1
|
Clain JA, Picard M, Rabezanahary H, André S, Boutrais S, Goma Matsetse E, Dewatines J, Dueymes Q, Thiboutot E, Racine G, Soundaramourty C, Mammano F, Corbeau P, Zghidi-Abouzid O, Estaquier J. Immune Alterations and Viral Reservoir Atlas in SIV-Infected Chinese Rhesus Macaques. Infect Dis Rep 2025; 17:12. [PMID: 39997464 PMCID: PMC11855486 DOI: 10.3390/idr17010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Over the last decades, our projects have been dedicated to clarifying immunopathological and virological events associated with Human Immunodeficiency Virus (HIV) infection. METHODS By using non-human primate models of pathogenic and non-pathogenic lentiviral infections, we aimed at identifying the cells and tissues in which the virus persists, despite antiretroviral therapy (ART). Indeed, the eradication of viral reservoirs is a major challenge for HIV cure. RESULTS We present a series of results performed in rhesus macaques of Chinese origin deciphering the virological and immunological events associated with ART that can be of interest for people living with HIV. CONCLUSIONS This model could be of interest for understanding in whole body the clinical alteration that persist despite ART.
Collapse
Affiliation(s)
- Julien A. Clain
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Morgane Picard
- Institut national de la santé et de la recherche médicale (INSERM) U1124, Université Paris Cité, 75006 Paris, France; (M.P.); (S.A.); (C.S.); (F.M.)
| | - Henintsoa Rabezanahary
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Sonia André
- Institut national de la santé et de la recherche médicale (INSERM) U1124, Université Paris Cité, 75006 Paris, France; (M.P.); (S.A.); (C.S.); (F.M.)
| | - Steven Boutrais
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Ella Goma Matsetse
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Juliette Dewatines
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Quentin Dueymes
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Elise Thiboutot
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Gina Racine
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Calaiselvy Soundaramourty
- Institut national de la santé et de la recherche médicale (INSERM) U1124, Université Paris Cité, 75006 Paris, France; (M.P.); (S.A.); (C.S.); (F.M.)
| | - Fabrizio Mammano
- Institut national de la santé et de la recherche médicale (INSERM) U1124, Université Paris Cité, 75006 Paris, France; (M.P.); (S.A.); (C.S.); (F.M.)
- Institut national de la santé et de la recherche médicale (Inserm) U1259 MAVIVHe, Université de Tours, 37032 Tours, France
| | - Pierre Corbeau
- Institut de Génétique Humaine, CNRS-Université de Montpellier UMR9002, 34094 Montpellier, France;
| | - Ouafa Zghidi-Abouzid
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Jérôme Estaquier
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
- Institut national de la santé et de la recherche médicale (INSERM) U1124, Université Paris Cité, 75006 Paris, France; (M.P.); (S.A.); (C.S.); (F.M.)
| |
Collapse
|
2
|
Bayón-Gil Á, Martinez-Picado J, Puertas MC. Viremic non-progression in HIV/SIV infection: A tied game between virus and host. Cell Rep Med 2025; 6:101921. [PMID: 39842407 PMCID: PMC11866547 DOI: 10.1016/j.xcrm.2024.101921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/16/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025]
Abstract
High-efficacy antiretroviral treatment (ART) has been a game-changer for HIV/AIDS pandemic, but incomplete CD4+ T cell recovery and persistent chronic immune activation still affect HIV-suppressed people. Exceptional cases of HIV infection that naturally exhibit delayed disease progression provide invaluable insights into protective biological mechanisms with potential clinical application. Viremic non-progressors (VNPs) represent an extremely rare population of individuals with HIV, characterized by preservation of the CD4+ T cell compartment despite persistent high levels of viral load (>10,000 copies/mL). While only a few studies have investigated the immunovirological characteristics of adult and pediatric VNPs, most of our knowledge about this phenotype stems from its non-human-primate counterpart, the natural simian immunodeficiency virus (SIV) hosts. In this review, we synthesize the insights gained from recent studies of natural SIV hosts and VNPs and evaluate the potential similarities and differences in the mechanisms that underlie the absence of pathogenesis, with special focus on the control of immune activation.
Collapse
Affiliation(s)
- Ángel Bayón-Gil
- IrsiCaixa Immunopathology Research Institute, Badalona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa Immunopathology Research Institute, Badalona, Spain; Germans Trias i Pujol Research Institute, Badalona, Spain; CIBERINFEC, Institute of Health Carlos III, Madrid, Spain; University of Vic-Central University of Catalonia, Vic, Spain; Catalan Institution for Research and Advanced Studies, Barcelona, Spain.
| | - Maria C Puertas
- IrsiCaixa Immunopathology Research Institute, Badalona, Spain; Germans Trias i Pujol Research Institute, Badalona, Spain; CIBERINFEC, Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
De Meyer A, Meuleman P. Preclinical animal models to evaluate therapeutic antiviral antibodies. Antiviral Res 2024; 225:105843. [PMID: 38548022 DOI: 10.1016/j.antiviral.2024.105843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/25/2024] [Indexed: 04/05/2024]
Abstract
Despite the availability of effective preventative vaccines and potent small-molecule antiviral drugs, effective non-toxic prophylactic and therapeutic measures are still lacking for many viruses. The use of monoclonal and polyclonal antibodies in an antiviral context could fill this gap and provide effective virus-specific medical interventions. In order to develop these therapeutic antibodies, preclinical animal models are of utmost importance. Due to the variability in viral pathogenesis, immunity and overall characteristics, the most representative animal model for human viral infection differs between virus species. Therefore, throughout the years researchers sought to find the ideal preclinical animal model for each virus. The most used animal models in preclinical research include rodents (mice, ferrets, …) and non-human primates (macaques, chimpanzee, ….). Currently, antibodies are tested for antiviral efficacy against a variety of viruses including different hepatitis viruses, human immunodeficiency virus (HIV), influenza viruses, respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and rabies virus. This review provides an overview of the current knowledge about the preclinical animal models that are used for the evaluation of therapeutic antibodies for the abovementioned viruses.
Collapse
Affiliation(s)
- Amse De Meyer
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Philip Meuleman
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
4
|
Costa RL, Gadelha L, D'arc M, Ribeiro-Alves M, Robertson DL, Schwartz JM, Soares MA, Porto F. HIHISIV: a database of gene expression in HIV and SIV host immune response. BMC Bioinformatics 2024; 25:125. [PMID: 38519883 PMCID: PMC10958971 DOI: 10.1186/s12859-024-05740-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 03/11/2024] [Indexed: 03/25/2024] Open
Abstract
In the battle of the host against lentiviral pathogenesis, the immune response is crucial. However, several questions remain unanswered about the interaction with different viruses and their influence on disease progression. The simian immunodeficiency virus (SIV) infecting nonhuman primates (NHP) is widely used as a model for the study of the human immunodeficiency virus (HIV) both because they are evolutionarily linked and because they share physiological and anatomical similarities that are largely explored to understand the disease progression. The HIHISIV database was developed to support researchers to integrate and evaluate the large number of transcriptional data associated with the presence/absence of the pathogen (SIV or HIV) and the host response (NHP and human). The datasets are composed of microarray and RNA-Seq gene expression data that were selected, curated, analyzed, enriched, and stored in a relational database. Six query templates comprise the main data analysis functions and the resulting information can be downloaded. The HIHISIV database, available at https://hihisiv.github.io , provides accurate resources for browsing and visualizing results and for more robust analyses of pre-existing data in transcriptome repositories.
Collapse
Affiliation(s)
- Raquel L Costa
- DEXL Lab, National Laboratory for Scientific Computing, Petrópolis, Brazil.
| | - Luiz Gadelha
- German Human Genome-Phenome Archive (GHGA, W620), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Mirela D'arc
- Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Ribeiro-Alves
- Instituto Nacional de Infectologia Evandro Chagas, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - David L Robertson
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Jean-Marc Schwartz
- Division of Evolution, Infection and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Marcelo A Soares
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
- Programa de Oncovirologia, Divisão de Pesquisa Translacional, Instituto Nacional do Câncer, Rio de Janeiro, 20230-130, Brazil
| | - Fábio Porto
- DEXL Lab, National Laboratory for Scientific Computing, Petrópolis, Brazil
| |
Collapse
|
5
|
Huang Y, Dhummakupt A, Khetan P, Nilles T, Zhou W, Mudvari P, Szewczyk J, Chen YH, Boritz E, Ji H, Agwu A, Persaud D. Immune activation and exhaustion marker expression on T-cell subsets in ART-treated adolescents and young adults with perinatal HIV-1 infection as correlates of viral persistence. Front Immunol 2023; 14:1007626. [PMID: 37033916 PMCID: PMC10076634 DOI: 10.3389/fimmu.2023.1007626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
HIV-1 infection in memory CD4+ T cells forms a latent reservoir that is a barrier to cure. Identification of immune biomarkers that correlate with HIV-1 reservoir size may aid with evaluating efficacy of HIV-1 eradication strategies, towards ART-free remission and cure. In adults living with non-perinatal HIV-1, the immune exhaustion marker PD-1 on central memory CD4+ T cells (Tcm) correlates with measures of HIV-1 reservoir size. Immune correlates of HIV-1 are less defined in adolescents and young adults with perinatal HIV-1. With multi-parameter flow cytometry, we examined immune activation (CD69, CD25, HLA-DR), and exhaustion (PD-1, TIGIT, TIM-3 and LAG-3) markers on CD4+ T cell subsets (naïve (Tn), central memory (Tcm), and the combination (Ttem) of transitional (Ttm) and effector memory (Tem) cells, in 10 adolescents and young adults living with perinatal HIV-1 (median age 15.9 years; median duration of virologic suppression 7.0 years), in whom HIV-1 reservoir size was measured with the Intact Proviral HIV-1 DNA Assay (IPDA) and an enhanced Tat/Rev limiting dilution assay (TILDA). RNA-seq was also performed on the unstimulated CD4+ T cells. The median total HIV-1 DNA concentration in memory CD4+ T cells was 211.90 copies per million CD4+ T cells. In the 7 participants with subtype B HIV-1 infection, the median intact proviral DNA load was 7.96 copies per million CD4+ T cells. Levels of HLA-DR and TIGIT on the Ttem were correlated with total HIV-1 DNA (r=0.76, p=0.015) and (r=0.72, p=0.023), respectively, but not with intact proviral load or induced reservoir size. HIV-1 DNA load was also positively correlated with transcriptional clusters associated with HLA-DR expression by RNA-seq. In contrast, PD-1 expression on Tcm was inversely correlated with total HIV-1 DNA (r=-0.67, p=0.039). Reservoir size by IPDA and TILDA were correlated (r=0.81, p=0.036). Thus, in this cohort of youths with long-standing treated perinatal infection, HLA-DR and TIGIT on Ttem were the key correlates of HIV-1 infected cell frequencies by total HIV-1 DNA, and not PD-1. Total HIV-1 DNA was negatively correlated with PD-1 expressing Tcm. These differences in longstanding perinatal HIV-1 infection compared with adult infection requires further study in larger cohorts.
