1
|
Ceriani C, Beisner B, Crane M, Cohen J, Moore IN, Kulpa DA, Hahn BH, Silvestri G. SIV infection in sooty mangabeys does not impact survival but changes the relative frequency of the main cause of death. mBio 2024; 15:e0163924. [PMID: 39258922 PMCID: PMC11481876 DOI: 10.1128/mbio.01639-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024] Open
Abstract
Sooty mangabeys (SMs) are natural hosts of simian immunodeficiency virus (SIV) and do not progress to AIDS despite high viral replication. The main factors involved in the benign nature of this infection are (i) low level of immune activation, (ii) relative preservation of specific CD4+ T-cell subsets from direct virus infection, and (iii) absence of microbial translocation from the gut to the systemic circulation. To determine the impact of SIV infection on underlying cause of death, we retrospectively analyzed data from 307 SMs (219 SIV infected and 88 uninfected) housed at the Emory Primate Center that have died between 1986 and 2022. Interestingly, we found that SIV-infected SMs live ~4 years longer than SIV-uninfected SMs, although this result is hard to interpret due to differences in how animals were housed and assigned to specific experimental studies. While the causes of death were not different between SIV-infected and uninfected SMs that died before age 15 (i.e., adult), we found significant differences in the relative frequency of specific causes of death in the elderly population (≥15 years old). Specifically, we observed that SIV-infected SMs were more likely to die from infections but less likely to die from cardiovascular disease (and diabetes in female animals) as compared to uninfected SMs. While confirming the non-pathogenic nature of SIV infection in SMs, these data reveal, for the first time, a qualitative impact of SIV infection on the host physiology that induces a significant change in the mortality pattern in these natural SIV hosts. IMPORTANCE In this study, we demonstrate, for the first time, that the natural, non-pathogenic SIV infection of the African monkey SM has a clinical impact which is revealed in terms of main causes of mortality, which are significantly different in the infected animals as compared to the uninfected ones. Indeed, SIV-infected SMs are at higher risk of dying of infectious diseases but appear to be somewhat protected from cardiovascular causes of death. The identification of a specific pattern of mortality associated with the infection suggests that the host-pathogen interaction between SIV and the SM immune system, while non-pathogenic in nature, has a detectable impact on the overall health status of the animals.
Collapse
Affiliation(s)
- Cristina Ceriani
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Brianne Beisner
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Maria Crane
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Joyce Cohen
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Ian N. Moore
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Deanna A. Kulpa
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Beatrice H. Hahn
- Departments of Medicine and Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Guido Silvestri
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
2
|
Obregon-Perko V, Mannino A, Ladner JT, Hodara V, Ebrahimi D, Parodi L, Callery J, Palacios G, Giavedoni LD. Adaptation of SIVmac to baboon primary cells results in complete absence of in vivo baboon infectivity. Front Cell Infect Microbiol 2024; 14:1408245. [PMID: 39006742 PMCID: PMC11239360 DOI: 10.3389/fcimb.2024.1408245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/04/2024] [Indexed: 07/16/2024] Open
Abstract
While simian immunodeficiency virus (SIV) infection is non-pathogenic in naturally infected African nonhuman primate hosts, experimental or accidental infection in rhesus macaques often leads to AIDS. Baboons, widely distributed throughout Africa, do not naturally harbor SIV, and experimental infection of baboons with SIVmac results in transient low-level viral replication. Elucidation of mechanisms of natural immunity in baboons could uncover new targets of antiviral intervention. We tested the hypothesis that an SIVmac adapted to replicate in baboon primary cells will gain the capacity to establish chronic infections in vivo. Here, we generated SIVmac variants in baboon cells through serial passage in PBMC from different donors (SIVbn-PBMC s1), in PBMC from the same donors (SIVbn-PBMC s2), or in isolated CD4 cells from the same donors used for series 2 (SIVbn-CD4). While SIVbn-PBMC s1 and SIVbn-CD4 demonstrated increased replication capacity, SIVbn-PBMC s2 did not. Pharmacological blockade of CCR5 revealed SIVbn-PBMC s1 could more efficiently use available CCR5 than SIVmac, a trait we hypothesize arose to circumvent receptor occupation by chemokines. Sequencing analysis showed that all three viruses accumulated different types of mutations, and that more non-synonymous mutations became fixed in SIVbn-PBMC s1 than SIVbn-PBMC s2 and SIVbn-CD4, supporting the notion of stronger fitness pressure in PBMC from different genetic backgrounds. Testing the individual contribution of several newly fixed SIV mutations suggested that is the additive effect of these mutations in SIVbn-PBMC s1 that contributed to its enhanced fitness, as recombinant single mutant viruses showed no difference in replication capacity over the parental SIVmac239 strain. The replicative capacity of SIVbn-PBMC passage 4 (P4) s1 was tested in vivo by infecting baboons intravenously with SIVbn-PBMC P4 s1 or SIVmac251. While animals infected with SIVmac251 showed the known pattern of transient low-level viremia, animals infected with SIVbn-PBMC P4 s1 had undetectable viremia or viral DNA in lymphoid tissue. These studies suggest that adaptation of SIV to grow in baboon primary cells results in mutations that confer increased replicative capacity in the artificial environment of cell culture but make the virus unable to avoid the restrictive factors generated by a complex multicellular organism.
Collapse
Affiliation(s)
| | - Amanda Mannino
- Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jason T. Ladner
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| | - Vida Hodara
- Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Diako Ebrahimi
- Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Laura Parodi
- Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jessica Callery
- Department of Biology, Trinity University, San Antonio, TX, United States
| | - Gustavo Palacios
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Luis D. Giavedoni
- Department of Biology, Trinity University, San Antonio, TX, United States
| |
Collapse
|
3
|
Symmonds J, Gaufin T, Xu C, Raehtz KD, Ribeiro RM, Pandrea I, Apetrei C. Making a Monkey out of Human Immunodeficiency Virus/Simian Immunodeficiency Virus Pathogenesis: Immune Cell Depletion Experiments as a Tool to Understand the Immune Correlates of Protection and Pathogenicity in HIV Infection. Viruses 2024; 16:972. [PMID: 38932264 PMCID: PMC11209256 DOI: 10.3390/v16060972] [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: 03/27/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Understanding the underlying mechanisms of HIV pathogenesis is critical for designing successful HIV vaccines and cure strategies. However, achieving this goal is complicated by the virus's direct interactions with immune cells, the induction of persistent reservoirs in the immune system cells, and multiple strategies developed by the virus for immune evasion. Meanwhile, HIV and SIV infections induce a pandysfunction of the immune cell populations, making it difficult to untangle the various concurrent mechanisms of HIV pathogenesis. Over the years, one of the most successful approaches for dissecting the immune correlates of protection in HIV/SIV infection has been the in vivo depletion of various immune cell populations and assessment of the impact of these depletions on the outcome of infection in non-human primate models. Here, we present a detailed analysis of the strategies and results of manipulating SIV pathogenesis through in vivo depletions of key immune cells populations. Although each of these methods has its limitations, they have all contributed to our understanding of key pathogenic pathways in HIV/SIV infection.
Collapse
Affiliation(s)
- Jen Symmonds
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Thaidra Gaufin
- Tulane National Primate Research Center, Tulane University, Covington, LA 70433, USA;
| | - Cuiling Xu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Kevin D. Raehtz
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ruy M. Ribeiro
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Ivona Pandrea
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.S.); (C.X.); (K.D.R.); (I.P.)
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Cristian Apetrei
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
4
|
Fonseca JA, King AC, Chahroudi A. More than the Infinite Monkey Theorem: NHP Models in the Development of a Pediatric HIV Cure. Curr HIV/AIDS Rep 2024; 21:11-29. [PMID: 38227162 PMCID: PMC10859349 DOI: 10.1007/s11904-023-00686-6] [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] [Accepted: 12/29/2023] [Indexed: 01/17/2024]
Abstract
PURPOSE OF REVIEW An HIV cure that eliminates the viral reservoir or provides viral control without antiretroviral therapy (ART) is an urgent need in children as they face unique challenges, including lifelong ART adherence and the deleterious effects of chronic immune activation. This review highlights the importance of nonhuman primate (NHP) models in developing an HIV cure for children as these models recapitulate the viral pathogenesis and persistence. RECENT FINDINGS Several cure approaches have been explored in infant NHPs, although knowledge gaps remain. Broadly neutralizing antibodies (bNAbs) show promise for controlling viremia and delaying viral rebound after ART interruption but face administration challenges. Adeno-associated virus (AAV) vectors hold the potential for sustained bNAb expression. Therapeutic vaccination induces immune responses against simian retroviruses but has yet to impact the viral reservoir. Combining immunotherapies with latency reversal agents (LRAs) that enhance viral antigen expression should be explored. Current and future cure approaches will require adaptation for the pediatric immune system and unique features of virus persistence, for which NHP models are fundamental to assess their efficacy.
Collapse
Affiliation(s)
- Jairo A Fonseca
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Alexis C King
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA.
