
- National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil.,Laboratory of Inflammation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Gonzalo Bello
- Laboratory of AIDS and Molecular Immunology, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Dumith Chequer Bou-Habib
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
92
|
Host MicroRNAs-221 and -222 Inhibit HIV-1 Entry in Macrophages by Targeting the CD4 Viral Receptor. Cell Rep 2018; 21:141-153. [PMID: 28978468 DOI: 10.1016/j.celrep.2017.09.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/18/2017] [Accepted: 09/07/2017] [Indexed: 12/19/2022] Open
Abstract
Macrophages are heterogeneous immune cells with distinct origins, phenotypes, functions, and tissue localization. Their susceptibility to HIV-1 is subject to variations from permissiveness to resistance, owing in part to regulatory microRNAs. Here, we used RNA sequencing (RNA-seq) to examine the expression of >400 microRNAs in productively infected and bystander cells of HIV-1-exposed macrophage cultures. Two microRNAs upregulated in bystander macrophages, miR-221 and miR-222, were identified as negative regulators of CD4 expression and CD4-mediated HIV-1 entry. Both microRNAs were enhanced by tumor necrosis factor alpha (TNF-α), an inhibitor of CD4 expression. MiR-221/miR-222 inhibitors recovered HIV-1 entry in TNF-α-treated macrophages by enhancing CD4 expression and increased HIV-1 replication and spread in macrophages by countering TNF-α-enhanced miR-221/miR-222 expression in bystander cells. In line with these findings, HIV-1-resistant intestinal myeloid cells express higher levels of miR-221 than peripheral blood monocytes. Thus, miR-221/miR-222 act as effectors of the antiviral host response activated during macrophage infection that restrict HIV-1 entry.
Collapse
|
93
|
SAMHD1 Phosphorylation Coordinates the Anti-HIV-1 Response by Diverse Interferons and Tyrosine Kinase Inhibition. mBio 2018; 9:mBio.00819-18. [PMID: 29764952 PMCID: PMC5954222 DOI: 10.1128/mbio.00819-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Macrophages are susceptible to human immunodeficiency virus type 1 (HIV-1) infection despite abundant expression of antiviral proteins. Perhaps the most important antiviral protein is the restriction factor sterile alpha motif domain and histidine/aspartic acid domain-containing protein 1 (SAMHD1). We investigated the role of SAMHD1 and its phospho-dependent regulation in the context of HIV-1 infection in primary human monocyte-derived macrophages and the ability of various interferons (IFNs) and pharmacologic agents to modulate SAMHD1. Here we show that stimulation by type I, type II, and to a lesser degree, type III interferons share activation of SAMHD1 via dephosphorylation at threonine-592 as a consequence of signaling. Cyclin-dependent kinase 1 (CDK1), a known effector kinase for SAMHD1, was downregulated at the protein level by all IFN types tested. Pharmacologic inhibition or small interfering RNA (siRNA)-mediated knockdown of CDK1 phenocopied the effects of IFN on SAMHD1. A panel of FDA-approved tyrosine kinase inhibitors potently induced activation of SAMHD1 and subsequent HIV-1 inhibition. The viral restriction imposed via IFNs or dasatinib could be overcome through depletion of SAMHD1, indicating that their effects are exerted primarily through this pathway. Our results demonstrate that SAMHD1 activation, but not transcriptional upregulation or protein induction, is the predominant mechanism of HIV-1 restriction induced by type I, type II, and type III IFN signaling in macrophages. Furthermore, SAMHD1 activation presents a pharmacologically actionable target through which HIV-1 infection can be subverted. Our experimental results demonstrate that SAMHD1 dephosphorylation at threonine-592 represents a central mechanism of HIV-1 restriction that is common to the three known families of IFNs. While IFN types I and II were potent inhibitors of HIV-1, type III IFN showed modest to undetectable activity. Regulation of SAMHD1 by IFNs involved changes in phosphorylation status but not in protein levels. Phosphorylation of SAMHD1 in macrophages occurred at least in part via CDK1. Tyrosine kinase inhibitors similarly induced SAMHD1 dephosphorylation, which protects macrophages from HIV-1 in a SAMHD1-dependent manner. SAMHD1 is a critical restriction factor regulating HIV-1 infection of macrophages.
