51
|
Barrenas F, Raehtz K, Xu C, Law L, Green RR, Silvestri G, Bosinger SE, Nishida A, Li Q, Lu W, Zhang J, Thomas MJ, Chang J, Smith E, Weiss JM, Dawoud RA, Richter GH, Trichel A, Ma D, Peng X, Komorowski J, Apetrei C, Pandrea I, Gale M. Macrophage-associated wound healing contributes to African green monkey SIV pathogenesis control. Nat Commun 2019; 10:5101. [PMID: 31704931 PMCID: PMC6841668 DOI: 10.1038/s41467-019-12987-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/08/2019] [Indexed: 01/13/2023] Open
Abstract
Natural hosts of simian immunodeficiency virus (SIV) avoid AIDS despite lifelong infection. Here, we examined how this outcome is achieved by comparing a natural SIV host, African green monkey (AGM) to an AIDS susceptible species, rhesus macaque (RM). To asses gene expression profiles from acutely SIV infected AGMs and RMs, we developed a systems biology approach termed Conserved Gene Signature Analysis (CGSA), which compared RNA sequencing data from rectal AGM and RM tissues to various other species. We found that AGMs rapidly activate, and then maintain, evolutionarily conserved regenerative wound healing mechanisms in mucosal tissue. The wound healing protein fibronectin shows distinct tissue distribution and abundance kinetics in AGMs. Furthermore, AGM monocytes exhibit an embryonic development and repair/regeneration signature featuring TGF-β and concomitant reduced expression of inflammatory genes compared to RMs. This regenerative wound healing process likely preserves mucosal integrity and prevents inflammatory insults that underlie immune exhaustion in RMs.
Collapse
Affiliation(s)
- Fredrik Barrenas
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Kevin Raehtz
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cuiling Xu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lynn Law
- Department of Immunology, University of Washington, Seattle, WA, USA
- Center for Innate Immunity and Immune Diseases, University of Washington, Seattle, WA, USA
| | - Richard R Green
- Department of Immunology, University of Washington, Seattle, WA, USA
- Center for Innate Immunity and Immune Diseases, University of Washington, Seattle, WA, USA
| | - Guido Silvestri
- Department of Pathology & Laboratory Medicine, Emory University, Atlanta, GA, USA
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Steven E Bosinger
- Department of Pathology & Laboratory Medicine, Emory University, Atlanta, GA, USA
- Division of Microbiology & Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Wuxun Lu
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jianshui Zhang
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Matthew J Thomas
- Department of Immunology, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | - Jean Chang
- Department of Immunology, University of Washington, Seattle, WA, USA
- Center for Innate Immunity and Immune Diseases, University of Washington, Seattle, WA, USA
| | - Elise Smith
- Department of Immunology, University of Washington, Seattle, WA, USA
- Center for Innate Immunity and Immune Diseases, University of Washington, Seattle, WA, USA
| | - Jeffrey M Weiss
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Reem A Dawoud
- Department of Pathology & Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - George H Richter
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anita Trichel
- Divison of Laboratory Animal Resources, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dongzhu Ma
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xinxia Peng
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Jan Komorowski
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
- Institute of Computer Science, PAN, Warsaw, Poland
| | - Cristian Apetrei
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ivona Pandrea
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, WA, USA.
- Center for Innate Immunity and Immune Diseases, University of Washington, Seattle, WA, USA.
- Washington National Primate Research Center, University of Washington, Seattle, WA, USA.
| |
Collapse
|
52
|
Abstract
Viruses are causative agents for many diseases and infect all living organisms on the planet. Development of effective therapies has relied on our ability to isolate and culture viruses in vitro, allowing mechanistic studies and strategic interventions. While this reductionist approach is necessary, testing the relevance of in vitro findings often takes a very long time. New developments in imaging technologies are transforming our experimental approach where viral pathogenesis can be studied in vivo at multiple spatial and temporal resolutions. Here, we outline a vision of a top-down approach using noninvasive whole-body imaging as a guide for in-depth characterization of key tissues, physiologically relevant cell types, and pathways of spread to elucidate mechanisms of virus spread and pathogenesis. Tool development toward imaging of infectious diseases is expected to transform clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Pradeep D Uchil
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06510, USA; , , ,
| | - Kelsey A Haugh
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06510, USA; , , ,
| | - Ruoxi Pi
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06510, USA; , , ,
| | - Walther Mothes
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06510, USA; , , ,
| |
Collapse
|
53
|
Differential Dynamics of Regulatory T-Cell and Th17 Cell Balance in Mesenteric Lymph Nodes and Blood following Early Antiretroviral Initiation during Acute Simian Immunodeficiency Virus Infection. J Virol 2019; 93:JVI.00371-19. [PMID: 31315987 PMCID: PMC6744245 DOI: 10.1128/jvi.00371-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 07/05/2019] [Indexed: 12/24/2022] Open
Abstract