- Emory+Children's Center for Childhood Infections and Vaccines, Atlanta, GA, USA.
| |
Collapse
|
5
|
Mazzuti L, Turriziani O, Mezzaroma I. The Many Faces of Immune Activation in HIV-1 Infection: A Multifactorial Interconnection. Biomedicines 2023; 11:biomedicines11010159. [PMID: 36672667 PMCID: PMC9856151 DOI: 10.3390/biomedicines11010159] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/10/2023] Open
Abstract
Chronic immune activation has a significant role in HIV-1 disease pathogenesis and CD4+ T-cell depletion. The causes of chronic inflammation and immune activation are incompletely understood, but they are likely multifactorial in nature, involving both direct and indirect stimuli. Possible explanations include microbial translocation, coinfection, and continued presence of competent replicating virus. In fact, long-term viral suppression treatments are unable to normalize elevated markers of systemic immune activation. Furthermore, high levels of pro-inflammatory cytokines increase susceptibility to premature aging of the immune system. The phenomenon of "inflammaging" has begun to be evident in the last decades, as a consequence of increased life expectancy due to the introduction of cART. Quality of life and survival have improved substantially; however, PLWH are predisposed to chronic inflammatory conditions leading to age-associated diseases, such as inflammatory bowel disease, neurocognitive disorders, cardiovascular diseases, metabolic syndrome, bone abnormalities, and non-HIV-associated cancers. Several approaches have been studied in numerous uncontrolled and/or randomized clinical trials with the aim of reducing immune activation/inflammatory status in PLWH, none of which have achieved consistent results.
Collapse
Affiliation(s)
- Laura Mazzuti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Ombretta Turriziani
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Ivano Mezzaroma
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
6
|
Kleinman AJ, Pandrea I, Apetrei C. So Pathogenic or So What?-A Brief Overview of SIV Pathogenesis with an Emphasis on Cure Research. Viruses 2022; 14:135. [PMID: 35062339 PMCID: PMC8781889 DOI: 10.3390/v14010135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/10/2021] [Accepted: 12/25/2021] [Indexed: 02/07/2023] Open
Abstract
HIV infection requires lifelong antiretroviral therapy (ART) to control disease progression. Although ART has greatly extended the life expectancy of persons living with HIV (PWH), PWH nonetheless suffer from an increase in AIDS-related and non-AIDS related comorbidities resulting from HIV pathogenesis. Thus, an HIV cure is imperative to improve the quality of life of PWH. In this review, we discuss the origins of various SIV strains utilized in cure and comorbidity research as well as their respective animal species used. We briefly detail the life cycle of HIV and describe the pathogenesis of HIV/SIV and the integral role of chronic immune activation and inflammation on disease progression and comorbidities, with comparisons between pathogenic infections and nonpathogenic infections that occur in natural hosts of SIVs. We further discuss the various HIV cure strategies being explored with an emphasis on immunological therapies and "shock and kill".
Collapse
Affiliation(s)
- Adam J. Kleinman
- Division of Infectious Diseases, DOM, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Ivona Pandrea
- Department of Infectious Diseases and Immunology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Cristian Apetrei
- Division of Infectious Diseases, DOM, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Department of Infectious Diseases and Immunology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| |
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: 5.0] [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
|
Control of HIV-1 Pathogenesis in Viremic Nonprogressors Is Independent of Gag-Specific Cytotoxic T Lymphocyte Responses. J Virol 2018; 92:JVI.00346-18. [PMID: 29593044 PMCID: PMC5974496 DOI: 10.1128/jvi.00346-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/21/2018] [Indexed: 01/01/2023] Open
Abstract
Viremic nonprogressors (VNPs) constitute a very scarce group of untreated human immunodeficiency virus type 1 (HIV-1)-infected individuals who maintain stable CD4+ T cell counts despite high levels of HIV-1 replication. The specific factors associated with this atypical control of the HIV infection have been poorly described. Since specific T cell responses seem to be one of the main causes of HIV-1 control in elite controllers, we studied whether HIV-1 Gag-specific cytotoxic T lymphocyte (CTL) responses could also modulate disease control in VNPs. We characterized the immune responses from four VNPs compared to those of five standard progressors (SPs) during the first years of HIV-1 infection. We observed no differences in the breadth and frequency of Gag-specific cellular responses. Furthermore, we obtained 217 HIV-1Gag clonal sequences in which the viral variability of Gag increased over 3 years of infection for synonymous and nonsynonymous mutations in both VNPs and SPs. VNPs evolution rates in gag were comparable to SPs. This observation is in line with a similar accumulation of CTL putative escape mutations in Gag epitopes targeted by CTL responses. Altogether, the absence of viral pathogenesis in VNP individuals seems to be independent of HIV-Gag-specific CTL responses. This novel information guides to the study of alternative mechanism of HIV-1 pathogenesis control. IMPORTANCE Control of HIV infection has been widely studied in elite controllers or long-term nonprogressor models. However, there is a less-known group of individuals, termed viremic nonprogressors (VNPs), who maintain stable CD4+ T cell counts despite high plasma viremia. The mechanisms involved in this remarkable control of HIV-1 pathogenesis clearly have implications for the development of new drugs and vaccines. We show here for the first time that VNPs have immune responses and HIV-gag evolution similar to those of standard progressors. Remarkably, we demonstrate that the mechanism of pathogenesis control in these individuals differs from some elite controllers that are reported to have improved immune control. This is noteworthy since it opens the door to new, as-yet-unknown mechanisms for HIV control. Our novel results advance the understanding of mechanisms involved in viremic nonprogression and suggest that there are alternative mechanisms to the adaptive immune responses for an effective control of viral pathogenesis.
Collapse
|
9
|
Tuyishime S, Haut LH, Kurupati RK, Billingsley JM, Carnathan D, Gangahara S, Styles TM, Xiang Z, Li Y, Zopfs M, Liu Q, Zhou X, Lewis MG, Amara RR, Bosinger S, Silvestri G, Ertl HCJ. Correlates of Protection Against SIV mac251 Infection in Rhesus Macaques Immunized With Chimpanzee-Derived Adenovirus Vectors. EBioMedicine 2018; 31:25-35. [PMID: 29685793 PMCID: PMC6013748 DOI: 10.1016/j.ebiom.2018.02.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/28/2018] [Accepted: 02/28/2018] [Indexed: 11/25/2022] Open
Abstract
We report on prime-boost vaccine regimens with two simian adenovirus (Ad) vectors (SAdV) or two human serotype Ad vectors (HAdV) expressing Gag and gp160 of simian immunodeficiency virus (SIV)mac239 tested in HAdV-seropositive rhesus macaques (RMs) repeatedly challenged rectally with low doses of SIVmac251. Both vaccine regimens reduced set point and peak viral loads (PVL) and accelerated viral clearance. In SAdV-vaccinated controller genotype RMs resistance against infection correlated with levels of envelope (Env)-specific antibody (Ab) titers. In both vaccine groups CD8+T cells controlled viral loads (VL) upon infection. Circulating CD4+ and CD8+ T cells showed significant changes in their transcriptome over time following vaccination, which differed between the vaccine groups. T cells from SIV-resistant RMs had unique transcriptional profiles indicating that both follicular T helper (TFH) cell responses and highly activated CD8+ T cells may play a role in protection.
Collapse
Affiliation(s)
- Steven Tuyishime
- Wistar Institute, Philadelphia, PA, United States; Gene Therapy and Vaccines Graduate Group of the University of PA, Philadelphia, PA, United States
| | | | | | - James M Billingsley
- Emory University and Yerkes National Primate Center, Atlanta, GA, United States
| | - Diane Carnathan
- Emory University and Yerkes National Primate Center, Atlanta, GA, United States
| | - Sailaja Gangahara
- Emory University and Yerkes National Primate Center, Atlanta, GA, United States
| | - Tiffany M Styles
- Emory University and Yerkes National Primate Center, Atlanta, GA, United States
| | | | - Yan Li
- Wistar Institute, Philadelphia, PA, United States
| | - Malte Zopfs
- Harvard University, Cambridge, MA, United States
| | - Qin Liu
- Wistar Institute, Philadelphia, PA, United States
| | | | | | - Rama R Amara
- Emory University and Yerkes National Primate Center, Atlanta, GA, United States
| | - Steven Bosinger
- Emory University and Yerkes National Primate Center, Atlanta, GA, United States
| | - Guido Silvestri
- Emory University and Yerkes National Primate Center, Atlanta, GA, United States
| | | |
Collapse
|
10
|
McBrien JB, Kumar NA, Silvestri G. Mechanisms of CD8 + T cell-mediated suppression of HIV/SIV replication. Eur J Immunol 2018; 48:898-914. [PMID: 29427516 DOI: 10.1002/eji.201747172] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/29/2018] [Accepted: 02/02/2018] [Indexed: 12/13/2022]
Abstract
In this article, we summarize the role of CD8+ T cells during natural and antiretroviral therapy (ART)-treated HIV and SIV infections, discuss the mechanisms responsible for their suppressive activity, and review the rationale for CD8+ T cell-based HIV cure strategies. Evidence suggests that CD8+ T cells are involved in the control of virus replication during HIV and SIV infections. During early HIV infection, the cytolytic activity of CD8+ T cells is responsible for control of viremia. However, it has been proposed that CD8+ T cells also use non-cytolytic mechanisms to control SIV infection. More recently, CD8+ T cells were shown to be required to fully suppress virus production in ART-treated SIV-infected macaques, suggesting that CD8+ T cells are involved in the control of virus transcription in latently infected cells that persist under ART. A better understanding of the complex antiviral activities of CD8+ T cells during HIV/SIV infection will pave the way for immune interventions aimed at harnessing these functions to target the HIV reservoir.