Collapse
|
94
|
Clayton KL, Collins DR, Lengieza J, Ghebremichael M, Dotiwala F, Lieberman J, Walker BD. Resistance of HIV-infected macrophages to CD8 + T lymphocyte-mediated killing drives activation of the immune system. Nat Immunol 2018; 19:475-486. [PMID: 29670239 PMCID: PMC6025741 DOI: 10.1038/s41590-018-0085-3] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 03/13/2018] [Indexed: 12/19/2022]
Abstract
CD4+ T lymphocytes are the principal target of human immunodeficiency virus (HIV), but infected macrophages also contribute to viral pathogenesis. The killing of infected cells by CD8+ cytotoxic T lymphocytes (CTLs) leads to control of viral replication. Here we found that the killing of macrophages by CTLs was impaired relative to the killing of CD4+ T cells by CTLs, and this resulted in inefficient suppression of HIV. The killing of macrophages depended on caspase-3 and granzyme B, whereas the rapid killing of CD4+ T cells was caspase independent and did not require granzyme B. Moreover, the impaired killing of macrophages was associated with prolonged effector cell-target cell contact time and higher expression of interferon-γ by CTLs, which induced macrophage production of pro-inflammatory chemokines that recruited monocytes and T cells. Similar results were obtained when macrophages presented other viral antigens, suggestive of a general mechanism for macrophage persistence as antigen-presenting cells that enhance inflammation and adaptive immunity. Inefficient killing of macrophages by CTLs might contribute to chronic inflammation, a hallmark of chronic disease caused by HIV.
Collapse
Affiliation(s)
| | - David R Collins
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Josh Lengieza
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | | | - Farokh Dotiwala
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Bruce D Walker
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA. .,Howard Hughes Medical Institute, Chevy Chase, MD, USA. .,Institute of Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
95
|
Vicenzi E, Poli G. The interferon-stimulated gene TRIM22: A double-edged sword in HIV-1 infection. Cytokine Growth Factor Rev 2018; 40:40-47. [PMID: 29650252 DOI: 10.1016/j.cytogfr.2018.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/07/2018] [Indexed: 12/17/2022]
Abstract
Infection of target cells by the human immunodeficiency virus type-1 (HIV-1) is hampered by constitutively expressed host cell proteins preventing or curtailing virus replication and therefore defined as "restriction factors". Among them, members of the tripartite motif (TRIM) family have emerged as important players endowed with both antiviral effects and modulatory capacity of the innate immune response. TRIM5α and TRIM19 (i.e. promyelocytic leukemia, PML) are among the best-characterized family members; however, in this review we will focus on the potential role of another family member, i.e. TRIM22, a factor strongly induced by interferon stimulation, in HIV infection in vivo and in vitro in the context of its broader antiviral effects. We will also focus on the potential role of TRIM22 in HIV-1-infected individuals speculating on its dual role in controlling virus replication and more complex role in chronic infection. At the molecular levels, we will review the evidence in favor of a relevant role of TRIM22 as epigenetic inhibitor of HIV-1 transcription acting by preventing the binding of the host cell transcription factor Sp1 to the viral promoter. These evidences suggest that TRIM22 should be considered a potential new player in either the establishment or maintenance of HIV-1 reservoirs of latently infected cells unaffected by combination antiretroviral therapy.
Collapse
Affiliation(s)
- Elisa Vicenzi
- Viral Pathogens and Biosafety Unit, P2-P3 Laboratories, DIBIT, Via Olgettina n. 58, 20132, Milano, Italy.
| | - Guido Poli
- AIDS Immunopathogenesis Unit, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| |
Collapse
|
96
|
Abstract
Lymph nodes play a central role in the development of adaptive immunity against pathogens and particularly the generation of antigen-specific B cell responses in specialized areas called germinal centers (GCs). Lymph node (LN) pathology was recognized as an important consequence of human immunodeficiency virus (HIV) infection since the beginning of the HIV epidemic. Investigation into the structural and functional alterations induced by HIV and Simian immunodeficiency virus (SIV) has further cemented the central role that lymphoid tissue plays in HIV/SIV pathogenesis. The coexistence of constant local inflammation, altered tissue architecture, and relative exclusion of virus-specific CD8 T cells from the GCs creates a unique environment for the virus evolution and establishment of viral reservoir in specific GC cells, namely T follicular helper CD4 T cells (Tfh). A better understanding of the biology of immune cells in HIV-infected lymph nodes is a prerequisite to attaining the ultimate goal of complete viral eradication.