Collapse
Affiliation(s)
- Yuyang Huang
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Adit Dhummakupt
- Department of Pediatric Infectious Disease, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Priya Khetan
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Tricia Nilles
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Weiqiang Zhou
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Prakriti Mudvari
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, United States
| | - Joseph Szewczyk
- Department of Pediatric Infectious Disease, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ya Hui Chen
- Department of Pediatric Infectious Disease, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Eli Boritz
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, United States
| | - Hongkai Ji
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Allison Agwu
- Department of Pediatric Infectious Disease, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Deborah Persaud
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
- Department of Pediatric Infectious Disease, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Deborah Persaud,
| |
Collapse
|
6
|
Le Hingrat Q, Sereti I, Landay AL, Pandrea I, Apetrei C. The Hitchhiker Guide to CD4 + T-Cell Depletion in Lentiviral Infection. A Critical Review of the Dynamics of the CD4 + T Cells in SIV and HIV Infection. Front Immunol 2021; 12:695674. [PMID: 34367156 PMCID: PMC8336601 DOI: 10.3389/fimmu.2021.695674] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/09/2021] [Indexed: 01/02/2023] Open
Abstract
CD4+ T-cell depletion is pathognomonic for AIDS in both HIV and simian immunodeficiency virus (SIV) infections. It occurs early, is massive at mucosal sites, and is not entirely reverted by antiretroviral therapy (ART), particularly if initiated when T-cell functions are compromised. HIV/SIV infect and kill activated CCR5-expressing memory and effector CD4+ T-cells from the intestinal lamina propria. Acute CD4+ T-cell depletion is substantial in progressive, nonprogressive and controlled infections. Clinical outcome is predicted by the mucosal CD4+ T-cell recovery during chronic infection, with no recovery occurring in rapid progressors, and partial, transient recovery, the degree of which depends on the virus control, in normal and long-term progressors. The nonprogressive infection of African nonhuman primate SIV hosts is characterized by partial mucosal CD4+ T-cell restoration, despite high viral replication. Complete, albeit very slow, recovery of mucosal CD4+ T-cells occurs in controllers. Early ART does not prevent acute mucosal CD4+ T-cell depletion, yet it greatly improves their restoration, sometimes to preinfection levels. Comparative studies of the different models of SIV infection support a critical role of immune activation/inflammation (IA/INFL), in addition to viral replication, in CD4+ T-cell depletion, with immune restoration occurring only when these parameters are kept at bay. CD4+ T-cell depletion is persistent, and the recovery is very slow, even when both the virus and IA/INFL are completely controlled. Nevertheless, partial mucosal CD4+ T-cell recovery is sufficient for a healthy life in natural hosts. Cell death and loss of CD4+ T-cell subsets critical for gut health contribute to mucosal inflammation and enteropathy, which weaken the mucosal barrier, leading to microbial translocation, a major driver of IA/INFL. In turn, IA/INFL trigger CD4+ T-cells to become either viral targets or apoptotic, fueling their loss. CD4+ T-cell depletion also drives opportunistic infections, cancers, and comorbidities. It is thus critical to preserve CD4+ T cells (through early ART) during HIV/SIV infection. Even in early-treated subjects, residual IA/INFL can persist, preventing/delaying CD4+ T-cell restoration. New therapeutic strategies limiting mucosal pathology, microbial translocation and IA/INFL, to improve CD4+ T-cell recovery and the overall HIV prognosis are needed, and SIV models are extensively used to this goal.
Collapse
Affiliation(s)
- Quentin Le Hingrat
- Division of Infectious Diseases, DOM, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Irini Sereti
- HIV Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alan L Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Ivona Pandrea
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Infectious Diseases and Immunology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Cristian Apetrei
- Division of Infectious Diseases, DOM, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Infectious Diseases and Immunology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
7
|
Raehtz KD, Barrenäs F, Xu C, Busman-Sahay K, Valentine A, Law L, Ma D, Policicchio BB, Wijewardana V, Brocca-Cofano E, Trichel A, Gale M, Keele BF, Estes JD, Apetrei C, Pandrea I. African green monkeys avoid SIV disease progression by preventing intestinal dysfunction and maintaining mucosal barrier integrity. PLoS Pathog 2020; 16:e1008333. [PMID: 32119719 PMCID: PMC7077871 DOI: 10.1371/journal.ppat.1008333] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 03/17/2020] [Accepted: 01/18/2020] [Indexed: 12/12/2022] Open
Abstract
Unlike HIV infection, SIV infection is generally nonpathogenic in natural hosts, such as African green monkeys (AGMs), despite life-long high viral replication. Lack of disease progression was reportedly based on the ability of SIV-infected AGMs to prevent gut dysfunction, avoiding microbial translocation and the associated systemic immune activation and chronic inflammation. Yet, the maintenance of gut integrity has never been documented, and the mechanism(s) by which gut integrity is preserved are unknown. We sought to investigate the early events of SIV infection in AGMs, specifically examining the impact of SIVsab infection on the gut mucosa. Twenty-nine adult male AGMs were intrarectally infected with SIVsab92018 and serially sacrificed at well-defined stages of SIV infection, preramp-up (1-3 days post-infection (dpi)), ramp-up (4-6 dpi), peak viremia (9-12 dpi), and early chronic SIV infection (46-55 dpi), to assess the levels of immune activation, apoptosis, epithelial damage and microbial translocation in the GI tract and peripheral lymph nodes. Tissue viral loads, plasma cytokines and plasma markers of gut dysfunction were also measured throughout the course of early infection. While a strong, but transient, interferon-based inflammatory response was observed, the levels of plasma markers linked to enteropathy did not increase. Accordingly, no significant increases in apoptosis of either mucosal enterocytes or lymphocytes, and no damage to the mucosal epithelium were documented during early SIVsab infection of AGMs. These findings were supported by RNAseq of the gut tissue, which found no significant alterations in gene expression that would indicate microbial translocation. Thus, for the first time, we confirmed that gut epithelial integrity is preserved, with no evidence of microbial translocation, in AGMs throughout early SIVsab infection. This might protect AGMs from developing intestinal dysfunction and the subsequent chronic inflammation that drives both HIV disease progression and HIV-associated comorbidities.
Collapse
Affiliation(s)
- Kevin D. Raehtz
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Fredrik Barrenäs
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Cuiling Xu
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Kathleen Busman-Sahay
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Audrey Valentine
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Lynn Law
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
- Center for Innate Immunity and Immune Diseases, University of Washington, Washington, United States of America
| | - Dongzhu Ma
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Benjamin B. Policicchio
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Viskam Wijewardana
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Egidio Brocca-Cofano
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Anita Trichel
- Division of Laboratory Animal Resources, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
- Center for Innate Immunity and Immune Diseases, University of Washington, Washington, United States of America
- Washington National Primate Research Center, University of Washington, Seattle, Washington, United States of America
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory of Cancer Research, Frederick, Maryland, United States of America
| | - Jacob D. Estes
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Cristian Apetrei
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Ivona Pandrea
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
8
|
Chaudhary O, Narayan V, Lelis F, Linz B, Watkins M, Veazey R, Aldovini A. Inhibition of p38 MAPK in combination with ART reduces SIV-induced immune activation and provides additional protection from immune system deterioration. PLoS Pathog 2018; 14:e1007268. [PMID: 30161247 PMCID: PMC6135519 DOI: 10.1371/journal.ppat.1007268] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 09/12/2018] [Accepted: 08/08/2018] [Indexed: 12/12/2022] Open
Abstract
Differences in immune activation were identified as the most significant difference between AIDS-susceptible and resistant species. p38 MAPK, activated in HIV infection, is key to induction of interferon-stimulated genes and cytokine-mediated inflammation and is associated with some of the pathology produced by HIV or SIV infection in AIDS-susceptible primates. As small molecule p38 MAPK inhibitors are being tested in human trials for inflammatory diseases, we evaluated the effects of treating SIV-infected macaques with the p38 MAPK inhibitor PH-797804 in conjunction with ART. PH-797804 had no side effects, did not impact negatively the antiviral immune response and, used alone, had no significant effect on levels of immune activation and did not reduced the viremia. When administered with ART, it significantly reduced numerous immune activation markers compared to ART alone. CD38+/HLA-DR+ and Ki-67+ T-cell percentages in blood, lymph node and rectal CD4+ and CD8+ T cells, PD-1 expression in CD8+ T cells and plasma levels of IFNα, IFNγ, TNFα, IL-6, IP-10, sCD163 and C-reactive protein were all significantly reduced. Significant preservation of CD4+, CD4+ central memory, CD4+/IL-22+ and CD4+/IL-17+ T-cell percentages and improvement of Th17/Treg ratio in blood and rectal mucosa were also observed. Importantly, the addition of PH-797804 to ART initiated during chronic SIV infection reduced immune activation and restored immune system parameters to the levels observed when ART was initiated on week 1 after infection. After ART interruption, viremia rebounded in a similar fashion in all groups, regardless of when ART was initiated. We concluded that the inhibitor PH-797804 significantly reduced, even if did not normalized, the immune activation parameters evaluated during ART treatment, improved preservation of critical populations of the immune system targeted by SIV, and increased the efficacy of ART treatment initiated in chronic infection to levels similar to those observed when initiated in acute infection but did not affect positively or negatively viral reservoirs. The hallmark of Human Immunodeficiency Virus and Simian Immunodeficiency Virus infection in disease-susceptible species is the progressive decline of the CD4+ T cell population and heightened immune activation, which by itself can contribute to CD4+ T-cell death. The cellular pathway regulated by p38 MAPK, which is activated in HIV and SIV infection, can contribute significantly to immune activation. We tested in SIV-infected macaques a p38 MAPK inhibitor in combination with anti-retroviral therapy. This drug is already being evaluated in humans for treatment of immune activation associated with other diseases. We found that, when combined with antiretroviral therapy, the inhibitor PH-797804 significantly reduced a few parameters of SIV-induced immune activation and improved preservation of critical populations of the immune system targeted by SIV, but did not modulate viral reservoirs. Importantly, the addition of the inhibitor to anti-retroviral therapy during the chronic phase of the infection, which is the time when most HIV-infected individuals initiate treatment, permitted a more significant preservation of the immune system compared to antiretroviral therapy alone that was similar to that observed when anti-retroviral therapy was initiated in the acute phase of the infection, which rarely occurs in HIV infection.
Collapse
Affiliation(s)
- Omkar Chaudhary
- Boston Children’s Hospital, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston MA, United States of America
| | - Vivek Narayan
- Boston Children’s Hospital, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston MA, United States of America
| | - Felipe Lelis
- Boston Children’s Hospital, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston MA, United States of America
| | - Brandon Linz
- Boston Children’s Hospital, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston MA, United States of America
| | - Meagan Watkins
- Tulane National Primate Research Center, Division of Comparative Pathology, Covington LA, United States of America
| | - Ronald Veazey
- Tulane National Primate Research Center, Division of Comparative Pathology, Covington LA, United States of America
| | - Anna Aldovini
- Boston Children’s Hospital, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston MA, United States of America
- * E-mail:
| |
Collapse
|
9
|
CCR5 Revisited: How Mechanisms of HIV Entry Govern AIDS Pathogenesis. J Mol Biol 2018; 430:2557-2589. [PMID: 29932942 DOI: 10.1016/j.jmb.2018.06.027] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/10/2018] [Accepted: 06/13/2018] [Indexed: 01/01/2023]
Abstract
The chemokine receptor CCR5 has been the focus of intensive studies since its role as a coreceptor for HIV entry was discovered in 1996. These studies lead to the development of small molecular drugs targeting CCR5, with maraviroc becoming in 2007 the first clinically approved chemokine receptor inhibitor. More recently, the apparent HIV cure in a patient transplanted with hematopoietic stem cells devoid of functional CCR5 rekindled the interest for inactivating CCR5 through gene therapy and pharmacological approaches. Fundamental research on CCR5 has also been boosted by key advances in the field of G-protein coupled receptor research, with the realization that CCR5 adopts a variety of conformations, and that only a subset of these conformations may be targeted by chemokine ligands. In addition, recent genetic and pathogenesis studies have emphasized the central role of CCR5 expression levels in determining the risk of HIV and SIV acquisition and disease progression. In this article, we propose to review the key properties of CCR5 that account for its central role in HIV pathogenesis, with a focus on mechanisms that regulate CCR5 expression, conformation, and interaction with HIV envelope glycoproteins.