Tregs contribute to SIV/HIV disease progression by inhibition of antiviral specific responses and effector T-cell proliferation. Tregs also cause tissue fibrosis via transforming growth factor β1 production and collagen deposition, which are associated with microbial translocation and generalized immune activation. Early ARV initiation upon viral exposure is recommended globally and results in improved immune function recovery and reduced viral persistence. Here, using an acute SIV infection model of rhesus macaques, we demonstrated for the first time that despite clear improvements in mucosal CD4 T cells, in contrast to blood, Treg frequencies in MLNs remained elevated following early ARV initiation. The particular Th17/Treg balance observed in MLNs can contribute, in part, to the maintenance of mucosal fibrosis during suppressive ARV treatment. Our results provide a better understanding of gut mucosal immune dynamics following early ARV initiation. These findings suggest that Treg-based treatments could serve as a novel immunotherapeutic approach to decrease gut mucosal damage during SIV/HIV infections. Increased frequencies of immunosuppressive regulatory T cells (Tregs) are associated with gut lymphoid tissue fibrosis and dysfunction which, in turn, contribute to disease progression in chronic simian immunodeficiency virus/human immunodeficiency virus (SIV/HIV) infection. Mesenteric lymph nodes (MLNs), which drain the large and small intestine, are critical sites for the induction and maintenance of gut mucosal immunity. However, the dynamics of Tregs in MLNs are not well understood due to the lack of accessibility to these tissues in HIV-infected individuals. Here, the dynamics of Tregs in blood and MLNs were assessed in SIV-infected rhesus macaques (RMs) following early antiretroviral drug (ARV) initiation. Early ARV initiation reduced T-cell immune activation, as assessed by HLA-DR/CD39 expression, and prevented the depletion of memory CCR6+ Th17 cells in both blood and MLNs. Untreated animals showed higher frequencies of Tregs, CD39+ Tregs, thymic Tregs, and new memory CD4 populations sharing similarity with Tregs as CTLA4+ PD1– and CTLA4+ PD1– FoxP3+ T cells. Despite early ARV treatment, the frequencies of these Treg subsets remained unchanged within the MLNs and, in contrast to blood normalization, the Th17/Treg ratio remained distorted in MLNs. Furthermore, our results highlighted that the expressions of IDO-1, TGFβ1 and collagen-1 mRNA remained unchanged in MLN of ARV-treated RMs. ARV interruption did not affect T-cell immune activation and Th17/Treg ratios in MLN. Altogether, our data demonstrated that early ARV initiation within the first few days of SIV infection is unable to reduce the frequencies and homing of various subsets of Tregs within the MLNs which, in turn, may result in tissue fibrosis, impairment in MLN function, and HIV persistence. IMPORTANCE Tregs contribute to SIV/HIV disease progression by inhibition of antiviral specific responses and effector T-cell proliferation. Tregs also cause tissue fibrosis via transforming growth factor β1 production and collagen deposition, which are associated with microbial translocation and generalized immune activation. Early ARV initiation upon viral exposure is recommended globally and results in improved immune function recovery and reduced viral persistence. Here, using an acute SIV infection model of rhesus macaques, we demonstrated for the first time that despite clear improvements in mucosal CD4 T cells, in contrast to blood, Treg frequencies in MLNs remained elevated following early ARV initiation. The particular Th17/Treg balance observed in MLNs can contribute, in part, to the maintenance of mucosal fibrosis during suppressive ARV treatment. Our results provide a better understanding of gut mucosal immune dynamics following early ARV initiation. These findings suggest that Treg-based treatments could serve as a novel immunotherapeutic approach to decrease gut mucosal damage during SIV/HIV infections.
Collapse
|
54
|
Muñoz L, Borrero MJ, Úbeda M, Conde E, Del Campo R, Rodríguez-Serrano M, Lario M, Sánchez-Díaz AM, Pastor O, Díaz D, García-Bermejo L, Monserrat J, Álvarez-Mon M, Albillos A. Intestinal Immune Dysregulation Driven by Dysbiosis Promotes Barrier Disruption and Bacterial Translocation in Rats With Cirrhosis. Hepatology 2019; 70:925-938. [PMID: 30414342 DOI: 10.1002/hep.30349] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 11/05/2018] [Indexed: 12/17/2022]
Abstract
In cirrhosis, intestinal dysbiosis, intestinal barrier impairment, and systemic immune system abnormalities lead to gut bacterial translocation (GBT) and bacterial infection. However, intestinal immune system dysfunction and its contribution to barrier damage are poorly understood. This study correlates immune system dysregulation in the intestines of rats at different stages of CCl4 -induced cirrhosis with barrier function and pathogenic microbiota. The following variables were addressed in the small intestine: intraepithelial lymphocyte (IEL) and lamina propria lymphocyte (LPL) activation status and cytokine production (flow cytometry), cytokine mRNA and protein expression (quantitative real-time PCR and immunofluorescence), microbiota composition of ileum content (16S recombinant DNA massive sequencing), permeability (fecal albumin loss), and epithelial junctions (immunohistochemistry and immunofluorescence). The intestinal mucosa in rats with cirrhosis showed a proinflammatory pattern of immune dysregulation in IELs and LPLs, which featured the expansion of activated lymphocytes, switch to a T helper 1 (Th1) regulatory pattern, and Th17 reduction. In rats with cirrhosis with ascites, this state was associated with epithelial junction protein disruption, fecal albumin loss, and GBT. Direct correlations (P < 0.01) were observed between elevated interferon gamma (IFNγ)-expressing T cytotoxic LPLs and fecal albumin and between inflammatory taxa abundance and IFNγ-producing immune cells in the ileum. Bowel decontamination led to redistributed microbiota composition, reduced proinflammatory activation of mucosal immune cells, normalized fecal albumin levels, and diminished GBT; but there were no modifications in Th17 depletion. Conclusion: The intestinal mucosa of rats with cirrhosis acquires a proinflammatory profile of immune dysregulation that parallels the severity of cirrhosis; this impaired intestinal immune response is driven by gut dysbiosis and leads to disrupted barrier function, promoting GBT.