Collapse
Affiliation(s)
- Julia Bergild McBrien
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Nitasha A Kumar
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Guido Silvestri
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| |
Collapse
|
11
|
Muenchhoff M, Adland E, Karimanzira O, Crowther C, Pace M, Csala A, Leitman E, Moonsamy A, McGregor C, Hurst J, Groll A, Mori M, Sinmyee S, Thobakgale C, Tudor-Williams G, Prendergast AJ, Kloverpris H, Roider J, Leslie A, Shingadia D, Brits T, Daniels S, Frater J, Willberg CB, Walker BD, Ndung'u T, Jooste P, Moore PL, Morris L, Goulder P. Nonprogressing HIV-infected children share fundamental immunological features of nonpathogenic SIV infection. Sci Transl Med 2017; 8:358ra125. [PMID: 27683550 DOI: 10.1126/scitranslmed.aag1048] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/22/2016] [Indexed: 12/14/2022]
Abstract
Disease-free infection in HIV-infected adults is associated with human leukocyte antigen-mediated suppression of viremia, whereas in the sooty mangabey and other healthy natural hosts of simian immunodeficiency virus (SIV), viral replication continues unabated. To better understand factors preventing HIV disease, we investigated pediatric infection, where AIDS typically develops more rapidly than in adults. Among 170 nonprogressing antiretroviral therapy-naïve children aged >5 years maintaining normal-for-age CD4 T cell counts, immune activation levels were low despite high viremia (median, 26,000 copies/ml). Potent, broadly neutralizing antibody responses in most of the subjects and strong virus-specific T cell activity were present but did not drive pediatric nonprogression. However, reduced CCR5 expression and low HIV infection in long-lived central memory CD4 T cells were observed in pediatric nonprogressors. These children therefore express two cardinal immunological features of nonpathogenic SIV infection in sooty mangabeys-low immune activation despite high viremia and low CCR5 expression on long-lived central memory CD4 T cells-suggesting closer similarities with nonpathogenetic mechanisms evolved over thousands of years in natural SIV hosts than those operating in HIV-infected adults.
Collapse
Affiliation(s)
- Maximilian Muenchhoff
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. Max von Pettenkofer-Institute, Department of Virology, Ludwig-Maximilians-University Munich, Munich, Germany. German Center for Infection Research (DZIF), Partner Site Munich, Germany
| | - Emily Adland
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Owen Karimanzira
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Carol Crowther
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Matthew Pace
- Institute for Emerging Infections, Oxford Martin School, University of Oxford, Oxford, U.K. Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford OX1 3SY, U.K. Oxford National Institute of Health Research, Biomedical Research Centre, Oxford, U.K
| | - Anna Csala
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Ellen Leitman
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Angeline Moonsamy
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Callum McGregor
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Jacob Hurst
- Institute of Cancer Research, Old Brompton Road, London SW7 3RP, U.K
| | - Andreas Groll
- Department of Mathematics, Ludwig-Maximilians-University Munich, Theresienstrasse 39, 80333 Munich, Germany
| | - Masahiko Mori
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Smruti Sinmyee
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Christina Thobakgale
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | | | | | - Henrik Kloverpris
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa. Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Julia Roider
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa
| | - Alasdair Leslie
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa
| | - Delane Shingadia
- Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children, London, U.K
| | - Thea Brits
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - Samantha Daniels
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - John Frater
- Institute for Emerging Infections, Oxford Martin School, University of Oxford, Oxford, U.K. Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford OX1 3SY, U.K. Oxford National Institute of Health Research, Biomedical Research Centre, Oxford, U.K
| | - Christian B Willberg
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford OX1 3SY, U.K. Oxford National Institute of Health Research, Biomedical Research Centre, Oxford, U.K
| | - Bruce D Walker
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139-4307, USA
| | - Thumbi Ndung'u
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa. Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139-4307, USA. Max Planck Institute for Infection Biology, Berlin, Germany
| | - Pieter Jooste
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - Penny L Moore
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa. Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa. Center for the AIDS Programme of Research in South Africa (CAPRISA), 4001 Durban, South Africa
| | - Lynn Morris
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa. Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa. Center for the AIDS Programme of Research in South Africa (CAPRISA), 4001 Durban, South Africa
| | - Philip Goulder
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children, London, U.K.
| |
Collapse
|
12
|
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: 1.7] [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
|
13
|
Abstract
PURPOSE OF REVIEW The paediatric HIV epidemic is changing. Over the past decade, new infections have substantially reduced, whereas access to antiretroviral therapy (ART) has increased. Overall this success means that numbers of children living with HIV are climbing. In addition, the problems observed in adult infection resulting from chronic inflammation triggered by persistent immune activation even following ART mediated suppression of viral replication are magnified in children infected from birth. RECENT FINDINGS Features of immune ontogeny favour low immune activation in early life, whereas specific aspects of paediatric HIV infection tend to increase it. A subset of ART-naïve nonprogressing children exists in whom normal CD4 cell counts are maintained in the setting of persistent high viremia and yet in the context of low immune activation. This sooty mangabey-like phenotype contrasts with nonprogressing adult infection which is characterized by the expression of protective HLA class I molecules and low viral load. The particular factors contributing to raised or lowered immune activation in paediatric infection, which ultimately influence disease outcome, are discussed. SUMMARY Novel strategies to circumvent the unwanted long-term consequences of HIV infection may be possible in children in whom natural immune ontogeny in early life militates against immune activation. Defining the mechanisms underlying low immune activation in natural HIV infection would have applications beyond paediatric HIV.
Collapse
Affiliation(s)
- Julia M Roider
- aDepartment of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, UK bHIV Pathogenesis Programme, The Doris Duke Medical Research Institute cKwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | | |
Collapse
|
14
|
Chowdhury A, Hayes TL, Bosinger SE, Lawson BO, Vanderford T, Schmitz JE, Paiardini M, Betts M, Chahroudi A, Estes JD, Silvestri G. Differential Impact of In Vivo CD8+ T Lymphocyte Depletion in Controller versus Progressor Simian Immunodeficiency Virus-Infected Macaques. J Virol 2015; 89:8677-86. [PMID: 26063417 PMCID: PMC4524088 DOI: 10.1128/jvi.00869-15] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/03/2015] [Indexed: 02/03/2023] Open
Abstract
UNLABELLED Numerous studies have demonstrated that CD8(+) T lymphocytes suppress virus replication during human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) infection. However, the mechanisms underlying this activity of T cells remain incompletely understood. Here, we conducted CD8(+) T lymphocyte depletion in 15 rhesus macaques (RMs) infected intravenously (i.v.) with SIVmac239. At day 70 postinfection, the animals (10 progressors with high viremia and 5 controllers with low viremia) were CD8 depleted by i.v. administration of the antibody M-T807R1. As expected, CD8 depletion resulted in increased virus replication, more prominently in controllers than progressors, which correlated inversely with predepletion viremia. Of note, the feature of CD8(+) T lymphocyte predepletion that correlated best with the increase in viremia postdepletion was the level of CD8(+) T-bet(+) lymphocytes. We next found that CD8 depletion resulted in a homogenous increase of SIV RNA in superficial and mesenteric lymph nodes, spleen, and the gastrointestinal tract of both controllers and progressors. Interestingly, the level of SIV DNA increased postdepletion in both CD4(+) central memory T lymphocytes (TCM) and CD4(+) effector memory T lymphocytes (TEM) in progressor RMs but decreased in the CD4(+) TCM of 4 out of 5 controllers. Finally, we found that CD8 depletion is associated with a greater increase in CD4(+) T lymphocyte activation (measured by Ki-67 expression) in controllers than in progressors. Overall, these data reveal a differential impact of CD8(+) T lymphocyte depletion between controller and progressor SIV-infected RMs, emphasizing the complexity of the in vivo antiviral role of CD8(+) T lymphocytes. IMPORTANCE In this study, we further dissect the impact of CD8(+) T lymphocytes on HIV/SIV replication during SIV infection. CD8(+) T lymphocyte depletion leads to a relatively homogenous increase in viral replication in peripheral blood and tissues. CD8(+) T lymphocyte depletion resulted in a more prominent increase in viral loads and CD4(+) T lymphocyte activation in controllers than in progressors. Interestingly, we found T-bet expression on CD8(+) T lymphocytes to be the best predictor of viral load increase following depletion. The levels of SIV DNA increase postdepletion in both CD4(+) TCM and TEM in progressor RMs but decrease in the CD4(+) TCM of controllers. The findings described in this study provide key insights into the differential functions of CD8(+) T lymphocytes in controller and progressor RMs.
Collapse
Affiliation(s)
- Ankita Chowdhury
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Timothy L Hayes
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Steven E Bosinger
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Benton O Lawson
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Thomas Vanderford
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Joern E Schmitz
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mirko Paiardini
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Michael Betts
- Institute for Immunology and Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ann Chahroudi
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Guido Silvestri
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
15
|
Hao XP, Lucero CM, Turkbey B, Bernardo ML, Morcock DR, Deleage C, Trubey CM, Smedley J, Klatt NR, Giavedoni LD, Kristoff J, Xu A, Del Prete GQ, Keele BF, Rao SS, Alvord WG, Choyke PL, Lifson JD, Brenchley JM, Apetrei C, Pandrea I, Estes JD. Experimental colitis in SIV-uninfected rhesus macaques recapitulates important features of pathogenic SIV infection. Nat Commun 2015; 6:8020. [PMID: 26282376 PMCID: PMC4544774 DOI: 10.1038/ncomms9020] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/08/2015] [Indexed: 02/07/2023] Open
Abstract
Mucosal damage to the gastrointestinal (GI) tract with resulting microbial translocation is hypothesized to significantly contribute to the heightened and persistent chronic inflammation and immune activation characteristic to HIV infection. Here we employ a non-human primate model of chemically induced colitis in SIV-uninfected rhesus macaques that we developed using dextran sulfate sodium (DSS), to directly test this hypothesis. DSS treatment results in GI barrier damage with associated microbial translocation, inflammation and immune activation. The progression and severity of colitis are longitudinally monitored by a magnetic resonance imaging approach. DSS treatment of SIV-infected African green monkeys, a natural host species for SIV that does not manifest GI tract damage or chronic immune activation during infection, results in colitis with elevated levels of plasma SIV RNA, sCD14, LPS, CRP and mucosal CD4+ T-cell loss. Together these results support the hypothesis that GI tract damage leading to local and systemic microbial translocation, and associated immune activation, are important determinants of AIDS pathogenesis.