Collapse
|
10
|
Huot N, Bosinger SE, Paiardini M, Reeves RK, Müller-Trutwin M. Lymph Node Cellular and Viral Dynamics in Natural Hosts and Impact for HIV Cure Strategies. Front Immunol 2018; 9:780. [PMID: 29725327 PMCID: PMC5916971 DOI: 10.3389/fimmu.2018.00780] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 03/28/2018] [Indexed: 01/03/2023] Open
Abstract
Combined antiretroviral therapies (cARTs) efficiently control HIV replication leading to undetectable viremia and drastic increases in lifespan of people living with HIV. However, cART does not cure HIV infection as virus persists in cellular and anatomical reservoirs, from which the virus generally rebounds soon after cART cessation. One major anatomical reservoir are lymph node (LN) follicles, where HIV persists through replication in follicular helper T cells and is also trapped by follicular dendritic cells. Natural hosts of SIV, such as African green monkeys and sooty mangabeys, generally do not progress to disease although displaying persistently high viremia. Strikingly, these hosts mount a strong control of viral replication in LN follicles shortly after peak viremia that lasts throughout infection. Herein, we discuss the potential interplay between viral control in LNs and the resolution of inflammation, which is characteristic for natural hosts. We furthermore detail the differences that exist between non-pathogenic SIV infection in natural hosts and pathogenic HIV/SIV infection in humans and macaques regarding virus target cells and replication dynamics in LNs. Several mechanisms have been proposed to be implicated in the strong control of viral replication in natural host's LNs, such as NK cell-mediated control, that will be reviewed here, together with lessons and limitations of in vivo cell depletion studies that have been performed in natural hosts. Finally, we discuss the impact that these insights on viral dynamics and host responses in LNs of natural hosts have for the development of strategies toward HIV cure.
Collapse
Affiliation(s)
- Nicolas Huot
- HIV Inflammation and Persistence Unit, Institut Pasteur, Paris, France.,Vaccine Research Institute, Créteil, France
| | - Steven E Bosinger
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes Nonhuman Primate Genomics Core, Yerkes National Primate Research Center, Atlanta, GA, United States
| | - Mirko Paiardini
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA, United States
| | - R Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, United States.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Michaela Müller-Trutwin
- HIV Inflammation and Persistence Unit, Institut Pasteur, Paris, France.,Vaccine Research Institute, Créteil, France
| |
Collapse
|
11
|
Pathogenic Correlates of Simian Immunodeficiency Virus-Associated B Cell Dysfunction. J Virol 2017; 91:JVI.01051-17. [PMID: 28931679 DOI: 10.1128/jvi.01051-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/12/2017] [Indexed: 01/08/2023] Open
Abstract
We compared and contrasted pathogenic (in pig-tailed macaques [PTMs]) and nonpathogenic (in African green monkeys [AGMs]) SIVsab infections to assess the significance of the B cell dysfunction observed in simian (SIV) and human immunodeficiency virus (HIV) infections. We report that the loss of B cells is specifically associated with the pathogenic SIV infection, while in the natural hosts, in which SIV is nonpathogenic, B cells rapidly increase in both lymph nodes (LNs) and intestine. SIV-associated B cell dysfunction associated with the pathogenic SIV infection is characterized by loss of naive B cells, loss of resting memory B cells due to their redistribution to the gut, increases of the activated B cells and circulating tissue-like memory B cells, and expansion of the B regulatory cells (Bregs). While circulating B cells are virtually restored to preinfection levels during the chronic pathogenic SIV infection, restoration is mainly due to an expansion of the "exhausted," virus-specific B cells, i.e., activated memory cells and tissue-like memory B cells. Despite of the B cell dysfunction, SIV-specific antibody (Ab) production was higher in the PTMs than in AGMs, with the caveat that rapid disease progression in PTMs was strongly associated with lack of anti-SIV Ab. Neutralization titers and the avidity and maturation of immune responses did not differ between pathogenic and nonpathogenic infections, with the exception of the conformational epitope recognition, which evolved from low to high conformations in the natural host. The patterns of humoral immune responses in the natural host are therefore more similar to those observed in HIV-infected subjects, suggesting that natural hosts may be more appropriate for modeling the immunization strategies aimed at preventing HIV disease progression. The numerous differences between the pathogenic and nonpathogenic infections with regard to dynamics of the memory B cell subsets point to their role in the pathogenesis of HIV/SIV infections and suggest that monitoring B cells may be a reliable approach for assessing disease progression.IMPORTANCE We report here that the HIV/SIV-associated B cell dysfunction (defined by loss of total and memory B cells, increased B regulatory cell [Breg] counts, and B cell activation and apoptosis) is specifically associated with pathogenic SIV infection and absent during the course of nonpathogenic SIV infection in natural nonhuman primate hosts. Alterations of the B cell population are not correlated with production of neutralizing antibodies, the levels of which are similar in the two species. Rapid progressive infections are associated with a severe impairment in SIV-specific antibody production. While we did not find major differences in avidity and maturation between the pathogenic and nonpathogenic SIV infections, we identified a major difference in conformational epitope recognition, with the nonpathogenic infection being characterized by an evolution from low to high conformations. B cell dysfunction should be considered in designing immunization strategies aimed at preventing HIV disease progression.
Collapse
|
12
|
Immune activation in HIV infection: what can the natural hosts of simian immunodeficiency virus teach us? Curr Opin HIV AIDS 2016; 11:201-8. [PMID: 26845673 DOI: 10.1097/coh.0000000000000238] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW The review summarizes studies in natural hosts, with a particular focus on the control of immune activation and new insights into viral reservoirs. We discuss why these findings are relevant for HIV research today. RECENT FINDINGS AIDS resistance in natural hosts is characterized by a rapid control of inflammatory processes in response to simian immunodeficiency virus infection despite persistent viremia. Although CD4 T cells are dramatically depleted in the intestine in primary infection, interleukin 17-producing T helper cells (Th17) are preserved and natural hosts lack microbial translocation. Thus, viral replication in the gut is not sufficient to explain mucosal damage, but additional factors are necessary. Natural hosts also display a lower infection rate of stem-cell memory, central memory and follicular helper T cells. The follicles are characterized by a lack of viral trapping and the viral replication in secondary lymphoid organs is rapidly controlled. Hence, the healthy status of natural hosts is associated with preserved lymphoid environments. SUMMARY Understanding the underlying mechanisms of preservation of Th17 and of the low contribution of stem-cell memory, central memory and follicular helper T cells to viral reservoirs could benefit the search for preventive and curative approaches of HIV. Altogether, the complementarity of the model helps to identify strategies aiming at restoring full capacity of the immune system and decreasing the size of the viral reservoirs.
Collapse
|
13
|
Visseaux B, Damond F, Matheron S, Descamps D, Charpentier C. Hiv-2 molecular epidemiology. INFECTION GENETICS AND EVOLUTION 2016; 46:233-240. [PMID: 27530215 DOI: 10.1016/j.meegid.2016.08.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 12/13/2022]
Abstract
The Simian Immunodeficiency Virus of sooty mangabeys (SIVsmm) has been revealed to be at the origin of Human Immunodeficiency Virus type 2 (HIV-2) in humans, firstly detected from two Portuguese patients in 1986. HIV-2 is mainly restricted to West Africa where it infects up to 1 to 2 million people. HIV-2 is also present in Europe, mainly Portugal and France, India and United States of America. Two major HIV-2 groups, groups A and B, were generated by two independent transmission events involving infected sooty mangabeys from the Taï forest in Ivory Coast. Seven other HIV-2 groups have been described, but each has only been identified in one patient. To date, no subtypes have been formally described but some preliminary data suggest that HIV-2 group A may be divided in two distinct subtypes with distinct geographical origins. To date only two recombinant forms have been described: one circulating recombinant form (CRF01_AB) and one unique recombinant form. In this review, we focused mainly on molecular data available and their insights about HIV-2 origins, diversity, drug resistance and global epidemiology.
Collapse
Affiliation(s)
- Benoit Visseaux
- INSERM, IAME, UMR 1137, F-75018 Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France; AP-HP, Hôpital Bichat, Laboratoire de Virologie, F-75018 Paris, France.
| | - Florence Damond
- INSERM, IAME, UMR 1137, F-75018 Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France; AP-HP, Hôpital Bichat, Laboratoire de Virologie, F-75018 Paris, France
| | - Sophie Matheron
- INSERM, IAME, UMR 1137, F-75018 Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France; AP-HP, Hôpital Bichat, Service de Maladies Infectieuses et Tropicales, F-75018 Paris, France
| | - Diane Descamps
- INSERM, IAME, UMR 1137, F-75018 Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France; AP-HP, Hôpital Bichat, Laboratoire de Virologie, F-75018 Paris, France
| | - Charlotte Charpentier
- INSERM, IAME, UMR 1137, F-75018 Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France; AP-HP, Hôpital Bichat, Laboratoire de Virologie, F-75018 Paris, France
| |
Collapse
|
14
|
Garcia-Tellez T, Huot N, Ploquin MJ, Rascle P, Jacquelin B, Müller-Trutwin M. Non-human primates in HIV research: Achievements, limits and alternatives. INFECTION GENETICS AND EVOLUTION 2016; 46:324-332. [PMID: 27469027 DOI: 10.1016/j.meegid.2016.07.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/07/2016] [Accepted: 07/12/2016] [Indexed: 12/20/2022]
Abstract
An ideal model for HIV-1 research is still unavailable. However, infection of non-human primates (NHP), such as macaques, with Simian Immunodeficiency Virus (SIV) recapitulates most virological, immunological and clinical hallmarks of HIV infection in humans. It has become the most suitable model to study the mechanisms of transmission and physiopathology of HIV/AIDS. On the other hand, natural hosts of SIV, such as African green monkeys and sooty mangabeys that when infected do not progress to AIDS, represent an excellent model to elucidate the mechanisms involved in the capacity of controlling inflammation and disease progression. The use of NHP-SIV models has indeed enriched our knowledge in the fields of: i) viral transmission and viral reservoirs, ii) early immune responses, iii) host cell-virus interactions in tissues, iv) AIDS pathogenesis, v) virulence factors, vi) prevention and vii) drug development. The possibility to control many variables during experimental SIV infection, together with the resemblance between SIV and HIV infections, make the NHP model the most appropriate, so far, for HIV/AIDS research. Nonetheless, some limitations in using these models have to be considered. Alternative models for HIV/AIDS research, such as humanized mice and recombinant forms of HIV-SIV viruses (SHIV) for NHP infection, have been developed. The improvement of SHIV viruses that mimic even better the natural history of HIV infection and of humanized mice that develop a greater variety of human immune cell lineages, is ongoing. None of these models is perfect, but they allow contributing to the progress in managing or preventing HIV infection.
Collapse
Affiliation(s)
- Thalía Garcia-Tellez
- Institut Pasteur, Unité HIV, Inflammation and Persistence. 25-28 Rue du Doctor Roux,75015 Paris, France.
| | - Nicolas Huot
- Institut Pasteur, Unité HIV, Inflammation and Persistence. 25-28 Rue du Doctor Roux,75015 Paris, France; Vaccine Research Institute, Créteil, France.
| | - Mickaël J Ploquin
- Institut Pasteur, Unité HIV, Inflammation and Persistence. 25-28 Rue du Doctor Roux,75015 Paris, France.
| | - Philippe Rascle
- Institut Pasteur, Unité HIV, Inflammation and Persistence. 25-28 Rue du Doctor Roux,75015 Paris, France.
| | - Beatrice Jacquelin
- Institut Pasteur, Unité HIV, Inflammation and Persistence. 25-28 Rue du Doctor Roux,75015 Paris, France.
| | - Michaela Müller-Trutwin
- Institut Pasteur, Unité HIV, Inflammation and Persistence. 25-28 Rue du Doctor Roux,75015 Paris, France; Vaccine Research Institute, Créteil, France.
| |
Collapse
|
15
|
The well-tempered SIV infection: Pathogenesis of SIV infection in natural hosts in the wild, with emphasis on virus transmission and early events post-infection that may contribute to protection from disease progression. INFECTION GENETICS AND EVOLUTION 2016; 46:308-323. [PMID: 27394696 DOI: 10.1016/j.meegid.2016.07.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/04/2016] [Accepted: 07/05/2016] [Indexed: 12/25/2022]
Abstract
African NHPs are infected by over 40 different simian immunodeficiency viruses. These viruses have coevolved with their hosts for long periods of time and, unlike HIV in humans, infection does not generally lead to disease progression. Chronic viral replication is maintained for the natural lifespan of the host, without loss of overall immune function. Lack of disease progression is not correlated with transmission, as SIV infection is highly prevalent in many African NHP species in the wild. The exact mechanisms by which these natural hosts of SIV avoid disease progression are still unclear, but a number of factors might play a role, including: (i) avoidance of microbial translocation from the gut lumen by preventing or repairing damage to the gut epithelium; (ii) control of immune activation and apoptosis following infection; (iii) establishment of an anti-inflammatory response that resolves chronic inflammation; (iv) maintenance of homeostasis of various immune cell populations, including NK cells, monocytes/macrophages, dendritic cells, Tregs, Th17 T-cells, and γδ T-cells; (v) restriction of CCR5 availability at mucosal sites; (vi) preservation of T-cell function associated with down-regulation of CD4 receptor. Some of these mechanisms might also be involved in protection of natural hosts from mother-to-infant SIV transmission during breastfeeding. The difficulty of performing invasive studies in the wild has prohibited investigation of the exact events surrounding transmission in natural hosts. Increased understanding of the mechanisms of SIV transmission in natural hosts, and of the early events post-transmission which may contribute to avoidance of disease progression, along with better comprehension of the factors involved in protection from SIV breastfeeding transmission in the natural hosts, could prove invaluable for the development of new prevention strategies for HIV.