Collapse
Affiliation(s)
- Leticia Muñoz
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - María-José Borrero
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - María Úbeda
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Elisa Conde
- Unidad de Biomarcadores y Dianas Terapéuticas, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Rosa Del Campo
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, IRYCIS, Red Española de Investigación en Enfermedades Infecciosas (REIPI), Instituto de Salud Carlos III, Madrid, Spain
| | - Macarena Rodríguez-Serrano
- Unidad de Biomarcadores y Dianas Terapéuticas, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Margaret Lario
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana-María Sánchez-Díaz
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, IRYCIS, Red Española de Investigación en Enfermedades Infecciosas (REIPI), Instituto de Salud Carlos III, Madrid, Spain
| | - Oscar Pastor
- Servicio de Bioquímica Clínica, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - David Díaz
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.,Servicio de Enfermedades del Sistema Inmune y Oncología, Hospital Universitario Príncipe de Asturias, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Laura García-Bermejo
- Unidad de Biomarcadores y Dianas Terapéuticas, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Jorge Monserrat
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.,Servicio de Enfermedades del Sistema Inmune y Oncología, Hospital Universitario Príncipe de Asturias, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Melchor Álvarez-Mon
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.,Servicio de Enfermedades del Sistema Inmune y Oncología, Hospital Universitario Príncipe de Asturias, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Agustín Albillos
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.,Servicio de Gastroenterología y Hepatología, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, IRYCIS, Madrid, Spain
| |
Collapse
|
55
|
Swainson LA, Ahn H, Pajanirassa P, Khetarpal V, Deleage C, Estes JD, Hunt PW, Munoz-Sanjuan I, McCune JM. Kynurenine 3-Monooxygenase Inhibition during Acute Simian Immunodeficiency Virus Infection Lowers PD-1 Expression and Improves Post-Combination Antiretroviral Therapy CD4 + T Cell Counts and Body Weight. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:899-910. [PMID: 31285277 PMCID: PMC6684450 DOI: 10.4049/jimmunol.1801649] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/17/2019] [Indexed: 01/31/2023]
Abstract
The kynurenine pathway (KP) is a key regulator of many important physiological processes and plays a harmful role in cancer, many neurologic conditions, and chronic viral infections. In HIV infection, KP activity is consistently associated with reduced CD4 T cell counts and elevated levels of T cell activation and viral load; it also independently predicts mortality and morbidity from non-AIDS events. Kynurenine 3-monooxygenase (KMO) is a therapeutically important target in the KP. Using the nonhuman primate model of SIV infection in rhesus macaques, we investigated whether KMO inhibition could slow the course of disease progression. We used a KMO inhibitor, CHDI-340246, to perturb the KP during early acute infection and followed the animals for 1 y to assess clinical outcomes and immune phenotype and function during pre-combination antiretroviral therapy acute infection and combination antiretroviral therapy-treated chronic infection. Inhibition of KMO in acute SIV infection disrupted the KP and prevented SIV-induced increases in downstream metabolites, improving clinical outcome as measured by both increased CD4+ T cell counts and body weight. KMO inhibition increased naive T cell frequency and lowered PD-1 expression in naive and memory T cell subsets. Importantly, early PD-1 expression during acute SIV infection predicted clinical outcomes of body weight and CD4+ T cell counts. Our data indicate that KMO inhibition in early acute SIV infection provides clinical benefit and suggest a rationale for testing KMO inhibition as an adjunctive treatment in SIV/HIV infection to slow the progression of the disease and improve immune reconstitution.
Collapse
Affiliation(s)
- Louise A Swainson
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110;
| | - Haelee Ahn
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110
| | - Priya Pajanirassa
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110
| | | | - Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21701
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21701
| | - Peter W Hunt
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110
| | | | - Joseph M McCune
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94110
| |
Collapse
|
56
|
Morou A, Brunet-Ratnasingham E, Dubé M, Charlebois R, Mercier E, Darko S, Brassard N, Nganou-Makamdop K, Arumugam S, Gendron-Lepage G, Yang L, Niessl J, Baxter AE, Billingsley JM, Rajakumar PA, Lefebvre F, Johnson RP, Tremblay C, Routy JP, Wyatt RT, Finzi A, Douek DC, Kaufmann DE. Altered differentiation is central to HIV-specific CD4 + T cell dysfunction in progressive disease. Nat Immunol 2019; 20:1059-1070. [PMID: 31308541 PMCID: PMC6642691 DOI: 10.1038/s41590-019-0418-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 05/03/2019] [Indexed: 12/19/2022]
Abstract
Dysfunction of virus-specific CD4+ T cells in chronic human infections is poorly understood. We performed genome-wide transcriptional analyses and functional assays of CD4+ T cells specific for human immunodeficiency virus (HIV) from HIV-infected people before and after initiation of antiretroviral therapy (ART). A follicular helper T cell (TFH cell)-like profile characterized HIV-specific CD4+ T cells in viremic infection. HIV-specific CD4+ T cells from people spontaneously controlling the virus (elite controllers) robustly expressed genes associated with the TH1, TH17 and TH22 subsets of helper T cells. Viral suppression by ART resulted in a distinct transcriptional landscape, with a reduction in the expression of genes associated with TFH cells, but persistently low expression of genes associated with TH1, TH17 and TH22 cells compared to the elite controller profile. Thus, altered differentiation is central to the impairment of HIV-specific CD4+ T cells and involves both gain of function and loss of function.