Collapse
Affiliation(s)
- Xing Pei Hao
- Pathology and Histotechnology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 539, Post Office Box B, Frederick, Maryland 21702, USA
| | - Carissa M. Lucero
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B69F, Bethesda, Maryland 20814, USA
| | - Marcelino L. Bernardo
- Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B69F, Bethesda, Maryland 20814, USA
| | - David R. Morcock
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Charles M. Trubey
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Jeremy Smedley
- Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 14D RM 233, 14 Service RD West, Bethesda, Maryland 20814, USA
- Washington National Primate Research Center, University of Washington, 1705 NE Pacific Street, Box 357330, Seattle, Washington 98195, USA
| | - Nichole R. Klatt
- Department of Pharmaceutics, WaNPRC, University of Washington, 3018 Western Avenue, Box 357331, Seattle, Washington 98121, USA
| | - Luis D. Giavedoni
- Department of Virology and Immunology, Southwest National Primate Research Center, Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, Texas 78227, USA
| | - Jan Kristoff
- Center for Vaccine Research, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
- School of Public Health, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
| | - Amy Xu
- Center for Vaccine Research, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
| | - Gregory Q. Del Prete
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Srinivas S. Rao
- Laboratory Animal Medicine, Vaccine Research Center, NIAID, NIH, BG 40, 40 Convent Drive, Bethesda, Maryland 20814, USA
| | - W. Gregory Alvord
- Statistical Consulting, Data Management Services, Inc., National Cancer Institute at Frederick, Post Office Box B, Frederick, Maryland 21702, USA
| | - Peter L. Choyke
- Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B69F, Bethesda, Maryland 20814, USA
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| | - Jason M. Brenchley
- Immunopathogenesis Section, Lab of Molecular Microbiology, NIAID, NIH, BG 4 RM 201, 4 Memorial Drive, Bethesda, Maryland 20814, USA
| | - Cristian Apetrei
- Center for Vaccine Research, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
| | - Ivona Pandrea
- Center for Vaccine Research, University of Pittsburgh, 9044 BST3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
- Department of Pathology and School of Medicine, University of Pittsburgh, 9017 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, Pennsylvania 15261, USA
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, BG 535, Post Office Box B, Frederick, Maryland 21702, USA
| |
Collapse
|
16
|
Smith GR, Bauer L, Crane MM, Johnson ZP. Immunogenetic characterization of a captive colony of sooty mangabeys (Cercocebus atys) used for SIV research. J Med Primatol 2015; 44:76-88. [PMID: 25645218 DOI: 10.1111/jmp.12161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2014] [Indexed: 11/27/2022]
Abstract
BACKGROUND African non-human primates are SIV natural hosts and do not develop disease following infection. Understanding disease avoidance mechanisms in these species is important for HIV vaccine development. The largest captive population of sooty mangabeys, a SIV natural host species, resides at the Yerkes National Primate Research Center. METHODS Thirteen primer sets that amplify polymorphic microsatellite loci within the MHC region were used to genotype 144 animals. Immunogenetic Management Software (IMS) was used to identify MHC haplotypes and organize data. RESULTS Seventy-three haplotypes were identified. Limited haplotype diversity was observed in this population with 88.2% of included animals carrying one of 18 haplotypes. Differences in haplotype frequency were observed between SIV (+) and SIV (-) populations. CONCLUSIONS We have developed a novel tool for others to use in the analysis of the role of the MHC in a natural host non-human primate model species used for SIV research.
Collapse
Affiliation(s)
- Geary R Smith
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | | | | | | |
Collapse
|
17
|
Xu H, Wang X, Veazey RS. Simian Immunodeficiency Virus Infection and Mucosal Immunity. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00076-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
18
|
Chahroudi A, Cartwright E, Lee ST, Mavigner M, Carnathan DG, Lawson B, Carnathan PM, Hashempoor T, Murphy MK, Meeker T, Ehnert S, Souder C, Else JG, Cohen J, Collman RG, Vanderford TH, Permar SR, Derdeyn CA, Villinger F, Silvestri G. Target cell availability, rather than breast milk factors, dictates mother-to-infant transmission of SIV in sooty mangabeys and rhesus macaques. PLoS Pathog 2014; 10:e1003958. [PMID: 24604066 PMCID: PMC3946396 DOI: 10.1371/journal.ppat.1003958] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/05/2014] [Indexed: 12/15/2022] Open
Abstract
Mother-to-infant transmission (MTIT) of HIV is a serious global health concern, with over 300,000 children newly infected in 2011. SIV infection of rhesus macaques (RMs) results in similar rates of MTIT to that of HIV in humans. In contrast, SIV infection of sooty mangabeys (SMs) rarely results in MTIT. The mechanisms underlying protection from MTIT in SMs are unknown. In this study we tested the hypotheses that breast milk factors and/or target cell availability dictate the rate of MTIT in RMs (transmitters) and SMs (non-transmitters). We measured viral loads (cell-free and cell-associated), levels of immune mediators, and the ability to inhibit SIV infection in vitro in milk obtained from lactating RMs and SMs. In addition, we assessed the levels of target cells (CD4+CCR5+ T cells) in gastrointestinal and lymphoid tissues, including those relevant to breastfeeding transmission, as well as peripheral blood from uninfected RM and SM infants. We found that frequently-transmitting RMs did not have higher levels of cell-free or cell-associated viral loads in milk compared to rarely-transmitting SMs. Milk from both RMs and SMs moderately inhibited in vitro SIV infection, and presence of the examined immune mediators in these two species did not readily explain the differential rates of transmission. Importantly, we found that the percentage of CD4+CCR5+ T cells was significantly lower in all tissues in infant SMs as compared to infant RMs despite robust levels of CD4+ T cell proliferation in both species. The difference between the frequently-transmitting RMs and rarely-transmitting SMs was most pronounced in CD4+ memory T cells in the spleen, jejunum, and colon as well as in central and effector memory CD4+ T cells in the peripheral blood. We propose that limited availability of SIV target cells in infant SMs represents a key evolutionary adaptation to reduce the risk of MTIT in SIV-infected SMs. Currently 2.5 million children are infected with HIV, largely as a result of mother-to-child transmission, and there is no effective vaccine or cure. Studies of Simian Immunodeficiency Virus (SIV) infection of nonhuman primate species termed “natural hosts” have shown that mother-to-infant transmission of SIV in these animals is rare. Natural hosts are African monkey species that are naturally infected with SIV in the wild but do not develop AIDS. We sought to understand the mechanism by which natural hosts are protected from mother-to-infant transmission of SIV, aiming to translate our findings into novel strategies to prevent perinatal HIV infection. We found that natural host sooty mangabey infants have extremely low levels of target cells for SIV infection in lymphoid and gastrointestinal tissues. Direct comparison of infant sooty mangabeys and infant rhesus macaques (non-natural host species with high SIV transmission rates) confirmed that natural hosts have significantly lower levels of SIV target cells compared with non-natural hosts. Analysis of the breast milk of sooty mangabeys and rhesus macaques revealed similar levels of virus and ability to inhibit SIV infection. Our study provides evidence for target cell restriction as the main mechanism of protection from mother-to-infant SIV transmission in natural hosts.
Collapse
Affiliation(s)
- Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- * E-mail:
| | - Emily Cartwright
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - S. Thera Lee
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Maud Mavigner
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Diane G. Carnathan
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Benton Lawson
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Paul M. Carnathan
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Tayebeh Hashempoor
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Megan K. Murphy
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Tracy Meeker
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Stephanie Ehnert
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Christopher Souder
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - James G. Else
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- Departments of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Joyce Cohen
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Ronald G. Collman
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Thomas H. Vanderford
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - Cynthia A. Derdeyn
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- Departments of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Francois Villinger
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- Departments of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Guido Silvestri
- Yerkes National Primate Research Center, Atlanta, Georgia, United States of America
- Departments of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
19
|
Increased stability and limited proliferation of CD4+ central memory T cells differentiate nonprogressive simian immunodeficiency virus (SIV) infection of sooty mangabeys from progressive SIV infection of rhesus macaques. J Virol 2014; 88:4533-42. [PMID: 24501416 DOI: 10.1128/jvi.03515-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Depletion of CD4(+) central memory T (TCM) cells dictates the tempo of progression to AIDS in simian immunodeficiency virus (SIV)-infected rhesus macaques (RMs) both in the natural history of infection and in the context of vaccination. CD4(+) TCM cells of sooty mangabeys (SMs), a natural host for SIV in which infection is nonpathogenic, are less susceptible to SIV infection than CD4(+) TCM cells of RMs. Whether this relative protection from infection translates into increased stability of CD4(+) TCM cells in natural versus nonnatural hosts has not yet been determined. Here we compared, both cross-sectionally and longitudinally, the levels of CD4(+) TCM cells in a large cohort of SMs and RMs and the association between CD4(+) TCM levels and the main virologic and immunologic markers of disease progression. Consistent with their lower susceptibility to infection, CD4(+) TCM cells of SIV-infected SMs are lost with kinetics 20 times slower than those of SIV-infected RMs. Remarkably, the estimated length of time of SIV infection needed for CD4(+) TCM cells to fall to half of their initial levels is <16 months for RMs but >17 years for SMs. Furthermore, the fraction of proliferating CD4(+) TCM cells is significantly lower in SIV-infected SMs than in SIV-infected RMs, and the extent of CD4(+) TCM cell proliferation is associated positively with CD4(+) T cell levels in SIV-infected SMs but negatively with CD4(+) T cell levels in SIV-infected RMs. Collectively, these findings identify increased stability and maintenance of the prohomeostatic role of CD4(+) TCM cells as features distinguishing nonprogressive from progressive SIV infections and support the hypothesis of a direct mechanistic link between the loss of CD4(+) TCM cells and disease progression. IMPORTANCE Comparison of the immunologic effects of simian immunodeficiency virus (SIV) infection on rhesus macaques (RMs), a species characterized by progression to AIDS, and natural host sooty mangabeys (SMs), a species which remains AIDS free, has become a useful tool for identifying mechanisms of human immunodeficiency virus (HIV) disease progression. One such distinguishing feature is that CD4(+) central memory T (TCM) cells in SIV-infected SMs are less infected than the same cells in RMs. Here we investigated whether lower levels of infection in SMs translate into a better-preserved CD4(+) TCM compartment. We found that the CD4(+) TCM compartment is significantly more stable in SIV-infected SMs. Likely to compensate for this cell loss, we also found that CD4(+) TCM cells increase their level of proliferation upon SIV infection in RMs but not in SMs, which mechanistically supports their preferential infectivity. Our study provides new insights into the importance of long-term maintenance of CD4(+) TCM homeostasis during HIV/SIV infection.