Collapse
|
16
|
Abstract
Although the replicative life cycle of HIV within CD4 T cells is understood in molecular detail, less is known about how this human retrovirus promotes the loss of CD4 T lymphocytes. It is this cell death process that drives clinical progression to acquired immune deficiency syndrome (AIDS). Recent studies have highlighted how abortive infection of resting and thus nonpermissive CD4 T cells in lymphoid tissues triggers a lethal innate immune response against the incomplete DNA products generated by inefficient viral reverse transcription in these cells. Sensing of these DNA fragments results in pyroptosis, a highly inflammatory form of programmed cell death, that potentially further perpetuates chronic inflammation and immune activation. As discussed here, these studies cast CD4 T cell death during HIV infection in a different light. Further, they identify drug targets that may be exploited to both block CD4 T cell demise and the chronic inflammatory response generated during pyroptosis.
Collapse
Affiliation(s)
- Gilad Doitsh
- Gladstone Institute of Virology and Immunology, 1650 Owens Street, San Francisco, CA 94158, USA.
| | - Warner C Greene
- Gladstone Institute of Virology and Immunology, 1650 Owens Street, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
17
|
Simian Immunodeficiency Virus SIVagm Efficiently Utilizes Non-CCR5 Entry Pathways in African Green Monkey Lymphocytes: Potential Role for GPR15 and CXCR6 as Viral Coreceptors. J Virol 2015; 90:2316-31. [PMID: 26656714 DOI: 10.1128/jvi.02529-15] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/04/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED African green monkeys (AGM) are natural hosts of simian immunodeficiency virus (SIV), and infection in these animals is generally nonpathogenic, whereas infection of nonnatural hosts, such as rhesus macaques (RM), is commonly pathogenic. CCR5 has been described as the primary entry coreceptor for SIV in vivo, while human-derived CXCR6 and GPR15 also appear to be used in vitro. However, sooty mangabeys that are genetically deficient in CCR5 due to an out-of-frame deletion are infectible with SIVsmm, indicating that SIVsmm can use alternative coreceptors in vivo. In this study, we examined the CCR5 dependence of SIV strains derived from vervet AGM (SIVagmVer) and the ability of AGM-derived GPR15 and CXCR6 to serve as potential entry coreceptors. We found that SIVagmVer replicated efficiently in AGM and RM peripheral blood mononuclear cells (PBMC) in the presence of the CCR5 antagonist maraviroc, despite the fact that maraviroc was capable of blocking the CCR5-tropic strains SIVmac239, SIVsmE543-3, and simian-human immunodeficiency virus SHIV-AD8 in RM PBMC. We also found that AGM CXCR6 and AGM GPR15, to a lesser extent, supported entry of pseudotype viruses bearing SIVagm envelopes, including SIVagm transmitted/founder envelopes. Lastly, we found that CCR5, GPR15, and CXCR6 mRNAs were detected in AGM and RM memory CD4(+) T cells. These results suggest that GPR15 and CXCR6 are expressed on AGM CD4(+) T cells and are potential alternative coreceptors for SIVagm use in vivo. These data suggest that the use of non-CCR5 entry pathways may be a common feature of SIV replication in natural host species, with the potential to contribute to nonpathogenicity in these animals. IMPORTANCE African green monkeys (AGM) are natural hosts of SIV, and infection in these animals generally does not cause AIDS, whereas SIV-infected rhesus macaques (RM) typically develop AIDS. Although it has been reported that SIV generally uses CD4 and CCR5 to enter target cells in vivo, other molecules, such as GPR15 and CXCR6, also function as SIV coreceptors in vitro. In this study, we investigated whether SIV from vervet AGM can use non-CCR5 entry pathways, as has been observed in sooty mangabeys. We found that SIVagmVer efficiently replicated in AGM and RM peripheral blood mononuclear cells in the presence of the CCR5 antagonist maraviroc, suggesting that non-CCR5 entry pathways can support SIVagm entry. We found that AGM-derived GPR15 and CXCR6 support SIVagmVer entry in vitro and may serve as entry coreceptors for SIVagm in vivo, since their mRNAs were detected in AGM memory CD4(+) T cells, the preferred target cells of SIV.
Collapse
|
18
|
Pulliam L. Cognitive consequences of a sustained monocyte type 1 IFN response in HIV-1 infection. Curr HIV Res 2015; 12:77-84. [PMID: 24862334 DOI: 10.2174/1570162x12666140526113544] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 12/16/2022]
Abstract
With successful antiretroviral therapy, HIV-1-infected subjects can achieve undetectable peripheral viral loads and immune homeostasis. However, in a subset of individuals on therapy, peripheral monocytes have a gene expression profile characteristic of a type 1 interferon α (IFN) response. This type 1 IFN response correlates with a number of pathogenic conditions including neural cell injury and in combination with HCV infection, cognitive impairment. Lessons from the non-human primate models of pathogenic and nonpathogenic SIV suggest that returning the initial IFN spike in acute SIV infection to normal allows the immune system to control infection and return to homeostasis. An IFN "alarm" signature, defined as monocyte activation with overexpression of the type1 IFN genes IFI27 and CD169, would be useful for identifying a subset of subjects with HIV-1 infection that could progress to a number of pathologies associated with immune activation including cognitive dysfunction. This strategy is being actively pursued for autoimmune diseases that are characterized by an IFN signature. Therapies to block the IFN signature are under investigation as a means to reset the immune system and in a subset of HIV-1-infected subjects may be an adjuvant to standard antiviral therapy to return cognitive function.
Collapse
Affiliation(s)
- Lynn Pulliam
- Veterans Affairs Medical Center (113A), 4150 Clement St., San Francisco, CA 94121, USA.
| |
Collapse
|
19
|
Jacquelin B, Petitjean G, Kunkel D, Liovat AS, Jochems SP, Rogers KA, Ploquin MJ, Madec Y, Barré-Sinoussi F, Dereuddre-Bosquet N, Lebon P, Le Grand R, Villinger F, Müller-Trutwin M. Innate immune responses and rapid control of inflammation in African green monkeys treated or not with interferon-alpha during primary SIVagm infection. PLoS Pathog 2014; 10:e1004241. [PMID: 24991927 PMCID: PMC4081777 DOI: 10.1371/journal.ppat.1004241] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 05/26/2014] [Indexed: 12/20/2022] Open
Abstract
Chronic immune activation (IA) is considered as the driving force of CD4+ T cell depletion and AIDS. Fundamental clues in the mechanisms that regulate IA could lie in natural hosts of SIV, such as African green monkeys (AGMs). Here we investigated the role of innate immune cells and IFN-α in the control of IA in AGMs. AGMs displayed significant NK cell activation upon SIVagm infection, which was correlated with the levels of IFN-α. Moreover, we detected cytotoxic NK cells in lymph nodes during the early acute phase of SIVagm infection. Both plasmacytoid and myeloid dendritic cell (pDC and mDC) homing receptors were increased, but the maturation of mDCs, in particular of CD16+ mDCs, was more important than that of pDCs. Monitoring of 15 cytokines showed that those, which are known to be increased early in HIV-1/SIVmac pathogenic infections, such as IL-15, IFN-α, MCP-1 and CXCL10/IP-10, were significantly increased in AGMs as well. In contrast, cytokines generally induced in the later stage of acute pathogenic infection, such as IL-6, IL-18 and TNF-α, were less or not increased, suggesting an early control of IA. We then treated AGMs daily with high doses of IFN-α from day 9 to 24 post-infection. No impact was observed on the activation or maturation profiles of mDCs, pDCs and NK cells. There was also no major difference in T cell activation or interferon-stimulated gene (ISG) expression profiles and no sign of disease progression. Thus, even after administration of high levels of IFN-α during acute infection, AGMs were still able to control IA, showing that IA control is independent of IFN-α levels. This suggests that the sustained ISG expression and IA in HIV/SIVmac infections involves non-IFN-α products. Chronic inflammation is considered as directly involved in AIDS pathogenesis. The role of IFN-α as a driving force of chronic inflammation is under debate. Natural hosts of SIV, such as African green monkeys (AGMs), avoid chronic inflammation. We show for the first time that NK cells are strongly activated during acute SIVagm infection. This further demonstrates that AGMs mount a strong early innate immune response. Myeloid and plasmacytoid dendritic cells (mDCs and pDCs) homed to lymph nodes; however mDCs showed a stronger maturation profile than pDCs. Monitoring of cytokine profiles in plasma suggests that the control of inflammation in AGMs is starting earlier than previously considered, weeks before the end of the acute infection. We tested whether the capacity to control inflammation depends on the levels of IFN-α produced. When treated with high doses of IFN-α during acute SIVagm infection, AGMs did not show increase of immune activation or signs of disease progression. Our study provides evidence that the control of inflammation in SIVagm infection is not the consequence of weaker IFN-α levels. These data indicate that the sustained interferon-stimulated gene induction and chronic inflammation in HIV/SIVmac infections is driven by factors other than IFN-α.
Collapse
Affiliation(s)
- Béatrice Jacquelin
- Institut Pasteur, Regulation of Retroviral Infection Unit, Paris, France
| | - Gaël Petitjean
- Institut Pasteur, Regulation of Retroviral Infection Unit, Paris, France
| | - Désirée Kunkel
- Institut Pasteur, Regulation of Retroviral Infection Unit, Paris, France
| | - Anne-Sophie Liovat
- Institut Pasteur, Regulation of Retroviral Infection Unit, Paris, France
| | - Simon P. Jochems
- Institut Pasteur, Regulation of Retroviral Infection Unit, Paris, France
- Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Kenneth A. Rogers
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Mickaël J. Ploquin
- Institut Pasteur, Regulation of Retroviral Infection Unit, Paris, France
| | - Yoann Madec
- Institut Pasteur, Emerging Diseases Epidemiology Unit, Paris, France
| | | | | | - Pierre Lebon
- Saint-Vincent de Paul Hospital & Paris Descartes University, Paris, France
| | - Roger Le Grand
- CEA, Division of Immuno-Virology, DSV, iMETI, Fontenay-aux-Roses, France
| | - François Villinger
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | | |
Collapse
|
20
|
Factors associated with siman immunodeficiency virus transmission in a natural African nonhuman primate host in the wild. J Virol 2014; 88:5687-705. [PMID: 24623416 DOI: 10.1128/jvi.03606-13] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED African green monkeys (AGMs) are naturally infected with simian immunodeficiency virus (SIV) at high prevalence levels and do not progress to AIDS. Sexual transmission is the main transmission route in AGM, while mother-to-infant transmission (MTIT) is negligible. We investigated SIV transmission in wild AGMs to assess whether or not high SIV prevalence is due to differences in mucosal permissivity to SIV (i.e., whether the genetic bottleneck of viral transmission reported in humans and macaques is also observed in AGMs in the wild). We tested 121 sabaeus AGMs (Chlorocebus sabaeus) from the Gambia and found that 53 were SIV infected (44%). By combining serology and viral load quantitation, we identified 4 acutely infected AGMs, in which we assessed the diversity of the quasispecies by single-genome amplification (SGA) and documented that a single virus variant established the infections. We thus show that natural SIV transmission in the wild is associated with a genetic bottleneck similar to that described for mucosal human immunodeficiency virus (HIV) transmission in humans. Flow cytometry assessment of the immune cell populations did not identify major differences between infected and uninfected AGM. The expression of the SIV coreceptor CCR5 on CD4+ T cells dramatically increased in adults, being higher in infected than in uninfected infant and juvenile AGMs. Thus, the limited SIV MTIT in natural hosts appears to be due to low target cell availability in newborns and infants, which supports HIV MTIT prevention strategies aimed at limiting the target cells at mucosal sites. Combined, (i) the extremely high prevalence in sexually active AGMs, (ii) the very efficient SIV transmission in the wild, and (iii) the existence of a fraction of multiparous females that remain uninfected in spite of massive exposure to SIV identify wild AGMs as an acceptable model of exposed, uninfected individuals. IMPORTANCE We report an extensive analysis of the natural history of SIVagm infection in its sabaeus monkey host, the African green monkey species endemic to West Africa. Virtually no study has investigated the natural history of SIV infection in the wild. The novelty of our approach is that we report for the first time that SIV infection has no discernible impact on the major immune cell populations in natural hosts, thus confirming the nonpathogenic nature of SIV infection in the wild. We also focused on the correlates of SIV transmission, and we report, also for the first time, that SIV transmission in the wild is characterized by a major genetic bottleneck, similar to that described for HIV-1 transmission in humans. Finally, we report here that the restriction of target cell availability is a major correlate of the lack of SIV transmission to the offspring in natural hosts of SIVs.