Collapse
Affiliation(s)
- Antigoni Morou
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Elsa Brunet-Ratnasingham
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Mathieu Dubé
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, La Jolla, CA, USA
| | - Roxanne Charlebois
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Eloi Mercier
- Canadian Centre for Computational Genomics-Montréal Node, Montreal, Quebec, Canada
| | - Sam Darko
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Nathalie Brassard
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | | | - Sahaana Arumugam
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Gabrielle Gendron-Lepage
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Lifei Yang
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Julia Niessl
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, La Jolla, CA, USA
| | - Amy E Baxter
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, La Jolla, CA, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - James M Billingsley
- Yerkes National Primate Research Center and Emory University, Atlanta, GA, USA
| | | | - François Lefebvre
- Canadian Centre for Computational Genomics-Montréal Node, Montreal, Quebec, Canada
| | - R Paul Johnson
- Yerkes National Primate Research Center and Emory University, Atlanta, GA, USA
| | - Cécile Tremblay
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illnesses Service and Division of Hematology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Richard T Wyatt
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, La Jolla, CA, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Andrés Finzi
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Daniel C Douek
- Human Immunology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Daniel E Kaufmann
- Research Centre of the Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.
- Université de Montréal, Montreal, Quebec, Canada.
- Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, La Jolla, CA, USA.
| |
Collapse
|
57
|
Ceccarelli G, Statzu M, Santinelli L, Pinacchio C, Bitossi C, Cavallari EN, Vullo V, Scagnolari C, d'Ettorre G. Challenges in the management of HIV infection: update on the role of probiotic supplementation as a possible complementary therapeutic strategy for cART treated people living with HIV/AIDS. Expert Opin Biol Ther 2019; 19:949-965. [PMID: 31260331 DOI: 10.1080/14712598.2019.1638907] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Recent insights show that gut-mucosal immunity and intestinal microbiota play a key role in the pathogenesis of HIV infection. Alterations in the composition of intestinal flora (dysbiosis) could be associated with an impaired intestinal epithelium barrier activity and an impaired mucosal immunity function, significantly contributing to microbial translocation which is considered a major driver of chronic immune activation. Areas covered: This article provides an overview on the novel trends in probiotic therapy application. A particular emphasis is addressed to the importance of probiotics as a novel strategy to attenuate or prevent gastrointestinal involvement and to improve gut-mucosal immunity in HIV-infected subjects. Therefore, opportunities, limits and methodological criticalities of supplementation with probiotic therapy are considered and analyzed. Expert opinion: Use of probiotics is emerging as a novel strategy to manage dysbiosis and gut-mucosal impairment, to reduce immune activation and to limit a number of non-AIDS-related disorders. However, despite the growing use of probiotic therapy, mechanisms by which oral bacteria intake exhibits its effects are strain-related and disease-specific, hence clinicians need to take these two factors into consideration when suggesting probiotic supplementation to HIV-infected patients.
Collapse
Affiliation(s)
- Giancarlo Ceccarelli
- a Department of Public Health and Infectious Diseases, Sapienza University of Rome , Rome , Italy
| | - Maura Statzu
- b Laboratory of Virology, Department of Molecular Medicine, affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome , Rome , Italy
| | - Letizia Santinelli
- b Laboratory of Virology, Department of Molecular Medicine, affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome , Rome , Italy
| | - Claudia Pinacchio
- a Department of Public Health and Infectious Diseases, Sapienza University of Rome , Rome , Italy
| | - Camilla Bitossi
- b Laboratory of Virology, Department of Molecular Medicine, affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome , Rome , Italy
| | - Eugenio Nelson Cavallari
- a Department of Public Health and Infectious Diseases, Sapienza University of Rome , Rome , Italy
| | - Vincenzo Vullo
- a Department of Public Health and Infectious Diseases, Sapienza University of Rome , Rome , Italy
| | - Carolina Scagnolari
- b Laboratory of Virology, Department of Molecular Medicine, affiliated to Istituto Pasteur Italia - Cenci Bolognetti Foundation, Sapienza University of Rome , Rome , Italy
| | - GabrieIla d'Ettorre
- a Department of Public Health and Infectious Diseases, Sapienza University of Rome , Rome , Italy
| |
Collapse
|
58
|
Nugeyre MT, Tchitchek N, Adapen C, Cannou C, Contreras V, Benjelloun F, Ravel J, Le Grand R, Marlin R, Menu E. Dynamics of Vaginal and Rectal Microbiota Over Several Menstrual Cycles in Female Cynomolgus Macaques. Front Cell Infect Microbiol 2019; 9:188. [PMID: 31249812 PMCID: PMC6582644 DOI: 10.3389/fcimb.2019.00188] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/16/2019] [Indexed: 12/12/2022] Open
Abstract
The composition of the microbiota in cynomolgus macaques is only partially characterized, although this animal model is often used to study pathogenesis and preventive strategies against infections. We thus performed, for the first time, a longitudinal characterization of the vaginal and rectal microbiota of five cycling female cynomolgus macaques. Samples were collected weekly for 15 weeks and the V3/V4 regions of the16S rRNA gene sequenced. Sequences were analyzed with QIIME for OTU detection and taxonomic assignment. Progesterone levels were also determined to evaluate hormonal influence on bacteria relative abundance. The rectal and vaginal bacterial composition in cynomolgus macaques is polymicrobial and clearly distinct, with larger individual variability in the vagina. Rectal microbiota profiles were consistent between animals, whereas they were highly variable and animal-specific in the vagina. In the rectum, the most abundant taxa were Ruminococcaceae, Prevotella, and Clostridiales. In the vagina, the most abundant genera were Sneathia, Porphyromonas, Prevotella, and Fusobacterium. Lactobacillus were found at relative abundances higher than 1% in only one animal and were not predominant. Comparison of the vaginal cynomolgus macaque microbiota with that of humans showed similarity to community state type IV-A usually associated with dysbiosis. In the vagina, the relative abundance of 12 bacterial genera was found to be associated with progesterone levels. Our study provides a detailed characterization of the rectal and vaginal microbiota in female cynomolgus macaques and opens new perspectives of this animal model.