Collapse
|
20
|
Cook R, Leroux C, Issel C. Equine infectious anemia and equine infectious anemia virus in 2013: A review. Vet Microbiol 2013; 167:181-204. [DOI: 10.1016/j.vetmic.2013.09.031] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 09/16/2013] [Accepted: 09/21/2013] [Indexed: 10/26/2022]
|
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: 4.6] [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: 50] [Impact Index Per Article: 4.5] [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
|
Shaw JM, Hunt PW, Critchfield JW, McConnell DH, Garcia JC, Pollard RB, Somsouk M, Deeks SG, Shacklett BL. Short communication: HIV+ viremic slow progressors maintain low regulatory T cell numbers in rectal mucosa but exhibit high T cell activation. AIDS Res Hum Retroviruses 2013; 29:172-7. [PMID: 22998457 DOI: 10.1089/aid.2012.0268] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Viremic slow progressors (VSP) are a rare subset of HIV-infected persons who exhibit slow immunologic progression despite high viremia. The mechanisms associated with this slow progression remain to be defined. Clinical characteristics of VSP are similar to those of natural hosts for simian immunodeficiency virus (SIV), such as sooty mangabeys (SM) and African green monkeys (AGM), who maintain near-normal CD4 counts despite high-level viremia but maintain low immune activation. Immune activation is a powerful predictor of disease progression, and we hypothesized that low immune activation might also explain the VSP phenotype. Using multiparameter flow cytometry, we assessed levels of T cell activation and regulatory T cells (Treg) in blood and rectal mucosa of VSP, typical progressors, virologic controllers, and seronegative controls. We also assessed Treg function and CD4 T cell proliferative capacity in VSP. Contrary to expectations, we found that VSP subjects have high levels of T cell activation in the gastrointestinal mucosa. The ratio of Treg to CD3+ T cells in the mucosa of VSP was relatively low, potentially contributing to increased immune activation. Nonetheless, CD4+CD25- T cells isolated from these individuals displayed a comparatively weak proliferative response to anti-CD3 stimulation. These data reveal that the VSP phenotype is associated with elevated markers of mucosal immune activation and low numbers of mucosal Treg, suggesting that factors other than immune activation account for this phenotype.
Collapse
Affiliation(s)
- Julia M. Shaw
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California
| | - Peter W. Hunt
- Positive Health Program, Department of Medicine, University of California, San Francisco, California
| | - J. William Critchfield
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California
| | - Delandy H. McConnell
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California
| | - Juan Carlos Garcia
- Division of Gastroenterology, School of Medicine, University of California, Davis, California
| | - Richard B. Pollard
- Division of Infectious Diseases, School of Medicine, University of California, Davis, California
| | - Ma Somsouk
- Division of Gastroenterology, San Francisco General Hospital, University of California, San Francisco, California
| | - Steven G. Deeks
- Positive Health Program, Department of Medicine, University of California, San Francisco, California
| | - Barbara L. Shacklett
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California
- Division of Infectious Diseases, School of Medicine, University of California, Davis, California
| |
Collapse
|
24
|
|
25
|
Brenchley JM, Vinton C, Tabb B, Hao XP, Connick E, Paiardini M, Lifson JD, Silvestri G, Estes JD. Differential infection patterns of CD4+ T cells and lymphoid tissue viral burden distinguish progressive and nonprogressive lentiviral infections. Blood 2012; 120:4172-81. [PMID: 22990012 PMCID: PMC3501715 DOI: 10.1182/blood-2012-06-437608] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 08/31/2012] [Indexed: 01/14/2023] Open
Abstract
Nonhuman primate natural hosts for simian immunodeficiency viruses (SIV) develop a nonresolving chronic infection but do not develop AIDS. Mechanisms to explain the nonprogressive nature of SIV infection in natural hosts that underlie maintained high levels of plasma viremia without apparent loss of target cells remain unclear. Here we used comprehensive approaches (ie, FACS sorting, quantitative RT-PCR, immunohistochemistry, and in situ hybridization) to study viral infection within subsets of peripheral blood and lymphoid tissue (LT) CD4(+) T cells in cohorts of chronically SIV-infected rhesus macaques (RMs), HIV-infected humans, and SIVsmm-infected sooty mangabeys (SMs). We find: (1) infection frequencies among CD4(+) T cells in chronically SIV-infected RMs are significantly higher than those in SIVsmm-infected SMs; (2) infected cells are found in distinct anatomic LT niches and different CD4(+) T-cell subsets in SIV-infected RMs and SMs, with infection patterns of RMs reflecting HIV infection in humans; (3) T(FH) cells are infected at higher frequencies in RMs and humans than in SMs; and (4) LT viral burden, including follicular dendritic cell deposition of virus, is increased in RMs and humans compared with SMs. These data provide insights into how natural hosts are able to maintain high levels of plasma viremia while avoiding development of immunodeficiency.
Collapse
Affiliation(s)
- Jason M Brenchley
- Program in Barrier Immunity and Repair, National Institute of Allergy and Infectious Diseases, National Institutes ofHealth, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Micci L, Cervasi B, Ende ZS, Iriele RI, Reyes-Aviles E, Vinton C, Else J, Silvestri G, Ansari AA, Villinger F, Pahwa S, Estes JD, Brenchley JM, Paiardini M. Paucity of IL-21-producing CD4(+) T cells is associated with Th17 cell depletion in SIV infection of rhesus macaques. Blood 2012; 120:3925-35. [PMID: 22990011 PMCID: PMC3496953 DOI: 10.1182/blood-2012-04-420240] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 08/31/2012] [Indexed: 12/21/2022] Open
Abstract
IL-21 regulates Th17 cell homeostasis, enhances the differentiation of memory B cells and antibody-secreting plasma cells, and promotes the maintenance of CD8(+) T-cell responses. In this study, we investigated the phenotype, function, and frequency of blood and intestinal IL-21-producing cells in nonhuman primates that are hosts of progressive (rhesus macaques [RMs]) and nonprogressive (sooty mangabeys [SMs]) SIV infection. We found that, in both species, memory CD4(+)CD95(+)CCR6(-) T cells are the main IL-21 producers, and that only a small fraction of CD4(+)IL-21(+) T cells produce IL-17. During chronic SIV infection of RMs, CD4(+)IL-21(+) T cells were significantly depleted in both blood and rectal mucosa, with the extent of this depletion correlating with the loss of Th17 cells. Furthermore, treatment with IL-21 increased the in vivo levels of Th17 cells in SIV-infected RMs. In contrast, normal levels of CD4(+)IL-21(+) T cells were found in SIV-infected SMs. Collectively, these data indicate that depletion of IL-21-producing CD4(+) T cells distinguishes progressive from nonprogressive SIV infection of RMs and SMs, and suggest that depletion of CD4(+)IL-21(+) T cells is involved in the preferential loss of Th17 cells that is associated with SIV disease progression. Further preclinical studies of IL-21 as a potential immunotherapeutic agent for HIV infection may be warranted.
Collapse
Affiliation(s)
- Luca Micci
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Distinct evolutionary pressures underlie diversity in simian immunodeficiency virus and human immunodeficiency virus lineages. J Virol 2012; 86:13217-31. [PMID: 23055550 DOI: 10.1128/jvi.01862-12] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Simian immunodeficiency virus (SIV) infection of rhesus macaques causes immune depletion and disease closely resembling human AIDS and is well recognized as the most relevant animal model for the human disease. Experimental investigations of viral pathogenesis and vaccine protection primarily involve a limited set of related viruses originating in sooty mangabeys (SIVsmm). The diversity of human immunodeficiency virus type 1 (HIV-1) has evolved in humans in about a century; in contrast, SIV isolates used in the macaque model evolved in sooty mangabeys over millennia. To investigate the possible consequences of such different evolutionary histories for selection pressures and observed diversity in SIVsmm and HIV-1, we isolated, sequenced, and analyzed 20 independent isolates of SIVsmm, including representatives of 7 distinct clades of viruses isolated from natural infection. We found SIVsmm diversity to be lower overall than HIV-1 M group diversity. Reduced positive selection (i.e., less diversifying evolution) was evident in extended regions of SIVsmm proteins, most notably in Gag p27 and Env gp120. In addition, the relative diversities of proteins in the two lineages were distinct: SIVsmm Env and Gag were much less diverse than their HIV-1 counterparts. This may be explained by lower SIV-directed immune activity in mangabeys relative to HIV-1-directed immunity in humans. These findings add an additional layer of complexity to the interpretation and, potentially, to the predictive utility of the SIV/macaque model, and they highlight the unique features of human and simian lentiviral evolution that inform studies of pathogenesis and strategies for AIDS vaccine design.