Collapse
|
21
|
Miedema F, Hazenberg MD, Tesselaar K, van Baarle D, de Boer RJ, Borghans JAM. Immune activation and collateral damage in AIDS pathogenesis. Front Immunol 2013; 4:298. [PMID: 24133492 PMCID: PMC3783946 DOI: 10.3389/fimmu.2013.00298] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 09/09/2013] [Indexed: 12/17/2022] Open
Abstract
In the past decade, evidence has accumulated that human immunodeficiency virus (HIV)-induced chronic immune activation drives progression to AIDS. Studies among different monkey species have shown that the difference between pathological and non-pathological infection is determined by the response of the immune system to the virus, rather than its cytopathicity. Here we review the current understanding of the various mechanisms driving chronic immune activation in HIV infection, the cell types involved, its effects on HIV-specific immunity, and how persistent inflammation may cause AIDS and the wide spectrum of non-AIDS related pathology. We argue that therapeutic relief of inflammation may be beneficial to delay HIV-disease progression and to reduce non-AIDS related pathological side effects of HIV-induced chronic immune stimulation.
Collapse
Affiliation(s)
- Frank Miedema
- Department of Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mette D. Hazenberg
- Department of Internal Medicine and Hematology, Academic Medical Center, Amsterdam, Netherlands
| | - Kiki Tesselaar
- Department of Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Debbie van Baarle
- Department of Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Rob J. de Boer
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, Netherlands
| | - José A. M. Borghans
- Department of Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
22
|
Klatt NR, Silvestri G, Hirsch V. Nonpathogenic simian immunodeficiency virus infections. Cold Spring Harb Perspect Med 2013; 2:a007153. [PMID: 22315718 DOI: 10.1101/cshperspect.a007153] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The simian immunodeficiency viruses (SIVs) are a diverse group of viruses that naturally infect a wide range of African primates, including African green monkeys (AGMs) and sooty mangabey monkeys (SMs). Although natural infection is widespread in feral populations of AGMs and SMs, this infection generally does not result in immunodeficiency. However, experimental inoculation of Asian macaques results in an immunodeficiency syndrome remarkably similar to human AIDS. Thus, natural nonprogressive SIV infections appear to represent an evolutionary adaptation between these animals and their primate lentiviruses. Curiously, these animals maintain robust virus replication but have evolved strategies to avoid disease progression. Adaptations observed in these primates include phenotypic changes to CD4(+) T cells, limited chronic immune activation, and altered mucosal immunity. It is probable that these animals have achieved a unique balance between T-cell renewal and proliferation and loss through activation-induced apoptosis, and virus-induced cell death. A clearer understanding of the mechanisms underlying the lack of disease progression in natural hosts for SIV infection should therefore yield insights into the pathogenesis of AIDS and may inform vaccine design.
Collapse
Affiliation(s)
- Nichole R Klatt
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
23
|
De Boer RJ, Perelson AS. Quantifying T lymphocyte turnover. J Theor Biol 2013; 327:45-87. [PMID: 23313150 PMCID: PMC3640348 DOI: 10.1016/j.jtbi.2012.12.025] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 12/13/2012] [Accepted: 12/30/2012] [Indexed: 01/13/2023]
Abstract
Peripheral T cell populations are maintained by production of naive T cells in the thymus, clonal expansion of activated cells, cellular self-renewal (or homeostatic proliferation), and density dependent cell life spans. A variety of experimental techniques have been employed to quantify the relative contributions of these processes. In modern studies lymphocytes are typically labeled with 5-bromo-2'-deoxyuridine (BrdU), deuterium, or the fluorescent dye carboxy-fluorescein diacetate succinimidyl ester (CFSE), their division history has been studied by monitoring telomere shortening and the dilution of T cell receptor excision circles (TRECs) or the dye CFSE, and clonal expansion has been documented by recording changes in the population densities of antigen specific cells. Proper interpretation of such data in terms of the underlying rates of T cell production, division, and death has proven to be notoriously difficult and involves mathematical modeling. We review the various models that have been developed for each of these techniques, discuss which models seem most appropriate for what type of data, reveal open problems that require better models, and pinpoint how the assumptions underlying a mathematical model may influence the interpretation of data. Elaborating various successful cases where modeling has delivered new insights in T cell population dynamics, this review provides quantitative estimates of several processes involved in the maintenance of naive and memory, CD4(+) and CD8(+) T cell pools in mice and men.
Collapse
Affiliation(s)
- Rob J De Boer
- Theoretical Biology & Bioinformatics, Utrecht University, The Netherlands; Santa Fe Institute, Santa Fe, NM 87501, USA.
| | | |
Collapse
|
24
|
Martinot AJ, Meythaler M, Pozzi LA, Dalecki Boisvert K, Knight H, Walsh D, Westmoreland S, Anderson DC, Kaur A, O'Neil SP. Acute SIV infection in sooty mangabey monkeys is characterized by rapid virus clearance from lymph nodes and absence of productive infection in germinal centers. PLoS One 2013; 8:e57785. [PMID: 23472105 PMCID: PMC3589484 DOI: 10.1371/journal.pone.0057785] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 01/24/2013] [Indexed: 11/23/2022] Open
Abstract
Lymphoid tissue immunopathology is a characteristic feature of chronic HIV/SIV infection in AIDS-susceptible species, but is absent in SIV-infected natural hosts. To investigate factors contributing to this difference, we compared germinal center development and SIV RNA distribution in peripheral lymph nodes during primary SIV infection of the natural host sooty mangabey and the non-natural host pig-tailed macaque. Although SIV-infected cells were detected in the lymph node of both species at two weeks post infection, they were confined to the lymph node paracortex in immune-competent mangabeys but were seen in both the paracortex and the germinal center of SIV-infected macaques. By six weeks post infection, SIV-infected cells were no longer detected in the lymph node of sooty mangabeys. The difference in localization and rate of disappearance of SIV-infected cells between the two species was associated with trapping of cell-free virus on follicular dendritic cells and higher numbers of germinal center CD4+ T lymphocytes in macaques post SIV infection. Our data suggests that fundamental differences in the germinal center microenvironment prevent productive SIV infection within the lymph node germinal centers of natural hosts contributing to sustained immune competency.
Collapse
Affiliation(s)
- Amanda J Martinot
- Division of Comparative Pathology, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Convergence and divergence in the evolution of the APOBEC3G-Vif interaction reveal ancient origins of simian immunodeficiency viruses. PLoS Pathog 2013; 9:e1003135. [PMID: 23359341 PMCID: PMC3554591 DOI: 10.1371/journal.ppat.1003135] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 12/03/2012] [Indexed: 01/05/2023] Open
Abstract
Naturally circulating lentiviruses are abundant in African primate species today, yet their origins and history of transmitting between hosts remain obscure. As a means to better understand the age of primate lentiviruses, we analyzed primate genomes for signatures of lentivirus-driven evolution. Specifically, we studied the adaptive evolution of host restriction factor APOBEC3G (A3G) in Old World Monkey (OWM) species. We find recurrent mutation of A3G in multiple primate lineages at sites that determine susceptibility to antagonism by the lentiviral accessory protein Vif. Using a broad panel of SIV Vif isolates, we demonstrate that natural variation in OWM A3G confers resistance to Vif-mediated degradation, suggesting that adaptive variants of the host factor were selected upon exposure to pathogenic lentiviruses at least 5–6 million years ago (MYA). Furthermore, in members of the divergent Colobinae subfamily of OWM, a multi-residue insertion event in A3G that arose at least 12 MYA blocks the activity of Vif, suggesting an even more ancient origin of SIV. Moreover, analysis of the lentiviruses associated with Colobinae monkeys reveal that the interface of the A3G-Vif interaction has shifted and given rise to a second genetic conflict. Our analysis of virus-driven evolution describes an ancient yet ongoing genetic conflict between simian primates and lentiviruses on a million-year time scale. The emergence of AIDS in the late 20th century has provoked studies to better understand the evolutionary history of viruses and the factors that govern their spread. Pandemic human immunodeficiency virus-type 1 (HIV-1), which currently infects 34 million people worldwide, emerged following the transmission of a lentivirus between chimpanzees and humans. A growing list of apparently nonpathogenic, species-specific strains has now been characterized in dozens of African primates, suggesting that primate lentiviruses are older and more widespread than originally thought. To estimate the extent to which primates and lentiviruses have coexisted, we examined the interaction between host and virus on a molecular level and tracked its dynamics over evolutionary time. We report that the immunity factor APOBEC3G is evolving in tandem with the lentiviral accessory gene vif, allowing us to associate instances of host evolution with instances of lentivirus infection in deep and shallow timescales. Specifically, we show that the region of APOBEC3G targeted by Vif is adaptively diversifying in independent primate lineages in a manner that suggests that lentiviruses are millions of years old. Our study reveals that, while primate lentiviruses may have modern consequences for human health, they have ancient origins in our non-human primate relatives.
Collapse
|
26
|
Abstract
Apoptosis of uninfected bystander cells is a key element of HIV pathogenesis and believed to be the driving force behind the selective depletion of CD4+ T cells leading to immunodeficiency. While several viral proteins have been implicated in this process the complex interaction between Env glycoprotein expressed on the surface of infected cells and the receptor and co-receptor expressing bystander cells has been proposed as a major mechanism. HIV-1 utilizes CD4 as the primary receptor for entry into cells; however, it is the viral co-receptor usage that greatly influences CD4 decline and progression to AIDS. This phenomenon is relatively simple for X4 viruses, which arise later during the course of the disease, are considered to be highly fusogenic, and cause a rapid CD4+ T cell decline. However, in contrast, R5 viruses in general have a greater transmissibility, are encountered early during the disease and have a lesser pathogenic potential than the former. The above generalization gets complicated in numerous situations where R5 viruses persist throughout the disease and are capable of causing a rigorous CD4+ T cell decline. This review will discuss the multiple factors that are reported to influence HIV induced bystander apoptosis and pathogenesis including Env glycoprotein phenotype, virus tropism, disease stage, co-receptor expression on CD4+ T cells, immune activation and therapies targeting the viral envelope.