Collapse
Affiliation(s)
- Marie-Thérèse Nugeyre
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| | - Nicolas Tchitchek
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Cindy Adapen
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Claude Cannou
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| | - Vanessa Contreras
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Fahd Benjelloun
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| | - Jacques Ravel
- Institute for Genome Sciences and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Roger Le Grand
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Romain Marlin
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Elisabeth Menu
- CEA, Université Paris-Sud, Inserm, U1184 "Immunology of Viral Infections and Autoimmune Diseases" (IMVA), IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| |
Collapse
|
59
|
Lv Y, Wang X. Interleukin-37 Inhibits the Imbalance Between T Helper 17 Cells and Regulatory T Cells in Hand, Foot, and Mouth Disease. J Interferon Cytokine Res 2019; 39:421-427. [PMID: 31090483 DOI: 10.1089/jir.2019.0005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The aim of this study was to explore the role of interleukin-37 (IL-37) in imbalance of T helper (Th)17/regulatory T cells (Tregs) in hand, foot, and mouth disease (HFMD). The proportions of CD4+ IL-17A+ Th17 cells and CD4+ CD25+Foxp3+ Tregs in peripheral blood or peripheral blood mononuclear cells (PBMCs) from HFMD patients and healthy controls were measured by fluorescence activated cell sorter. The level of IL-37, IL-10, IL-17A, IL-23, and transforming growth factor β1 (TGF-β1) in serum or PBMCs of HFMD patients and control subjects were detected using enzyme-linked immunosorbent assay. Results showed that Th17 cells proportion and IL-17A and IL-23 levels were highly increased, whereas Tregs proportion and IL-10 and TGF-β1 levels were significantly decreased in HFMD patients. Moreover, IL-37 stimulation elevated Tregs proportion but reduced Th17 cell proportion in subjects with HFMD. On the contrary, we found methylprednisolone pulse therapy/methylprednisolone combinated with intravenous gamma globulin inhibits Th17/Treg imbalance through upregulation of IL-37 in HFMD. In conclusion, IL-37 inhibits the imbalance of Th17/Tregs in HFMD.
Collapse
Affiliation(s)
- Yong Lv
- Department of Pediatric, The First Affiliated Hospital of University of Science and Technology of China (USTC), Hefei, China
| | - Xuesong Wang
- Department of Pediatric, The First Affiliated Hospital of University of Science and Technology of China (USTC), Hefei, China
| |
Collapse
|
60
|
Simian Immunodeficiency Virus Infects Functionally Polarized Memory CD4 T Cells Equivalently In Vivo. J Virol 2019; 93:JVI.02163-18. [PMID: 30787150 DOI: 10.1128/jvi.02163-18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/11/2019] [Indexed: 11/20/2022] Open
Abstract
Among the numerous immunological abnormalities observed in chronically human immunodeficiency virus (HIV)-infected individuals, perturbations in memory CD4 T cells are thought to contribute specifically to disease pathogenesis. Among these, functional imbalances in the frequencies of T regulatory cells (Tregs) and interleukin 17 (IL-17)/IL-22-producing Th cells (Th17/Th22) from mucosal sites and T follicular helper (Tfh) cells in lymph nodes are thought to facilitate specific aspects of disease pathogenesis. However, while preferential infection of Tfh cells is widely thought to create an important viral reservoir in an immunologically privileged site in vivo, whether immunological perturbations among memory CD4 T cell populations are attributable to their relative infectivity by the virus in vivo is unclear. Here we studied peripheral blood and lymphoid tissues from antiretroviral (ARV)-treated and ARV-naive Asian macaques and isolated functionally defined populations of memory CD4 T cells. We then assessed the degree to which these populations were infected by simian immunodeficiency virus (SIV) in vivo, to determine whether particular functionally identified populations of memory CD4 T cells were preferentially infected by the virus. We found that SIV did not preferentially infect Th17 cells, compared to Th1 cells, Th2 cells, or Tregs. Moreover, Th17 cells contributed proportionately to the total pool of infected cells. Taken together, our data suggest that, although Tfh cells are more prone to harbor viral DNA, other functionally polarized cells are equally infected by the virus in vivo and Th17 cells are not preferentially infected.IMPORTANCE Functional perturbations of memory CD4 T cells have been suggested to underlie important aspects of HIV disease progression. However, the mechanisms underlying these perturbations remain unclear. Using a nonhuman primate model of HIV, we show that SIV infects functionally defined populations of memory CD4 T cells equally in different anatomic sites. Thus, preferential infection by the virus is unlikely to cause functional perturbations.