Collapse
Affiliation(s)
- Yiannis Dimopoulos
- Tissue Analysis Core, Vaccine Research Center, NIAID, NIH, 40 Convent Drive, MSC 3022, Building 40, Room 3612B, Bethesda, MD, 20892, USA
| | - Eirini Moysi
- Tissue Analysis Core, Vaccine Research Center, NIAID, NIH, 40 Convent Drive, MSC 3022, Building 40, Room 3612B, Bethesda, MD, 20892, USA
| | - Constantinos Petrovas
- Tissue Analysis Core, Vaccine Research Center, NIAID, NIH, 40 Convent Drive, MSC 3022, Building 40, Room 3612B, Bethesda, MD, 20892, USA.
| |
Collapse
|
97
|
Bone degradation machinery of osteoclasts: An HIV-1 target that contributes to bone loss. Proc Natl Acad Sci U S A 2018; 115:E2556-E2565. [PMID: 29463701 DOI: 10.1073/pnas.1713370115] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bone deficits are frequent in HIV-1-infected patients. We report here that osteoclasts, the cells specialized in bone resorption, are infected by HIV-1 in vivo in humanized mice and ex vivo in human joint biopsies. In vitro, infection of human osteoclasts occurs at different stages of osteoclastogenesis via cell-free viruses and, more efficiently, by transfer from infected T cells. HIV-1 infection markedly enhances adhesion and osteolytic activity of human osteoclasts by modifying the structure and function of the sealing zone, the osteoclast-specific bone degradation machinery. Indeed, the sealing zone is broader due to F-actin enrichment of its basal units (i.e., the podosomes). The viral protein Nef is involved in all HIV-1-induced effects partly through the activation of Src, a regulator of podosomes and of their assembly as a sealing zone. Supporting these results, Nef-transgenic mice exhibit an increased osteoclast density and bone defects, and osteoclasts derived from these animals display high osteolytic activity. Altogether, our study evidences osteoclasts as host cells for HIV-1 and their pathological contribution to bone disorders induced by this virus, in part via Nef.
Collapse
|
98
|
Taylor JP, Cash MN, Santostefano KE, Nakanishi M, Terada N, Wallet MA. CRISPR/Cas9 knockout of USP18 enhances type I IFN responsiveness and restricts HIV-1 infection in macrophages. J Leukoc Biol 2018; 103:1225-1240. [PMID: 29437254 PMCID: PMC6754309 DOI: 10.1002/jlb.3mia0917-352r] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/14/2017] [Accepted: 01/05/2018] [Indexed: 12/11/2022] Open
Abstract
The IFN-stimulated gene ubiquitin-specific proteinase 18 (USP18) encodes a protein that negatively regulates T1 IFN signaling via stearic inhibition of JAK1 recruitment to the IFN-α receptor 2 subunit (IFNAR2). Here, we demonstrate that USP18 expression is induced by HIV-1 in a T1 IFN-dependent manner. Experimental depletion of USP18 by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) gene editing results in a significant restriction of HIV-1 replication in an induced pluripotent stem cell (iPSC)-derived macrophage model. In the absence of USP18, macrophages have increased responsiveness to stimulation with T1 IFNs with prolonged phosphorylation of STAT1 and STAT2 and increased expression of IFN-stimulated genes that are key for antiviral responses. Interestingly, HIV-1 requires some signaling through the T1 IFN receptor to replicate efficiently because a neutralizing antibody that inhibits T1 IFN activity reduces HIV-1 replication rate in monocyte-derived macrophages. USP18 induction by HIV-1 tunes the IFN response to optimal levels allowing for efficient transcription from the HIV-1 LTR promoter while minimizing the T1 IFN-induced antiviral response that would otherwise restrict viral replication and spread. Finally, iPSC and CRISPR/Cas9 gene targeting offer a powerful tool to study host factors that regulate innate immune responses.