Collapse
Affiliation(s)
- Himanshu Garg
- Center of Excellence for Infectious Disease, Department of Biomedical Science, Texas Tech University Health Sciences Center, 5001 El Paso Dr, MSB-1 Annex, El Paso, TX 79905, USA.
| | | | | |
Collapse
|
27
|
Chahroudi A, Bosinger SE, Vanderford TH, Paiardini M, Silvestri G. Natural SIV hosts: showing AIDS the door. Science 2012; 335:1188-93. [PMID: 22403383 DOI: 10.1126/science.1217550] [Citation(s) in RCA: 204] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Many species of African nonhuman primates are naturally infected with simian immunodeficiency viruses (SIVs) in the wild and in captivity. In contrast to HIV-infected humans, these natural SIV hosts typically do not develop AIDS, despite chronic infection with a highly replicating virus. In this Review, we discuss the most recent advances on the mechanisms of protection from disease progression in natural SIV hosts, with emphasis on how they differ from pathogenic HIV/SIV infections of humans and rhesus macaques. These mechanisms include: (i) resolution of immune activation after acute infection, (ii) restricted pattern of target cell infection, and (iii) protection from mother-to-infant transmission. We highlight the areas that should be pursued in future studies, focusing on potential applications for the treatment and prevention of HIV infection.
Collapse
Affiliation(s)
- Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
28
|
Abstract
Human immunodeficiency virus (HIV) pathogenesis has proven to be quite complex and dynamic with most of the critical events (e.g., transmission, CD4(+) T-cell destruction) occurring in mucosal tissues. In addition, although the resulting disease can progress over years, it is clear that many critical events happen within the first few weeks of infection when most patients are unaware that they are infected. These events occur predominantly in tissues other than the peripheral blood, particularly the gastrointestinal tract, where massive depletion of CD4(+) T cells occurs long before adverse consequences of HIV infection are otherwise apparent. Profound insights into these early events have been gained through the use of nonhuman primate models, which offer the opportunity to examine the early stages of infection with the simian immunodeficiency virus (SIV), a close relative of HIV that induces an indistinguishable clinical picture from AIDS in Asian primate species, but importantly, fails to cause disease in its natural African hosts, such as sooty mangabeys and African green monkeys. This article draws from data derived from both human and nonhuman primate studies.
Collapse
Affiliation(s)
- A A Lackner
- Tulane National Primate Research Center, Tulane University Health Science Center, Covington, LA 70443, USA.
| | | | | |
Collapse
|
29
|
Petrovas C, Yamamoto T, Gerner MY, Boswell KL, Wloka K, Smith EC, Ambrozak DR, Sandler NG, Timmer KJ, Sun X, Pan L, Poholek A, Rao SS, Brenchley JM, Alam SM, Tomaras GD, Roederer M, Douek DC, Seder RA, Germain RN, Haddad EK, Koup RA. CD4 T follicular helper cell dynamics during SIV infection. J Clin Invest 2012; 122:3281-94. [PMID: 22922258 DOI: 10.1172/jci63039] [Citation(s) in RCA: 278] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 07/05/2012] [Indexed: 12/16/2022] Open
Abstract
CD4 T follicular helper (TFH) cells interact with and stimulate the generation of antigen-specific B cells. TFH cell interaction with B cells correlates with production of SIV-specific immunoglobulins. However, the fate of TFH cells and their participation in SIV-induced antibody production is not well understood. We investigated the phenotype, function, location, and molecular signature of TFH cells in rhesus macaques. Similar to their human counterparts, TFH cells in rhesus macaques represented a heterogeneous population with respect to cytokine function. In a highly differentiated subpopulation of TFH cells, characterized by CD150lo expression, production of Th1 cytokines was compromised while IL-4 production was augmented, and cells exhibited decreased survival, cycling, and trafficking capacity. TFH cells exhibited a distinct gene profile that was markedly altered by SIV infection. TFH cells were infected by SIV; yet, in some animals, these cells actually accumulated during chronic SIV infection. Generalized immune activation and increased IL-6 production helped drive TFH differentiation during SIV infection. Accumulation of TFH cells was associated with increased frequency of activated germinal center B cells and SIV-specific antibodies. Therefore, chronic SIV does not disturb the ability of TFH cells to help B cell maturation and production of SIV-specific immunoglobulins.
Collapse
|
30
|
Memory CD4(+) T lymphocytes in the gastrointestinal tract are a major source of cell-associated simian immunodeficiency virus in chronic nonpathogenic infection of African green monkeys. J Virol 2012; 86:11380-5. [PMID: 22896600 DOI: 10.1128/jvi.01556-12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Simian immunodeficiency virus (SIV) infection of natural hosts is characterized by nonpathogenic chronic viremia, maintenance of gastrointestinal epithelial barrier integrity, and low numbers of target cells. Assessment of cell-associated virus load in T cell subsets in multiple anatomic compartments of chronically SIV-infected sabeus African green monkeys (AGMs) revealed that gastrointestinal memory CD4(+) T lymphocytes are a major source of cell-associated virus and a significant contributor to SIV viremia in AGMs.
Collapse
|
31
|
Taaffe JE, Bosinger SE, Del Prete GQ, Else JG, Ratcliffe S, Ward CD, Migone T, Paiardini M, Silvestri G. CCR5 blockade is well tolerated and induces changes in the tissue distribution of CCR5+ and CD25+ T cells in healthy, SIV-uninfected rhesus macaques. J Med Primatol 2011; 41:24-42. [PMID: 22077380 DOI: 10.1111/j.1600-0684.2011.00521.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND CCR5 is a main co-receptor for HIV, but also homes lymphocytes to sites of inflammation. We hypothesized that inhibition of CCR5 signaling would reduce HIV-associated chronic immune activation. METHODS To test this hypothesis, we administered an antagonistic anti-CCR5 monoclonal antibody (HGS101) to five uninfected rhesus macaques (RMs) and monitored lymphocyte dynamics in blood and tissue. RESULTS CCR5 blockade resulted in decreased levels of CCR5+ T cells in blood and, at later timepoints, in lymph nodes. Additionally, the levels of CD25+ T cells increased in lymph nodes, but decreased in blood, bone marrow, and rectal mucosa. Finally, a profile of gene expression from HGS101-treated RMs revealed a subtle, but consistent, in vivo signature of CCR5 blockade that suggests a mild immune-modulatory effect. CONCLUSIONS Treatment with anti-CCR5 antibody induces changes in the tissue distribution of CCR5+ and CD25+ T cells that may impact on the overall levels of immune activation during HIV and SIV infection.
Collapse
Affiliation(s)
- Jessica E Taaffe
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Preclinical safety evaluation of subretinal AAV2.sFlt-1 in non-human primates. Gene Ther 2011; 19:999-1009. [DOI: 10.1038/gt.2011.169] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
33
|
CD4-like immunological function by CD4- T cells in multiple natural hosts of simian immunodeficiency virus. J Virol 2011; 85:8702-8. [PMID: 21715501 DOI: 10.1128/jvi.00332-11] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Many species of African nonhuman primates are natural hosts for individual strains of simian immunodeficiency virus (SIV). These infected animals do not, however, develop AIDS. Here we show that multiple species of African nonhuman primate species characteristically have low frequencies of CD4(+) T cells and high frequencies of both T cells that express only the alpha-chain of CD8 and double-negative T cells. These subsets of T cells are capable of eliciting functions generally associated with CD4(+) T cells, yet these cells lack surface expression of the CD4 protein and are, therefore, poor targets for SIV in vivo. These data demonstrate that coevolution with SIV has, in several cases, involved downregulation of receptors for the virus by otherwise-susceptible host target cells. Understanding the genetic factors that lead to downregulation of these receptors may lead to therapeutic interventions that mimic this modulation in progressive infections.
Collapse
|
34
|
Weinberger AD, Perelson AS. Persistence and emergence of X4 virus in HIV infection. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2011; 8:605-626. [PMID: 21631149 PMCID: PMC3118547 DOI: 10.3934/mbe.2011.8.605] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Approximately 50% of late-stage HIV patients develop CXCR4-tropic (X4) virus in addition to CCR5-tropic (R5) virus. X4 emergence occurs with a sharp decline in CD4+ T cell counts and accelerated time to AIDS. Why this phenotypic switch to X4 occurs is not well understood. Previously, we used numerical simulations of a mathematical model to show that across much of parameter space a promising new class of antiretroviral treatments, CCR5 inhibitors, can accelerate X4 emergence and immunodeficiency. Here, we show that mathematical model to be a minimal activation-based HIV model that produces a spontaneous switch to X4 virus at a clinically-representative time point, while also matching in vivo data showing X4 and R5 coexisting and competing to infect memory CD4+ T cells. Our analysis shows that X4 avoids competitive exclusion from an initially fitter R5 virus due to X4v unique ability to productively infect nave CD4+ T cells. We further justify the generalized conditions under which this minimal model holds, implying that a phenotypic switch can even occur when the fraction of activated nave CD4+ T cells increases at a slower rate than the fraction of activated memory CD4+ T cells. We find that it is the ratio of the fractions of activated nave and memory CD4+ T cells that must increase above a threshold to produce a switch. This occurs as the concentration of CD4+ T cells drops beneath a threshold. Thus, highly active antiretroviral therapy (HAART), which increases CD4+ T cell counts and decreases cellular activation levels, inhibits X4 viral growth. However, we show here that even in the simplest dual-strain framework, competition between R5 and X4 viruses often results in accelerated X4 emergence in response to CCR5 inhibition, further highlighting the potential danger of anti-CCR5 monotherapy in multi-strain HIV infection.
Collapse
Affiliation(s)
- Ariel D Weinberger
- Graduate Group in Biophysics, University of California, Berkeley, Berkeley, CA 94720, United States.
| | | |
Collapse
|
35
|
Estes JD, Harris LD, Klatt NR, Tabb B, Pittaluga S, Paiardini M, Barclay GR, Smedley J, Pung R, Oliveira KM, Hirsch VM, Silvestri G, Douek DC, Miller CJ, Haase AT, Lifson J, Brenchley JM. Damaged intestinal epithelial integrity linked to microbial translocation in pathogenic simian immunodeficiency virus infections. PLoS Pathog 2010; 6:e1001052. [PMID: 20808901 PMCID: PMC2924359 DOI: 10.1371/journal.ppat.1001052] [Citation(s) in RCA: 393] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Accepted: 07/20/2010] [Indexed: 12/12/2022] Open
Abstract
The chronic phase of HIV infection is marked by pathological activation of the immune system, the extent of which better predicts disease progression than either plasma viral load or CD4(+) T cell count. Recently, translocation of microbial products from the gastrointestinal tract has been proposed as an underlying cause of this immune activation, based on indirect evidence including the detection of microbial products and specific immune responses in the plasma of chronically HIV-infected humans or SIV-infected Asian macaques. We analyzed tissues from SIV-infected rhesus macaques (RMs) to provide direct in situ evidence for translocation of microbial constituents from the lumen of the intestine into the lamina propria and to draining and peripheral lymph nodes and liver, accompanied by local immune responses in affected tissues. In chronically SIV-infected RMs this translocation is associated with breakdown of the integrity of the epithelial barrier of the gastrointestinal (GI) tract and apparent inability of lamina propria macrophages to effectively phagocytose translocated microbial constituents. By contrast, in the chronic phase of SIV infection in sooty mangabeys, we found no evidence of epithelial barrier breakdown, no increased microbial translocation and no pathological immune activation. Because immune activation is characteristic of the chronic phase of progressive HIV/SIV infections, these findings suggest that increased microbial translocation from the GI tract, in excess of capacity to clear the translocated microbial constituents, helps drive pathological immune activation. Novel therapeutic approaches to inhibit microbial translocation and/or attenuate chronic immune activation in HIV-infected individuals may complement treatments aimed at direct suppression of viral replication.