Collapse
|
28
|
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: 201] [Impact Index Per Article: 16.8] [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
|
29
|
Garg H, Viard M, Jacobs A, Blumenthal R. Targeting HIV-1 gp41-induced fusion and pathogenesis for anti-viral therapy. Curr Top Med Chem 2012; 11:2947-58. [PMID: 22044225 DOI: 10.2174/156802611798808479] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 02/28/2011] [Accepted: 12/01/2011] [Indexed: 11/22/2022]
Abstract
HIV gp41 is a metastable protein whose native conformation is maintained in the form of a heterodimer with gp120. The non-covalently associated gp41/gp120 complex forms a trimer on the virus surface. As gp120 engages with HIV's receptor, CD4, and coreceptor, CXCR4 or CCR5, gp41 undergoes several conformational changes resulting in fusion between the viral and cellular membranes. Several lipophilic and amphiphilic domains have been shown to be critical in that process. While the obvious function of gp41 in viral entry is well-established its role in cellular membrane fusion and the link with pathogenesis are only now beginning to appear. Recent targeting of gp41 via fusion inhibitors has revealed an important role of this protein not only in viral entry but also in bystander apoptosis and HIV pathogenesis. Studies by our group and others have shown that the phenomenon of gp41-mediated hemifusion initiates apoptosis in bystander cells and correlates with virus pathogenesis. More interestingly, recent clinical evidence suggests that gp41 mutants arising after Enfuvirtide therapy are associated with CD4 cell increase and immunological benefits. This has in turn been correlated to a decrease in bystander apoptosis in our in vitro as well as in vivo assays. Although a great deal of work has been done to unravel HIV-1 gp41-mediated fusion mechanisms, the factors that regulate gp41-mediated fusion versus hemifusion and the mechanism by which hemifusion initiates bystander apoptosis are not fully understood. Further insight into these issues will open new avenues for drug development making gp41 a critical anti-HIV target both for neutralization and virus attenuation.
Collapse
Affiliation(s)
- Himanshu Garg
- Center of Excellence for Infectious Disease, Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | | | | | | |
Collapse
|
30
|
Treatment of SIV-infected sooty mangabeys with a type-I IFN agonist results in decreased virus replication without inducing hyperimmune activation. Blood 2012; 119:5750-7. [PMID: 22550346 DOI: 10.1182/blood-2012-02-411496] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A key feature differentiating nonpathogenic SIV infection of sooty mangabeys (SMs) from pathogenic HIV/SIV infections is the rapid resolution of type I IFN (IFN-I) responses and IFN-stimulated gene expression during the acute-to-chronic phase transition and the establishment of an immune quiescent state that persists throughout the chronic infection. We hypothesized that low levels of IFN-I signaling may help to prevent chronic immune activation and disease progression in SIV-infected SMs. To assess the effects of IFN-I signaling in this setting, in the present study, we administered recombinant rhesus macaque IFNα2-IgFc (rmIFNα2) to 8 naturally SIV-infected SMs weekly for 16 weeks. Gene-expression profiling revealed a strong up-regulation of IFN-stimulated genes in the blood of treated animals, confirming the reagent's bioactivity. Interestingly, we observed an approximately 1-log decrease in viral load that persisted through day 35 of treatment. Flow cytometric analysis of lymphocytes in the blood, lymph nodes, and rectal biopsies did not reveal a significant decline of CD4(+) T cells, a robust increase in lymphocyte activation, or change in the level of SIV-specific CD8(+) T cells. The results of the present study indicate that administration of type I IFNs in SIV-infected SMs induces a significant anti-viral effect that is not associated with a detectable increase in chronic immune activation.
Collapse
|
31
|
Sundaravaradan V, Mir KD, Sodora DL. Double-negative T cells during HIV/SIV infections: potential pinch hitters in the T-cell lineup. Curr Opin HIV AIDS 2012; 7:164-71. [PMID: 22241163 PMCID: PMC3639317 DOI: 10.1097/coh.0b013e3283504a66] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW This review summarizes the role of CD3+CD4-CD8- double-negative T cells, which have both regulatory and helper T-cell functions and may have the potential to compensate for the reduced levels of CD4 T cells during SIV/HIV infection. RECENT FINDINGS Double-negative T cells have been characterized in several human diseases and in murine models of autoimmunity and transplantation, where they exhibit both immunoregulatory and helper T-cell-like function. During the natural nonpathogenic SIV infection of African nonhuman primates, the lack of clinical disease progression is associated with the presence of double-negative T cells that maintain helper T-cell functions while remaining refractory to viral infection. Moreover, DN T cells may compensate for very low levels of CD4+ T cells observed in a cohort of SIV-infected sooty mangabeys that have remained free of clinical AIDS for over 10 years. These studies identify a potential for double-negative T cells to provide critical helper function during HIV infection. SUMMARY Double-negative T cells with some CD4+ T-cell functions are associated with a nonpathogenic outcome during SIV infection and represent a potential immune therapeutic target in HIV-infected patients.
Collapse
|
32
|
Mucosal simian immunodeficiency virus transmission in African green monkeys: susceptibility to infection is proportional to target cell availability at mucosal sites. J Virol 2012; 86:4158-68. [PMID: 22318138 DOI: 10.1128/jvi.07141-11] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
African green monkeys (AGMs) are naturally infected with a simian immunodeficiency virus (SIVagm) that is nonpathogenic in its host. Although SIVagm is common and widespread, little is known about the mechanisms that govern its transmission. Since the earliest virus-host interactions may provide key insights into the nonpathogenic phenotype of SIVagm, we developed a mucosal transmission model for this virus. Using plasma from an acutely infected AGM as the virus inoculum, we exposed adult and juvenile AGMs, as well as pigtailed macaques (PTMs) as a nonnatural host control, by mucosal routes to increasing titers of virus and compared the doses needed to establish a productive infection. Four juvenile and four adult AGMs as well as two PTMs were intrarectally (IR) exposed, while two additional adult female AGMs were intravaginally (IVAG) exposed. No animal became infected following exposure to 10(5) RNA copies. Both PTMs but none of the AGMs became infected following exposure to 10(6) RNA copies. Finally, all adult AGMs and two of the four juvenile AGMs became infected following exposure to 10(7) RNA copies, acquiring either one (2 IR infected juveniles, 1 IR infected adult, 2 IVAG infected adults) or two (3 IR infected adults) transmitted founder viruses. These results were consistent with immunophenotypic data, which revealed a significant correlation between the percentage of CD4(+) T cells expressing CCR5 in the mucosa and the susceptibility to infection, in terms of both the viral dose and the numbers of transmitted founder viruses. Moreover, studies of uninfected AGMs showed that the fraction of CCR5-expressing CD4(+) T cells increased significantly with age. These results indicate that (i) AGMs are readily infected with SIVagm by both intrarectal and intravaginal routes, (ii) susceptibility to infection is proportional to the number of available CCR5(+) CD4(+) target cells in the mucosa, and (iii) the paucity of CCR5(+) CD4(+) target cells in infant and juvenile AGMs may explain the near absence of vertical transmission.
Collapse
|
33
|
Ortiz AM, Klatt NR, Li B, Yi Y, Tabb B, Hao XP, Sternberg L, Lawson B, Carnathan PM, Cramer EM, Engram JC, Little DM, Ryzhova E, Gonzalez-Scarano F, Paiardini M, Ansari AA, Ratcliffe S, Else JG, Brenchley JM, Collman RG, Estes JD, Derdeyn CA, Silvestri G. Depletion of CD4⁺ T cells abrogates post-peak decline of viremia in SIV-infected rhesus macaques. J Clin Invest 2011; 121:4433-45. [PMID: 22005304 PMCID: PMC3204830 DOI: 10.1172/jci46023] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 09/07/2011] [Indexed: 12/13/2022] Open
Abstract
CD4+ T cells play a central role in the immunopathogenesis of HIV/AIDS, and their depletion during chronic HIV infection is a hallmark of disease progression. However, the relative contribution of CD4+ T cells as mediators of antiviral immune responses and targets for virus replication is still unclear. Here, we have generated data in SIV-infected rhesus macaques (RMs) that suggest that CD4+ T cells are essential in establishing control of virus replication during acute infection. To directly assess the role of CD4+ T cells during primary SIV infection, we in vivo depleted these cells from RMs prior to infecting the primates with a pathogenic strain of SIV. Compared with undepleted animals, CD4+ lymphocyte-depleted RMs showed a similar peak of viremia, but did not manifest any post-peak decline of virus replication despite CD8+ T cell- and B cell-mediated SIV-specific immune responses comparable to those observed in control animals. Interestingly, depleted animals displayed rapid disease progression, which was associated with increased virus replication in non-T cells as well as the emergence of CD4-independent SIV-envelopes. Our results suggest that the antiviral CD4+ T cell response may play an important role in limiting SIV replication, which has implications for the design of HIV vaccines.
Collapse
Affiliation(s)
- Alexandra M. Ortiz
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nichole R. Klatt
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Bing Li
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yanjie Yi
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Brian Tabb
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Xing Pei Hao
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lawrence Sternberg
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Benton Lawson
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Paul M. Carnathan
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Elizabeth M. Cramer
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jessica C. Engram
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dawn M. Little
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Elena Ryzhova
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Francisco Gonzalez-Scarano
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Mirko Paiardini
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Aftab A. Ansari
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Sarah Ratcliffe
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - James G. Else
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jason M. Brenchley
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ronald G. Collman
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jacob D. Estes
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Cynthia A. Derdeyn
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center and Emory Vaccine Center, Emory University, Atlanta, Georgia, USA.