Collapse
|
61
|
Abstract
As our understanding of mucosal immunity increases, it is becoming clear that the host response to HIV-1 is more complex and nuanced than originally believed. The mucosal landscape is populated with a variety of specialized cell types whose functions include combating infectious agents while preserving commensal microbiota, maintaining barrier integrity, and ensuring immune homeostasis. Advances in multiparameter flow cytometry, gene expression analysis and bioinformatics have allowed more detailed characterization of these cell types and their roles in host defense than was previously possible. This review provides an overview of existing literature on immunity to HIV-1 and SIVmac in mucosal tissues of the female reproductive tract and the gastrointestinal tract, focusing on major effector cell populations and briefly summarizing new information on tissue resident memory T cells, Treg, Th17, Th22 and innate lymphocytes (ILC), subsets that have been studied primarily in the gastrointestinal mucosa.
Collapse
Affiliation(s)
- Barbara L Shacklett
- Department of Medical Microbiology and Immunology.,Division of Infectious Diseases, Department of Medicine, School of Medicine, University of California, Davis, CA 95616
| |
Collapse
|
62
|
Wetzel KS, Elliott STC, Collman RG. SIV Coreceptor Specificity in Natural and Non-Natural Host Infection: Implications for Cell Targeting and Differential Outcomes from Infection. Curr HIV Res 2019; 16:41-51. [PMID: 29173179 DOI: 10.2174/1570162x15666171124121805] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/07/2017] [Accepted: 07/07/2017] [Indexed: 11/22/2022]
Abstract
Pathogenic HIV-1 infection of humans and SIVmac infection of macaques are the result of zoonotic transfer of primate immunodeficiency viruses from their natural hosts into non-natural host species. Natural host infections do not result in pathogenesis despite high levels of virus replication, and evidence suggests that differences in anatomical location and specific subsets of CD4+ T cells infected may underlie distinct outcomes from infection. The coreceptor CCR5 has long been considered the sole pathway for SIV entry and the key determinant of CD4+ cell targeting, but it has also been known that natural hosts express exceedingly low levels of CCR5 despite maintaining high levels of virus replication. This review details emerging data indicating that in multiple natural host species, CCR5 is dispensable for SIV infection ex vivo and/or in vivo and, contrary to the established dogma, alternative coreceptors, particularly CXCR6, play a central role in infection and cell targeting. Infections of non-natural hosts, however, are characterized by CCR5-exclusive entry. These findings suggest that alternative coreceptor-mediated cell targeting in natural hosts, combined with low CCR5 expression, may direct the virus to distinct populations of cells that are dispensable for immune homeostasis, particularly extralymphoid and more differentiated CD4+ T cells. In contrast, CCR5-mediated entry in non-natural hosts results in targeting of CD4+ T cells that are located in lymphoid tissues, critical for immune homeostasis, or necessary for gut barrier integrity. Thus, fundamental differences in viral entry coreceptor use may be central determinants of infection outcome. These findings redefine the normal SIV/host relationship in natural host species, shed new light on key features linked to zoonotic immunodeficiency virus transfer, and highlight important questions regarding how and why this coreceptor bottleneck occurs and the coevolutionary equilibrium is lost following cross-species transfer that results in AIDS.
Collapse
Affiliation(s)
- Katherine S Wetzel
- Department of Medicine and Penn Center for AIDS Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Sarah T C Elliott
- Department of Medicine and Penn Center for AIDS Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Ronald G Collman
- Department of Medicine and Penn Center for AIDS Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
63
|
Loiseau C, Requena M, Nayrac M, Mavigner M, Cazabat M, Iscache AL, Carrere N, Suc B, Alric L, Izopet J, Delobel P. Increased CXCR3+ T Cells Impairs Recruitment of T-Helper Type 17 Cells via Interferon γ and Interleukin 18 in the Small Intestine Mucosa During Treated HIV-1 Infection. J Infect Dis 2019; 220:830-840. [DOI: 10.1093/infdis/jiz123] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 03/14/2019] [Indexed: 12/14/2022] Open
Abstract
Abstract
The restoration of CD4+ T cells, especially T-helper type 17 (Th17) cells, remains incomplete in the gut mucosa of most human immunodeficiency virus type 1 (HIV-1)–infected individuals despite sustained antiretroviral therapy (ART). Herein, we report an increase in the absolute number of CXCR3+ T cells in the duodenal mucosa during ART. The frequencies of Th1 and CXCR3+ CD8+ T cells were increased and negatively correlated with CCL20 and CCL25 expression in the mucosa. In ex vivo analyses, we showed that interferon γ, the main cytokine produced by Th1 and effector CD8+ T cells, downregulates the expression of CCL20 and CCL25 by small intestine enterocytes, while it increases the expression of CXCL9/10/11, the ligands of CXCR3. Interleukin 18, a pro-Th1 cytokine produced by enterocytes, also contributes to the downregulation of CCL20 expression and increases interferon γ production by Th1 cells. This could perpetuate an amplification loop for CXCR3-driven Th1 and effector CD8+ T cells recruitment to the gut, while impairing Th17 cells homing through the CCR6-CCL20 axis in treated HIV-1–infected individuals.