Collapse
Affiliation(s)
- Jacob D. Estes
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland, United States of America
- * E-mail: (JDE); (JMB)
| | - Levelle D. Harris
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Nichole R. Klatt
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Brian Tabb
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland, United States of America
| | - Stefania Pittaluga
- Lab of Pathology, NCI, NIH, Bethesda, Maryland, United States of America
| | - Mirko Paiardini
- Department of Pathology and Laboratory of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - G. Robin Barclay
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Jeremy Smedley
- Laboratory Animal Science Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland, United States of America
| | - Rhonda Pung
- Laboratory Animal Science Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland, United States of America
| | | | - Vanessa M. Hirsch
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Guido Silvestri
- Department of Pathology and Laboratory of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Daniel C. Douek
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Christopher J. Miller
- Center for Comparative Medicine, California National Primate Research Center, University of California, Davis, California, United States of America
| | - Ashley T. Haase
- Department of Microbiology, Medical School, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jeffrey Lifson
- AIDS and Cancer Virus Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland, United States of America
| | - Jason M. Brenchley
- Lab of Molecular Microbiology, NIAID, NIH, Bethesda, Maryland, United States of America
- * E-mail: (JDE); (JMB)
| |
Collapse
|
36
|
Nonprogressive and progressive primate immunodeficiency lentivirus infections. Immunity 2010; 32:737-42. [PMID: 20620940 DOI: 10.1016/j.immuni.2010.06.004] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Indexed: 12/17/2022]
Abstract
Natural hosts for simian immunodeficiency virus (SIV) can be, and are often naturally, infected with species-specific SIVs, but do not develop acquired immunodeficiency syndrome (AIDS). These natural hosts maintain high SIV viral loads, but avoid immunodeficiency. Elucidating the mechanisms that allow natural hosts to coexist with SIV without overt disease may provide crucial information for understanding AIDS pathogenesis. Over the past few years, several key features of natural SIV infections have been described in studies conducted predominantly in sooty mangabeys (SMs), African green monkeys (AGMs), and mandrills. Natural SIV hosts are able to avoid the chronic, generalized immune system activation that is associated with disease progression in HIV-infected individuals and are known to downmodulate the expression of the receptors for SIV. In this perspective we propose that a critical factor that differentiates nonprogressive from progressive HIV or SIV infection is the maintenance of T cell immune competence in the face of a virus that infects and kills CD4(+) T cells. Elucidation of the mechanisms underlying the preservation of immune function during and after the acute phase of natural SIV infection may lead to the design of novel preventive and therapeutic interventions for treatment of chronic HIV infection.
Collapse
|
37
|
Chan ML, Petravic J, Ortiz AM, Engram J, Paiardini M, Cromer D, Silvestri G, Davenport MP. Limited CD4+ T cell proliferation leads to preservation of CD4+ T cell counts in SIV-infected sooty mangabeys. Proc Biol Sci 2010; 277:3773-81. [PMID: 20591864 DOI: 10.1098/rspb.2010.0972] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) infections result in chronic virus replication and progressive depletion of CD4+ T cells, leading to immunodeficiency and death. In contrast, 'natural hosts' of SIV experience persistent infection with high virus replication but no severe CD4+ T cell depletion, and remain AIDS-free. One important difference between pathogenic and non-pathogenic infections is the level of activation and proliferation of CD4+ T cells. We analysed the relationship between CD4+ T cell number and proliferation in HIV, pathogenic SIV in macaques, and non-pathogenic SIV in sooty mangabeys (SMs) and mandrills. We found that CD4+ T cell proliferation was negatively correlated with CD4+ T cell number, suggesting that animals respond to the loss of CD4+ T cells by increasing the proliferation of remaining cells. However, the level of proliferation seen in pathogenic infections (SIV in rhesus macaques and HIV) was much greater than in non-pathogenic infections (SMs and mandrills). We then used a modelling approach to understand how the host proliferative response to CD4+ T cell depletion may impact the outcome of infection. This modelling demonstrates that the rapid proliferation of CD4+ T cells in humans and macaques associated with low CD4+ T cell levels can act to 'fuel the fire' of infection by providing more proliferating cells for infection. Natural host species, on the other hand, have limited proliferation of CD4+ T cells at low CD4+ T cell levels, which allows them to restrict the number of proliferating cells susceptible to infection.
Collapse
Affiliation(s)
- Ming Liang Chan
- Complex Systems in Biology Group, Centre for Vascular Research, University of New South Wales, Kensington, NSW, Australia
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Shedlock DJ, Talbott KT, Morrow MP, Ferraro B, Hokey DA, Muthumani K, Weiner DB. Ki-67 staining for determination of rhesus macaque T cell proliferative responses ex vivo. Cytometry A 2010; 77:275-84. [PMID: 20104580 DOI: 10.1002/cyto.a.20857] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The capacity for robust proliferation upon re-infection is a hallmark of adaptive immunity and the basis of vaccination. A widely used animal model for the study of human disease is the rhesus macaque (RM), where capacity for proliferation can be assessed ex vivo using carboxyfluorescein succinimidyl ester (CFSE)-based dilution assays. However, we show over the course of the standard ex vivo proliferation assay that CFSE-labeling at commonly used dye concentrations induces significant cell death, but that this phenomenon is dose-dependent. Here, we describe an alternative semiquantitative method for estimating T cell proliferative responses that avoids the putative biases associated with chemical modification. RM peripheral blood mononuclear cells were stimulated ex vivo with cognate peptides for 5 days, immunostained for intracellular Ki-67, and then analyzed by flow cytometry. We describe a gating strategy using Ki-67 and side light scatter, also a marker of blastogenesis, which correlates strongly with data from CFSE dilution. We show that this method is a valid tool for measuring RM antigen-specific cellular proliferation ex vivo and can be used as an alternative to CFSE dilution assays.
Collapse
Affiliation(s)
- Devon J Shedlock
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Gaufin T, Ribeiro RM, Gautam R, Dufour J, Mandell D, Apetrei C, Pandrea I. Experimental depletion of CD8+ cells in acutely SIVagm-infected African Green Monkeys results in increased viral replication. Retrovirology 2010; 7:42. [PMID: 20459829 PMCID: PMC2879233 DOI: 10.1186/1742-4690-7-42] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 05/11/2010] [Indexed: 11/22/2022] Open
Abstract
Background In vivo CD8+ cell depletions in pathogenic SIV infections identified a key role for cellular immunity in controlling viral load (VL) and disease progression. However, similar studies gave discordant results in chronically-infected SMs, leading some authors to propose that in natural hosts, SIV replication is independent of cellular immunity. To assess the role of cellular immune responses in the control of SIV replication in natural hosts, we investigated the impact of CD8+ cell depletion during acute SIV infection in AGMs. Results Nine AGMs were infected with SIVagm.sab and were followed up to day 225 p.i. Four were intravenously infused with the cM-T807 antibody on days 0 (50 mg/kg), 6, and 13 (10 mg/kg, respectively) post infection (p.i.). CD8+ cells were depleted for up to 28 days p.i. in peripheral blood and LNs in all treated AGMs. Partial CD8+ T cell depletion occurred in the intestine. SIVagm VLs peaked at similar levels in both groups (107-108 RNA copies/ml). However, while VLs were controlled in undepleted AGMs, reaching set-point levels (104-105 RNA copies/ml) by day 28 p.i., high VLs (>106 RNA copies/ml) were maintained by day 21 p.i. in CD8-depleted AGMs. By day 42 p.i., VLs were comparable between the two groups. The levels of immune activation and proliferation remained elevated up to day 72 p.i. in CD8-depleted AGMs and returned to preinfection levels in controls by day 28 p.i. None of the CD8-depleted animals progressed to AIDS. Conclusion CD8+ cells are responsible for a partial control of postacute viral replication in SIVagm.sab-infected AGMs. In contrast to macaques, the SIVagm-infected AGMs are able to control viral replication after recovery of the CD8+ T cells and avoid disease progression.
Collapse
Affiliation(s)
- Thaidra Gaufin
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
A five-year longitudinal analysis of sooty mangabeys naturally infected with simian immunodeficiency virus reveals a slow but progressive decline in CD4+ T-cell count whose magnitude is not predicted by viral load or immune activation. J Virol 2010; 84:5476-84. [PMID: 20335252 DOI: 10.1128/jvi.00039-10] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Natural simian immunodeficiency virus (SIV) infection in sooty mangabeys (SMs) typically does not result in AIDS, despite high-level viremia and significant depletion of mucosal CD4(+) T cells. Here, we report the results of the first longitudinal study of a large cohort of SMs naturally infected with SIV (n = 78) housed at the Yerkes National Primate Research Center from which samples were obtained three times over a 5-year period. In this study, we observed (i) no signs of simian AIDS, (ii) stable SIV loads, (iii) a slow but progressive decline in CD4(+) T-cell counts (from a mean of 1,067.0 cells/mm(3) at time point 1 to 764.8 cells/mm(3) at time point 3) and increases in the numbers of animals with CD4(+) T-cell levels below 500 and 200 cells/mm(3) (from 8 to 28 of 78 and from 1 to 4 of 78, respectively), (iv) progressive declines in percentages of naïve CD4(+) and CD8(+) T cells (from 37.7 to 24.8% and from 21.0 to 13.0%, respectively), and (v) stably low levels of activated/proliferating T cells as well as CD4(+) CCR5(+) T cells. Since the level of total CD4(+) T cells and the fraction of naïve T cells in SIV-uninfected SMs also declined, it is possible that some of these observations are related to aging, as the SIV-infected animals were significantly older than the uninfected animals. In contrast to the decline in CD4(+) T cell counts in individuals infected with human immunodeficiency virus (HIV), the decline in CD4(+) T cell counts in SMs naturally infected with SIV over a 5-year period was not predicted by either plasma viremia or levels of T-cell activation. Taken together, these results confirm that natural SIV infection is nonprogressive from a clinical, virological, and immunological point of view and that stable levels of viremia associated with persistently low-level immune activation represent key differences from the natural course of HIV infection in humans.
Collapse
|
41
|
Rout N, Else JG, Yue S, Connole M, Exley MA, Kaur A. Paucity of CD4+ natural killer T (NKT) lymphocytes in sooty mangabeys is associated with lack of NKT cell depletion after SIV infection. PLoS One 2010; 5:e9787. [PMID: 20352088 PMCID: PMC2844411 DOI: 10.1371/journal.pone.0009787] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 03/02/2010] [Indexed: 12/22/2022] Open
Abstract
Lack of chronic immune activation in the presence of persistent viremia is a key feature that distinguishes nonpathogenic simian immunodeficiency virus (SIV) infection in natural hosts from pathogenic SIV and HIV infection. To elucidate novel mechanisms downmodulating immune activation in natural hosts of SIV infection, we investigated natural killer T (NKT) lymphocytes in sooty mangabeys. NKT lymphocytes are a potent immunoregulatory arm of the innate immune system that recognize glycolipid antigens presented on the nonpolymorphic MHC-class I-like CD1d molecules. In a cross-sectional analysis of 50 SIV-negative and 50 naturally SIV-infected sooty mangabeys, ligand alpha-galactosylceramide loaded CD1d tetramers co-staining with Valpha24-positive invariant NKT lymphocytes were detected at frequencies >or=0.002% of circulating T lymphocytes in approximately half of the animals. In contrast to published reports in Asian macaques, sooty mangabey NKT lymphocytes consisted of CD8(+) and CD4/CD8 double-negative T lymphocytes that were CXCR3-positive and CCR5-negative suggesting that they trafficked to sites of inflammation without being susceptible to SIV infection. Consistent with these findings, there was no difference in the frequency or phenotype of NKT lymphocytes between SIV-negative and SIV-infected sooty mangabeys. On stimulation with alpha-galactosylceramide loaded on human CD1d molecules, sooty mangabey NKT lymphocytes underwent degranulation and secreted IFN-gamma, TNF-alpha, IL-2, IL-13, and IL-10, indicating the presence of both effector and immunoregulatory functional capabilities. The unique absence of CD4(+) NKT lymphocytes in sooty mangabeys, combined with their IL-10 cytokine-secreting ability and preservation following SIV infection, raises the possibility that NKT lymphocytes might play a role in downmodulating immune activation in SIV-infected sooty mangabeys.