Department of Microbiology and
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Laboratory of Molecular Microbiology, NIH, Bethesda, Maryland, USA.
Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
AIDS and Cancer Virus Program, SAIC-Frederick, National Cancer Institute, NIH, Fredrick, Maryland, USA.
Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA.
Department of Neurology and
Department of Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
34
|
Immunovirological analyses of chronically simian immunodeficiency virus SIVmnd-1- and SIVmnd-2-infected mandrills (Mandrillus sphinx). J Virol 2011; 85:13077-87. [PMID: 21957286 DOI: 10.1128/jvi.05693-11] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Simian immunodeficiency virus (SIV) infection in African nonhuman primate (NHP) natural hosts is usually nonpathogenic, despite high levels of virus replication. We have previously shown that chronic SIV infection in sooty mangabeys (SMs) and African green monkeys (AGMs) is associated with low levels of immune activation and bystander T cell apoptosis. To compare these features with those observed in another natural host, the mandrill (MND), we conducted a cross-sectional survey of the 23 SIV-infected and 25 uninfected MNDs from the only semifree colony of mandrills available worldwide. Viral loads (VLs) were determined and phenotypic and functional analysis of peripheral blood- and lymph node-derived lymphocytes was performed. We found that mandrills chronically infected with SIVmnd-1 or SIVmnd-2 have similar levels of viral replication, and we observed a trend toward lower CD4+ T cell counts in chronically SIVmnd-2-infected MNDs than SIVmnd-1-infected MNDs. No correlation between CD4+ T cell counts and VLs in SIV-infected MNDs could be established. Of note, the levels of T cell activation, proliferation, and apoptosis were comparable between SIVmnd-1- and SIVmnd-2-infected MNDs and to those observed in uninfected animals, with the only exception being an increase in tumor necrosis factor alpha-producing CD8+ T cells in SIVmnd-2-infected MNDs. Overall, these findings recapitulate previous observations in SIV-infected SMs and AGMs and lend further evidence to the hypothesis that low levels of immune activation protect natural SIV hosts from disease progression.
Collapse
|
35
|
Mother-to-infant transmission of simian immunodeficiency virus is rare in sooty mangabeys and is associated with low viremia. J Virol 2011; 85:5757-63. [PMID: 21450815 DOI: 10.1128/jvi.02690-10] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mother-to-child transmission of human immunodeficiency virus type 1 (HIV-1) occurs in utero, intrapartum, and through breastfeeding, with a cumulative rate of transmission of 35 to 40%. As a result, ∼ 400,000 children become infected each year. Little is known about mother-to-infant transmission (MTIT) during natural simian immunodeficiency virus (SIV) infection of sooty mangabeys (SMs) that typically is nonpathogenic despite high viral loads. In this study, we retrospectively investigated the rates of MTIT in a large colony of naturally SIV-infected SMs using serological (anti-SIV antibody by enzyme-linked immunosorbent assay [ELISA] and Western blot analysis) and virological (SIV(smm) real-time reverse transcription-PCR) methods. We examined 161 SM infants born to SIV-infected mothers and found that 150 (93.2%) were infected by non-MTIT (n = 120) or remained uninfected (n = 30). The remaining 11 SM infants (6.8%) were defined as acquiring SIV by presumptive MTIT based on (i) the presence of anti-SIV antibodies without seroreversion and (ii) a viral load of >500 copies/ml of serum in the first year of life. SM infants infected with SIV by presumptive MTIT did not show any increased morbidity or mortality, indicating that the infection is nonpathogenic even when acquired early in life. Interestingly, viral loads of SIV-infected SM infants with presumptive MTIT were 2-log lower than those of SIV-infected adult SMs living in the same colony (i.e., ∼ 1,000 and 100,000 copies/ml, respectively). These results indicate that MTIT is substantially less frequent in naturally SIV-infected SMs than in HIV-1-infected humans and results in nonpathogenic infection associated with low SIV viremia. Evolutionary pressure to reduce MTIT may have contributed to the restriction of SIV pathogenesis in natural hosts.
Collapse
|
36
|
Meythaler M, Wang Z, Martinot A, Pryputniewicz S, Kasheta M, McClure HM, O'Neil SP, Kaur A. Early induction of polyfunctional simian immunodeficiency virus (SIV)-specific T lymphocytes and rapid disappearance of SIV from lymph nodes of sooty mangabeys during primary infection. THE JOURNAL OF IMMUNOLOGY 2011; 186:5151-61. [PMID: 21441446 DOI: 10.4049/jimmunol.1004110] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Although the cellular immune response is essential for controlling SIV replication in Asian macaques, its role in maintaining nonpathogenic SIV infection in natural hosts such as sooty mangabeys (SM) remains to be defined. We have previously shown that similar to rhesus macaques (RM), SM are able to mount a T lymphocyte response against SIV infection. To investigate early control of SIV replication in natural hosts, we performed a detailed characterization of SIV-specific cellular immunity and viral control in the first 6 mo following SIV infection in SM. Detection of the initial SIV-specific IFN-γ ELISPOT response in SIVsmE041-infected SM coincided temporally with a decline in peak plasma viremia and was similar in magnitude, specificity, and breadth to SIVsmE041-infected and SIVmac239-infected RM. Despite these similarities, SM showed a greater reduction in postpeak plasma viremia and a more rapid disappearance of productively SIV-infected cells from the lymph node compared with SIVmac239-infected RM. The early Gag-specific CD8(+) T lymphocyte response was significantly more polyfunctional in SM compared with RM, and granzyme B-positive CD8(+) T lymphocytes were present at significantly higher frequencies in SM even prior to SIV infection. These findings suggest that the early SIV-specific T cell response may be an important determinant of lymphoid tissue viral clearance and absence of lymph node immunopathology in natural hosts of SIV infection.
Collapse
Affiliation(s)
- Mareike Meythaler
- Department of Immunology, New England Primate Research Center, Harvard Medical School, Southborough, MA 01772, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Milush JM, Mir KD, Sundaravaradan V, Gordon SN, Engram J, Cano CA, Reeves JD, Anton E, O'Neill E, Butler E, Hancock K, Cole KS, Brenchley JM, Else JG, Silvestri G, Sodora DL. Lack of clinical AIDS in SIV-infected sooty mangabeys with significant CD4+ T cell loss is associated with double-negative T cells. J Clin Invest 2011; 121:1102-10. [PMID: 21317533 DOI: 10.1172/jci44876] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 12/15/2010] [Indexed: 11/17/2022] Open
Abstract
SIV infection of natural host species such as sooty mangabeys results in high viral replication without clinical signs of simian AIDS. Studying such infections is useful for identifying immunologic parameters that lead to AIDS in HIV-infected patients. Here we have demonstrated that acute, SIV-induced CD4(+) T cell depletion in sooty mangabeys does not result in immune dysfunction and progression to simian AIDS and that a population of CD3(+)CD4(-)CD8(-) T cells (double-negative T cells) partially compensates for CD4(+) T cell function in these animals. Passaging plasma from an SIV-infected sooty mangabey with very few CD4(+) T cells to SIV-negative animals resulted in rapid loss of CD4(+) T cells. Nonetheless, all sooty mangabeys generated SIV-specific antibody and T cell responses and maintained normal levels of plasma lipopolysaccharide. Moreover, all CD4-low sooty mangabeys elicited a de novo immune response following influenza vaccination. Such preserved immune responses as well as the low levels of immune activation observed in these animals were associated with the presence of double-negative T cells capable of producing Th1, Th2, and Th17 cytokines. These studies indicate that SIV-infected sooty mangabeys do not appear to rely entirely on CD4(+) T cells to maintain immunity and identify double-negative T cells as a potential subset of cells capable of performing CD4(+) T cell-like helper functions upon SIV-induced CD4(+) T cell depletion in this species.
Collapse
Affiliation(s)
- Jeffrey M Milush
- Division of Infectious Disease, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kobayashi Y. Molecular Target Therapy in Hematological Malignancy: Front-runners and Prototypes of Small Molecule and Antibody Therapy. Jpn J Clin Oncol 2010; 41:157-64. [DOI: 10.1093/jjco/hyq189] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
39
|
Mir KD, Gasper MA, Sundaravaradan V, Sodora DL. SIV infection in natural hosts: resolution of immune activation during the acute-to-chronic transition phase. Microbes Infect 2010; 13:14-24. [PMID: 20951225 DOI: 10.1016/j.micinf.2010.09.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 09/09/2010] [Indexed: 10/18/2022]
Abstract
SIV-infected natural hosts do not progress to clinical AIDS yet display high viral replication and an acute immunologic response similar to pathogenic SIV/HIV infections. During chronic SIV infection, natural hosts suppress their immune activation, whereas pathogenic hosts display a highly activated immune state. Here, we review natural host SIV infections with an emphasis on specific immune cells and their contribution to the transition from the acute-to-chronic phases of infection.