Collapse
Affiliation(s)
- C Loiseau
- INSERM, UMR1043, Toulouse, France
- aPresent affiliation: Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - M Requena
- Laboratoire de Virologie, Toulouse, France
| | - M Nayrac
- INSERM, UMR1043, Toulouse, France
| | - M Mavigner
- Department of Pediatrics, Atlanta, Georgia
- Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia
| | - M Cazabat
- Laboratoire de Virologie, Toulouse, France
| | | | - N Carrere
- Service de Chirurgie générale et digestive, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| | - B Suc
- Service de Chirurgie générale et digestive, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| | - L Alric
- Service de Médecine Interne, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
- IRD UMR152, Toulouse, France
| | - J Izopet
- INSERM, UMR1043, Toulouse, France
- Laboratoire de Virologie, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| | - P Delobel
- INSERM, UMR1043, Toulouse, France
- Service des Maladies Infectieuses et Tropicales, CHU de Toulouse, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
64
|
O'Connor MA, Munson PV, Tunggal HC, Hajari N, Lewis TB, Bratt D, Moats C, Smedley J, Bagley KC, Mullins JI, Fuller DH. Mucosal T Helper 17 and T Regulatory Cell Homeostasis Correlate with Acute Simian Immunodeficiency Virus Viremia and Responsiveness to Antiretroviral Therapy in Macaques. AIDS Res Hum Retroviruses 2019; 35:295-305. [PMID: 30398361 DOI: 10.1089/aid.2018.0184] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Depletion of gut T helper 17 (Th17) cells during HIV infection leads to decreased mucosal integrity and increased disease progression. Conversely, T regulatory (Treg) cells may inhibit antiviral responses or immune activation. In HIV elite controllers, a balanced Th17/Treg ratio is maintained in the blood, suggesting a role for these responses in controlling inflammation and viral replication. HIV-infected individuals exhibit a range in responsiveness to combination antiretroviral therapy (cART). Given the link between the Th17/Treg ratio and HIV disease, we reasoned these responses may play a role in cART responsiveness. In this study, we investigated the relationship between the mucosal Th17/Treg ratio to acute simian immunodeficiency virus (SIV) viremia and the response to cART. Nineteen rhesus macaques were infected with highly pathogenic SIVΔB670 virus and cART was initiated 6 weeks postinfection. Mucosal CD4 T cell subsets were assessed by intracellular cytokine staining in the colon and mesenteric lymph nodes. Higher baseline Th17/Treg ratios corresponded with increased acute SIV viremia. Th17/Treg ratios decreased during acute SIV infection and were not restored during cART, and this corresponded to increased gut immune activation (Ki67+), markers of microbial translocation (sCD14), and T cell exhaustion (TIGIT+). Animals that maintained a more balanced mucosal Th17/Treg ratio at the time of cART initiation exhibited a better virological response to cART and maintained higher peripheral CD4 counts. These results suggest mucosal Th17 and Treg homeostasis influences acute viremia and the response to cART, a result that suggests therapeutic interventions that improve the Th17/Treg ratio before or during cART may improve treatment of HIV.
Collapse
Affiliation(s)
- Megan A. O'Connor
- Department of Microbiology, University of Washington, Seattle, Washington
- Washington National Primate Research Center, Seattle, Washington
| | - Paul V. Munson
- Department of Microbiology, University of Washington, Seattle, Washington
- Washington National Primate Research Center, Seattle, Washington
| | - Hillary C. Tunggal
- Department of Microbiology, University of Washington, Seattle, Washington
- Washington National Primate Research Center, Seattle, Washington
| | - Nika Hajari
- Department of Microbiology, University of Washington, Seattle, Washington
- Washington National Primate Research Center, Seattle, Washington
| | - Thomas B. Lewis
- Department of Microbiology, University of Washington, Seattle, Washington
- Washington National Primate Research Center, Seattle, Washington
| | - Debra Bratt
- Washington National Primate Research Center, Seattle, Washington
| | - Cassie Moats
- Washington National Primate Research Center, Seattle, Washington
| | - Jeremy Smedley
- Washington National Primate Research Center, Seattle, Washington
| | | | - James I. Mullins
- Department of Microbiology, University of Washington, Seattle, Washington
| | - Deborah H. Fuller
- Department of Microbiology, University of Washington, Seattle, Washington
- Washington National Primate Research Center, Seattle, Washington
| |
Collapse
|
65
|
Roider J, Ngoepe A, Muenchhoff M, Adland E, Groll A, Ndung'u T, Kløverpris H, Goulder P, Leslie A. Increased Regulatory T-Cell Activity and Enhanced T-Cell Homeostatic Signaling in Slow Progressing HIV-infected Children. Front Immunol 2019; 10:213. [PMID: 30809229 PMCID: PMC6379343 DOI: 10.3389/fimmu.2019.00213] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/24/2019] [Indexed: 12/14/2022] Open
Abstract
Pediatric slow progressors (PSP) are rare ART-naïve, HIV-infected children who maintain high CD4 T-cell counts and low immune activation despite persistently high viral loads. Using a well-defined cohort of PSP, we investigated the role of regulatory T-cells (TREG) and of IL-7 homeostatic signaling in maintaining normal-for-age CD4 counts in these individuals. Compared to children with progressive disease, PSP had greater absolute numbers of TREG, skewed toward functionally suppressive phenotypes. As with immune activation, overall T-cell proliferation was lower in PSP, but was uniquely higher in central memory TREG (CM TREG), indicating active engagement of this subset. Furthermore, PSP secreted higher levels of the immunosuppressive cytokine IL-10 than children who progressed. The frequency of suppressive TREG, CM TREG proliferation, and IL-10 production were all lower in PSP who go on to progress at a later time-point, supporting the importance of an active TREG response in preventing disease progression. In addition, we find that IL-7 homeostatic signaling is enhanced in PSP, both through preserved surface IL-7receptor (CD127) expression on central memory T-cells and increased plasma levels of soluble IL-7receptor, which enhances the bioactivity of IL-7. Combined analysis, using a LASSO modeling approach, indicates that both TREG activity and homeostatic T-cell signaling make independent contributions to the preservation of CD4 T-cells in HIV-infected children. Together, these data demonstrate that maintenance of normal-for-age CD4 counts in PSP is an active process, which requires both suppression of immune activation through functional TREG, and enhanced T-cell homeostatic signaling.