Collapse
Affiliation(s)
- Namita Rout
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
| | - James G. Else
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Simon Yue
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michelle Connole
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
| | - Mark A. Exley
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Amitinder Kaur
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
42
|
Paiardini M, Pandrea I, Apetrei C, Silvestri G. Lessons learned from the natural hosts of HIV-related viruses. Annu Rev Med 2009; 60:485-95. [PMID: 19630581 DOI: 10.1146/annurev.med.60.041807.123753] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The fact that human immunodeficiency virus (HIV) causes a deadly disease in humans whereas its simian counterparts, the simian immunodeficiency viruses (SIVs), are virtually nonpathogenic in their natural hosts remains a fundamental mystery of modern medicine. Arguably, the pathogenesis of HIV infection will remain poorly understood until the mechanisms responsible for the AIDS resistance of natural SIV hosts are fully explained. Over the past few years, some key features of natural SIV infections have been described in studies conducted predominantly in sooty mangabeys (SMs), African green monkeys (AGMs), and mandrills. Natural SIV hosts are able to avoid the chronic, generalized immune system activation that is associated with disease progression in HIV-infected individuals and have evolved to down-modulate the expression of CCR5 on CD4(+) T cells. Better elucidation of the mechanisms underlying the lack of disease progression of natural SIV infections holds promise for the design of novel preventive and therapeutic approaches to HIV infection.
Collapse
Affiliation(s)
- Mirko Paiardini
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
43
|
Joyce EA, Popper SJ, Falkow S. Streptococcus pneumoniae nasopharyngeal colonization induces type I interferons and interferon-induced gene expression. BMC Genomics 2009; 10:404. [PMID: 19712482 PMCID: PMC2743716 DOI: 10.1186/1471-2164-10-404] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 08/27/2009] [Indexed: 01/22/2023] Open
Abstract
Background We employed DNA microarray technology to investigate the host response to Streptococcus pneumoniae in a mouse model of asymptomatic carriage. Over a period of six weeks, we profiled transcript abundance and complexity in the Nasal Associated Lymphoid Tissue (NALT) to identify genes whose expression differed between pneumococcal-colonized and uncolonized states. Results Colonization with S. pneumoniae altered the expression of hundreds of genes over the course of the study, demonstrating that carriage is a dynamic process characterized by increased expression of a set of early inflammatory responses, including induction of a Type I Interferon response, and the production of several antimicrobial factors. Subsequent to this initial inflammatory response, we observed increases in transcripts associated with T cell development and activation, as well as wounding, basement membrane remodeling, and cell proliferation. Our analysis suggests that microbial colonization induced expression of genes encoding components critical for controlling JAK/STAT signaling, including stat1, stat2, socs3, and mapk1, as well as induction of several Type I Interferon-inducible genes and other antimicrobial factors at the earliest stages of colonization. Conclusion Examining multiple time points over six weeks of colonization demonstrated that asymptomatic carriage stimulates a dynamic host response characterized by temporal waves with distinct biological programs. Our data suggest that the usual response to the presence of the pneumocccus is an initial controlled inflammatory response followed by activation of host physiological processes such as response to wounding, basement membrane remodeling, and increasing cellular numbers that ultimately allow the host to maintain an intact epithelium and eventually mount a preventive adaptive immune response.
Collapse
Affiliation(s)
- Elizabeth A Joyce
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94131, USA.
| | | | | |
Collapse
|
44
|
Sodora DL, Allan JS, Apetrei C, Brenchley JM, Douek DC, Else JG, Estes JD, Hahn BH, Hirsch VM, Kaur A, Kirchhoff F, Muller-Trutwin M, Pandrea I, Schmitz JE, Silvestri G. Toward an AIDS vaccine: lessons from natural simian immunodeficiency virus infections of African nonhuman primate hosts. Nat Med 2009; 15:861-5. [PMID: 19661993 DOI: 10.1038/nm.2013] [Citation(s) in RCA: 182] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The design of an effective AIDS vaccine has eluded the efforts of the scientific community to the point that alternative approaches to classic vaccine formulations have to be considered. We propose here that HIV vaccine research could greatly benefit from the study of natural simian immunodeficiency virus (SIV) infections of African nonhuman primates. Natural SIV hosts (for example, sooty mangabeys, African green monkeys and mandrills) share many features of HIV infection of humans; however, they usually do not develop immunodeficiency. These natural, nonprogressive SIV infections represent an evolutionary adaptation that allows a peaceful coexistence of primate lentiviruses and the host immune system. This adaptation does not result in reduced viral replication but, rather, involves phenotypic changes to CD4(+) T cell subsets, limited immune activation and preserved mucosal immunity, all of which contribute to the avoidance of disease progression and, possibly, to the reduction of vertical SIV transmission. Here we summarize the current understanding of SIV infection of African nonhuman primates and discuss how unraveling these evolutionary adaptations may provide clues for new vaccine designs that might induce effective immune responses without the harmful consequences of excessive immune activation.
Collapse
Affiliation(s)
- Donald L Sodora
- Seattle Biomedical Research Institute, Seattle, Washington, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Siegismund CS, Hohn O, Kurth R, Norley S. Enhanced T- and B-cell responses to simian immunodeficiency virus (SIV)agm, SIVmac and human immunodeficiency virus type 1 Gag DNA immunization and identification of novel T-cell epitopes in mice via codon optimization. J Gen Virol 2009; 90:2513-2518. [PMID: 19587137 DOI: 10.1099/vir.0.013730-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
As a prelude to primate studies, the immunogenicity of wild-type and codon-optimized versions of simian immunodeficiency virus (SIV)agm Gag DNA, with and without co-administered granulocyte-macrophage colony-stimulating factor (GM-CSF) DNA, was directly compared in two strains of mice. Gag-specific T cells in the splenocytes of BALB/c and C57BL/6 mice immunized by gene gun were quantified by ELISpot using panels of overlapping synthetic peptides (15mers) spanning the entire capsid proteins of SIVagm, SIVmac and human immunodeficiency virus type 1. Specific antibodies were measured by ELISA. Codon optimization was shown to significantly increase the immune response to the DNA immunogens, reducing the amount of DNA necessary to induce cellular and antibody responses by one and two orders of magnitude, respectively. Co-administration of murine GM-CSF DNA was necessary for the induction of high level T- and B-cell responses. Finally, it was possible to identify both known and novel T-cell epitopes in the Gag proteins of the three viruses.
Collapse
Affiliation(s)
| | - Oliver Hohn
- Robert Koch-Institute, Nordufer 20, 13353 Berlin, Germany
| | - Reinhard Kurth
- Robert Koch-Institute, Nordufer 20, 13353 Berlin, Germany
| | - Stephen Norley
- Robert Koch-Institute, Nordufer 20, 13353 Berlin, Germany
| |
Collapse
|
46
|
Beaumier CM, Harris LD, Goldstein S, Klatt NR, Whitted S, McGinty J, Apetrei C, Pandrea I, Hirsch VM, Brenchley JM. CD4 downregulation by memory CD4+ T cells in vivo renders African green monkeys resistant to progressive SIVagm infection. Nat Med 2009; 15:879-85. [PMID: 19525963 PMCID: PMC2723181 DOI: 10.1038/nm.1970] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Accepted: 04/28/2009] [Indexed: 02/07/2023]
Abstract
African green monkeys (genus Chlorocebus) can be infected with SIVagm, but do not develop AIDS. This natural host of SIV, like sooty mangabeys, maintains high levels of SIV replication but has evolved to avoid immunodeficiency. Elucidating the mechanisms that allow the natural hosts to co-exist with SIV without overt disease may provide crucial information to understand AIDS pathogenesis. Here we show: (1) many CD4+ T cells from African green monkeys down-regulate CD4 in vivo as they enter the memory pool, (2) down regulation of CD4 by memory T cells is independent of SIV infection, (3) the CD4− memory T cells maintain functions which are normally attributed to CD4 T cells including production of IL-2, production of IL-17, expression of FoxP3 and expression of CD40L (4) loss of CD4 expression protects these T cells from infection by SIVagm in vivo, and (5) these CD4− T cells can maintain MHC-II restriction. These data demonstrate that the absence of SIV-induced disease progression in natural hosts species may be partially explained by preservation of a subset of T cells that maintain CD4 T cell function while being resistant to SIV-infection in vivo.
Collapse
Affiliation(s)
- Coreen M Beaumier
- Lab of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Asquith B, Borghans JAM, Ganusov VV, Macallan DC. Lymphocyte kinetics in health and disease. Trends Immunol 2009; 30:182-9. [PMID: 19286425 DOI: 10.1016/j.it.2009.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 01/11/2009] [Accepted: 01/12/2009] [Indexed: 12/22/2022]
Abstract
Quantitative understanding of immunology requires the development of experimental and mathematical techniques for estimation of rates of division and death of lymphocytes under different conditions. Here, we review the advantages and limitations of several labelling methods that are currently used to quantify turnover of lymphocytes in vivo. In addition to highlighting insights into lymphocyte kinetics which have recently been gained thanks to the development of novel techniques, we discuss important directions for future experimental and theoretical work in the field of lymphocyte turnover.
Collapse
Affiliation(s)
- Becca Asquith
- Department of Immunology, Wright-Fleming Institute, Imperial College London, London W2 1PG, UK
| | | | | | | |
Collapse
|
48
|
Parameters of Soluble Immune Activation In Vivo Correlate Negatively With the Proliferative Capacity of Peripheral Blood Mononuclear Cells In Vitro in HIV-Infected Patients. J Acquir Immune Defic Syndr 2009; 50:354-9. [DOI: 10.1097/qai.0b013e318198a6e1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
49
|
Abstract
Despite many years of intense scientific effort, the pathogenic mechanisms underlying the immunodeficiency that follows human immunodeficiency virus (HIV) infection are still poorly understood. This lack of understanding is likely the main reason why at present there is neither a cure nor a vaccine for AIDS. Important clues on the immunopathogenesis of primate lentiviral infections have been provided by comparative studies of two simian models of SIV infection: the non-pathogenic SIV infection of sooty mangabey, an African natural host species, and the pathogenic SIV-infection of non-natural host rhesus macaques, that develop a disease that closely resembles AIDS in humans. While the final mechanisms underlying the difference in infection outcome between these two species are still incompletely understood, a series of recent studies has allowed the identification of key similarities and differences between the two models of infection. In this article we summarize these findings and review the main implications in terms of HIV pathogenesis, therapy, and vaccines.
Collapse
Affiliation(s)
- Guido Silvestri
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Finnefrock AC, Tang A, Li F, Freed DC, Feng M, Cox KS, Sykes KJ, Guare JP, Miller MD, Olsen DB, Hazuda DJ, Shiver JW, Casimiro DR, Fu TM. PD-1 blockade in rhesus macaques: impact on chronic infection and prophylactic vaccination. THE JOURNAL OF IMMUNOLOGY 2009; 182:980-7. [PMID: 19124741 DOI: 10.4049/jimmunol.182.2.980] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Programmed Cell Death 1 (PD-1) plays a crucial role in immunomodulation. Binding of PD-1 to its ligand receptors down-regulates immune responses, and published reports suggest that this immune modulation is exploited in cases of tumor progression or chronic viral infection to evade immune surveillance. Thus, blockade of this signal could restore or enhance host immune functions. To test this hypothesis, we generated a panel of mAbs specific to human PD-1 that block PD ligand 1 and tested them for in vitro binding, blocking, and functional T cell responses, and evaluated a lead candidate in two in vivo rhesus macaque (Macaca mulatta) models. In the first therapeutic model, chronically SIV-infected macaques were treated with a single infusion of anti-PD-1 mAb; viral loads increased transiently before returning to, or falling below, pretreatment baselines. In the second prophylactic model, naive macaques were immunized with an SIV-gag adenovirus vector vaccine. Induced PD-1 blockade caused a statistically significant (p<0.05) increase in the peak percentage of T cells specific for the CM9 Gag epitope. These new results on PD-1 blockade in nonhuman primates point to a broader role for PD-1 immunomodulation and to potential applications in humans.
Collapse
Affiliation(s)
- Adam C Finnefrock
- Vaccine Basic Research, Merck Research Laboratories, West Point, PA 19486, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|