Collapse
Affiliation(s)
- Kiran D Mir
- Seattle Biomedical Research Institute, Seattle, WA, USA
| | | | | | | |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW To summarize our current understanding of the regulation of Th17 cells in pathogenic and nonpathogenic lentiviral infections. RECENT FINDINGS It has been shown that Th17 cells, a recently identified T helper-cell subset deemed critical for antimicrobial mucosal immunity, are preferentially depleted in the gastrointestinal tracts of human immunodeficiency virus (HIV)-infected humans and simian immunodeficiency virus (SIV)-infected rhesus and pigtailed (PTMs) macaques. In contrast, Th17 cells are preserved at healthy levels in monkey species that are natural hosts for SIV, such as sooty mangabeys or African green monkeys (AGMs), which maintain mucosal immunity and remain AIDS free. These findings suggest that preservation of Th17 cells (or lack thereof) may be central in determining the pathogenic or nonpathogenic outcome of HIV/SIV infection. SUMMARY A preferential depletion of mucosal Th17 cells is a feature that distinguishes pathogenic HIV infection of humans from nonprogressive SIV infection of sooty mangabeys and AGMs. The exact mechanism accounting for this different phenotype is still unclear. To understand how natural hosts for SIV preserve Th17 cells and mucosal immunity might be central to the development of therapeutic interventions aimed at improving mucosal immunity in HIV-infected individuals.
Collapse
|
41
|
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: 94] [Impact Index Per Article: 6.7] [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
|
42
|
Cafaro A, Macchia I, Maggiorella MT, Titti F, Ensoli B. Innovative approaches to develop prophylactic and therapeutic vaccines against HIV/AIDS. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 655:189-242. [PMID: 20047043 DOI: 10.1007/978-1-4419-1132-2_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The acquired immunodeficiency syndrome (AIDS) emerged in the human population in the summer of 1981. According to the latest United Nations estimates, worldwide over 33 million people are infected with human immunodeficiency virus (HIV) and the prevalence rates continue to rise globally. To control the alarming spread of HIV, an urgent need exists for developing a safe and effective vaccine that prevents individuals from becoming infected or progressing to disease. To be effective, an HIV/AIDS vaccine should induce broad and long-lasting humoral and cellular immune responses, at both mucosal and systemic level. However, the nature of protective immune responses remains largely elusive and this represents one of the major roadblocks preventing the development of an effective vaccine. Here we summarize our present understanding of the factors responsible for resistance to infection or control of progression to disease in human and monkey that may be relevant to vaccine development and briefly review recent approaches which are currently being tested in clinical trials. Finally, the rationale and the current status of novel strategies based on nonstructural HIV-1 proteins, such as Tat, Nef and Rev, used alone or in combination with modified structural HIV-1 Env proteins are discussed.
Collapse
Affiliation(s)
- Aurelio Cafaro
- National AIDS Center, Istituto Superiore di Sanità, V.le Regina Elena 299, 00161, Rome, Italy
| | | | | | | | | |
Collapse
|
43
|
Sikder MA, Friedberg JW. Beyond rituximab: The future of monoclonal antibodies in B-cell non-Hodgkin lymphoma. Curr Hematol Malig Rep 2010; 3:187-93. [PMID: 20425465 DOI: 10.1007/s11899-008-0027-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The treatment of non-Hodgkin lymphoma (NHL) has changed dramatically since the introduction of rituximab, a monoclonal antibody that binds to the B-cell transmembrane protein CD20 and causes lysis of the lymphoma cells. Since then, a number of additional antibodies have been tested against other B-cell targets, resulting in variable efficacies. The goal of these newer agents is to achieve similar or better response rates as seen with rituximab and perhaps demonstrate activity in rituximab-refractory disease. Several of the antibodies have been investigated in combination with each other as well as with conventional chemotherapeutic regimens. Approval of such antibodies by regulatory committees and their eventual integration into clinical practice will likely depend on positive results from randomized trials.
Collapse
Affiliation(s)
- Manzurul A Sikder
- James P. Wilmot Cancer Center, Division of Hematology-Oncology, University of Rochester, 601 Elmwood Avenue,Box 704, Rochester, NY 14642, USA
| | | |
Collapse
|
44
|
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
|
45
|
Abstract
Twenty-five years after the discovery of HIV as the cause of AIDS, tremendous progress has been made in reducing the morbidity and mortality associated with this disease. However, there is still neither a cure nor a vaccine for HIV infection. This shortcoming is likely a consequence of our incomplete understanding of the mechanisms of AIDS pathogenesis, particularly with regard to the interaction between the virus and the host immune system. In this article we review a series of recent advances in specific areas of HIV immunopathogenesis research: virus-host cell interactions; immune responses to HIV; chronic immune activation; and natural simian immunodeficiency virus infections. Finally, we discuss the potential implications of these findings for HIV/AIDS therapy and vaccines.
Collapse
Affiliation(s)
- Alexandra M Ortiz
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 705 Stellar-Chance Laboratories, 422 Curie Boulevard, Philadelphia, PA 19104, USA
| | | |
Collapse
|
46
|
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: 2.2] [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
|
47
|
Nonpathogenic simian immunodeficiency virus infection of sooty mangabeys is not associated with high levels of autologous neutralizing antibodies. J Virol 2010; 84:6248-53. [PMID: 20375163 DOI: 10.1128/jvi.00295-10] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [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-host species, such as sooty mangabeys (SMs), is characterized by a high level of viral replication and a low level of generalized immune activation, despite evidence of an adaptive immune response. Here the ability of SIV-infected SMs to mount neutralizing antibodies (Nab) against autologous virus was compared to that of human immunodeficiency virus type 1 (HIV-1) subtype C-infected subjects. While high levels of Nab were observed in HIV-1 infection, samples obtained at comparable time points from SM exhibited relatively low titers of autologous Nab. Nevertheless, SM plasma with higher Nab titers also contained elevated peripheral CD4(+) T-cell levels, suggesting a potential immunologic benefit for SMs. These data indicate that AIDS resistance in these primates is not due to high Nab titers and raise the possibility that low levels of Nab might be an inherent feature of natural-host SIV infections.
Collapse
|
48
|
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: 2.2] [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
|
49
|
Arias JF, Nishihara R, Bala M, Ikuta K. High systemic levels of interleukin-10, interleukin-22 and C-reactive protein in Indian patients are associated with low in vitro replication of HIV-1 subtype C viruses. Retrovirology 2010; 7:15. [PMID: 20211031 PMCID: PMC2841095 DOI: 10.1186/1742-4690-7-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Accepted: 03/09/2010] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND HIV-1 subtype C (HIV-1C) accounts for almost 50% of all HIV-1 infections worldwide and predominates in countries with the highest case-loads globally. Functional studies suggest that HIV-1C is unique in its biological properties, and there are contradicting reports about its replicative characteristics. The present study was conducted to evaluate whether the host cytokine environment modulates the in vitro replication capacity of HIV-1C viruses. METHODS A small subset of HIV-1C isolates showing efficient replication in peripheral blood mononuclear cells (PBMC) is described, and the association of in vitro replication capacity with disease progression markers and the host cytokine response was evaluated. Viruses were isolated from patient samples, and the corresponding in vitro growth kinetics were determined by monitoring for p24 production. Genotype, phenotype and co-receptor usage were determined for all isolates, while clinical category, CD4 cell counts and viral loads were recorded for all patients. Plasmatic concentrations of cytokines and, acute-phase response, and microbial translocation markers were determined; and the effect of cytokine treatment on in vitro replication rates was also measured. RESULTS We identified a small number of viral isolates showing high in vitro replication capacity in healthy-donor PBMC. HIV-1C usage of CXCR4 co-receptor was rare; therefore, it did not account for the differences in replication potential observed. There was also no correlation between the in vitro replication capacity of HIV-1C isolates and patients' disease status. Efficient virus growth was significantly associated with low interleukin-10 (IL-10), interleukin-22 (IL-22), and C-reactive protein (CRP) levels in plasma (p < .0001). In vitro, pretreatment of virus cultures with IL-10 and CRP resulted in a significant reduction of virus production, whereas IL-22, which lacks action on immune cells appears to mediate its anti-HIV effect through interaction with both IL-10 and CRP, and its own protective effect on mucosal membranes. CONCLUSIONS These results indicate that high systemic levels of IL-10, CRP and IL-22 in HIV-1C-infected Indian patients are associated with low viral replication in vitro, and that the former two have direct inhibitory effects whereas the latter acts through downstream mechanisms that remain uncertain.
Collapse
Affiliation(s)
- Juan F Arias
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Viral Emergent Diseases Research Group (VIREM), Universidad del Valle, Cali, Colombia
| | - Reiko Nishihara
- Department of Health Promotion Sciences, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Manju Bala
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Regional STD Teaching, Training and Research Center, VM Medical College & Safdarjang Hospital, New Delhi, India
| | - Kazuyoshi Ikuta
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
50
|
Suppression of adaptive immune responses during primary SIV infection of sabaeus African green monkeys delays partial containment of viremia but does not induce disease. Blood 2010; 115:3070-8. [PMID: 20147699 DOI: 10.1182/blood-2009-10-245225] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
One of the most puzzling observations in HIV research is the lack of pathogenicity in most nonhuman primate species that are natural hosts of simian immunodeficiency virus (SIV) infection. Despite this, natural hosts experience a level of viremia similar to humans infected with HIV or macaques infected with SIV. To determine the role of adaptive immune responses in viral containment and lack of disease, we delayed the generation of cellular and humoral immune responses by administering anti-CD8- and anti-CD20 lymphocyte-depleting antibodies to sabaeus African green monkeys (Chlorocebus sabaeus) before challenge with SIV(sab9315BR). In vivo lymphocyte depletion during primary infection resulted in a brief elevation of viremia but not in disease. Based on the magnitude and timing of SIV-specific CD8(+) T-cell responses in the lymphocyte-depleted animals, CD8(+) T-cell responses appear to contribute to viral containment in natural hosts. We found no evidence for a contribution of humoral immune responses in viral containment. These studies indicate that natural hosts have developed mechanisms in addition to classic adaptive immune responses to cope with this lentiviral infection. Thus, adaptive immune responses in natural hosts appear to be less critical for viral containment than in HIV infection.
Collapse
|