Collapse
Affiliation(s)
- Jared P. Taylor
- Department of PathologyImmunology & Laboratory MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Melanie N. Cash
- Department of PathologyImmunology & Laboratory MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Katherine E. Santostefano
- Department of PathologyImmunology & Laboratory MedicineUniversity of FloridaGainesvilleFloridaUSA
- Center for Cellular ReprogrammingUniversity of FloridaGainesvilleFloridaUSA
| | - Mahito Nakanishi
- Research Center for Stem Cell EngineeringNational Institute of Advanced Industrial Science and TechnologyTsukubaIbarakiJapan
| | - Naohiro Terada
- Department of PathologyImmunology & Laboratory MedicineUniversity of FloridaGainesvilleFloridaUSA
- Center for Cellular ReprogrammingUniversity of FloridaGainesvilleFloridaUSA
| | - Mark A. Wallet
- Department of PathologyImmunology & Laboratory MedicineUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
99
|
The Biology of Monocytes and Dendritic Cells: Contribution to HIV Pathogenesis. Viruses 2018; 10:v10020065. [PMID: 29415518 PMCID: PMC5850372 DOI: 10.3390/v10020065] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 02/07/2023] Open
Abstract
Myeloid cells such as monocytes, dendritic cells (DC) and macrophages (MΦ) are key components of the innate immune system contributing to the maintenance of tissue homeostasis and the development/resolution of immune responses to pathogens. Monocytes and DC, circulating in the blood or infiltrating various lymphoid and non-lymphoid tissues, are derived from distinct bone marrow precursors and are typically short lived. Conversely, recent studies revealed that subsets of tissue resident MΦ are long-lived as they originate from embryonic/fetal precursors that have the ability to self-renew during the life of an individual. Pathogens such as the human immunodeficiency virus type 1 (HIV-1) highjack the functions of myeloid cells for viral replication (e.g., MΦ) or distal dissemination and cell-to-cell transmission (e.g., DC). Although the long-term persistence of HIV reservoirs in CD4+ T-cells during viral suppressive antiretroviral therapy (ART) is well documented, the ability of myeloid cells to harbor replication competent viral reservoirs is still a matter of debate. This review summarizes the current knowledge on the biology of monocytes and DC during homeostasis and in the context of HIV-1 infection and highlights the importance of future studies on long-lived resident MΦ to HIV persistence in ART-treated patients.
Collapse
|
100
|
Baxter AE, O'Doherty U, Kaufmann DE. Beyond the replication-competent HIV reservoir: transcription and translation-competent reservoirs. Retrovirology 2018; 15:18. [PMID: 29394935 PMCID: PMC5797386 DOI: 10.1186/s12977-018-0392-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/08/2018] [Indexed: 12/20/2022] Open
Abstract
Recent years have seen a substantial increase in the number of tools available to monitor and study HIV reservoirs. Here, we discuss recent technological advances that enable an understanding of reservoir dynamics beyond classical assays to measure the frequency of cells containing provirus able to propagate a spreading infection (replication-competent reservoir). Specifically, we focus on the characterization of cellular reservoirs containing proviruses able to transcribe viral mRNAs (so called transcription-competent) and translate viral proteins (translation-competent). We suggest that the study of these alternative reservoirs provides complementary information to classical approaches, crucially at a single-cell level. This enables an in-depth characterization of the cellular reservoir, both following reactivation from latency and, importantly, directly ex vivo at baseline. Furthermore, we propose that the study of cellular reservoirs that may not contain fully replication-competent virus, but are able to produce HIV mRNAs and proteins, is of biological importance. Lastly, we detail some of the key contributions that the study of these transcription and translation-competent reservoirs has made thus far to investigations into HIV persistence, and outline where these approaches may take the field next.
Collapse
Affiliation(s)
- Amy E Baxter
- CR-CHUM, Université de Montréal, Montréal, QC, Canada.,Scripps CHAVI-ID, La Jolla, CA, USA
| | - Una O'Doherty
- Department of Pathology and Laboratory Medicine, Division of Transfusion Medicine and Therapeutic Pathology, University of Pennsylvania, Philadelphia, PA, USA.
| | - Daniel E Kaufmann
- CR-CHUM, Université de Montréal, Montréal, QC, Canada. .,Scripps CHAVI-ID, La Jolla, CA, USA.
| |
Collapse
|