Collapse
Affiliation(s)
- Julia Roider
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, Oxford University, Oxford, United Kingdom
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Infectious Diseases, Medizinische Klinik IV, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Abigail Ngoepe
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Maximilian Muenchhoff
- Department of Virology, Max von Pettenkofer Institute, Ludwig-Maximilians-University Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Emily Adland
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, Oxford University, Oxford, United Kingdom
| | - Andreas Groll
- Faculty of Statistics, TU Dortmund University, Dortmund, Germany
| | - Thumbi Ndung'u
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
- Max Planck Institute for Infection Biology, Berlin, Germany
- Department of Infection and Immunity, University College London, London, United Kingdom
| | - Henrik Kløverpris
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Infection and Immunity, University College London, London, United Kingdom
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Philip Goulder
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, Oxford University, Oxford, United Kingdom
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Alasdair Leslie
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
66
|
Hensley-McBain T, Berard AR, Manuzak JA, Miller CJ, Zevin AS, Polacino P, Gile J, Agricola B, Cameron M, Hu SL, Estes JD, Reeves RK, Smedley J, Keele BF, Burgener AD, Klatt NR. Intestinal damage precedes mucosal immune dysfunction in SIV infection. Mucosal Immunol 2018; 11:1429-1440. [PMID: 29907866 PMCID: PMC6162106 DOI: 10.1038/s41385-018-0032-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/23/2018] [Accepted: 04/02/2018] [Indexed: 02/04/2023]
Abstract
HIV and pathogenic SIV infection are characterized by mucosal dysfunction including epithelial barrier damage, loss of Th17 cells, neutrophil infiltration, and microbial translocation with accompanying inflammation. However, it is unclear how and when these contributing factors occur relative to one another. In order to determine whether any of these features initiates the cycle of damage, we longitudinally evaluated the kinetics of mucosal and systemic T-cell activation, microbial translocation, and Th17 cell and neutrophil frequencies following intrarectal SIV infection of rhesus macaques. We additionally assessed the colon proteome to elucidate molecular pathways altered early after infection. We demonstrate increased T-cell activation (HLA-DR+) beginning 3-14 days post-SIV challenge, reduced peripheral zonulin 3-14 days post-SIV, and evidence of microbial translocation 14 days post-SIV. The onset of mucosal dysfunction preceded peripheral and mucosal Th17 depletion, which occurred 14-28 days post-SIV, and gut neutrophil accumulation was not observed. Proteins involved in epithelial structure were downregulated 3 days post-SIV followed by an upregulation of immune proteins 14 days post-SIV. These data demonstrate that immune perturbations such as Th17 loss and neutrophil infiltration occur after alterations to epithelial structural protein pathways, suggesting that epithelial damage occurs prior to widespread immune dysfunction.
Collapse
Affiliation(s)
- Tiffany Hensley-McBain
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | - Alicia R Berard
- National HIV and Retrovirology Labs, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Jennifer A Manuzak
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | - Charlene J Miller
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | - Alexander S Zevin
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | | | - Jillian Gile
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | - Brian Agricola
- Washington National Primate Research Center, Seattle, WA, USA
| | - Mark Cameron
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Shiu-Lok Hu
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, USA
| | - Jacob D Estes
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - R Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Jeremy Smedley
- Washington National Primate Research Center, Seattle, WA, USA
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Adam D Burgener
- National HIV and Retrovirology Labs, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
- Unit of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nichole R Klatt
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA.
- Washington National Primate Research Center, Seattle, WA, USA.
| |
Collapse
|
67
|
Chaudhary O, Narayan V, Lelis F, Linz B, Watkins M, Veazey R, Aldovini A. Inhibition of p38 MAPK in combination with ART reduces SIV-induced immune activation and provides additional protection from immune system deterioration. PLoS Pathog 2018; 14:e1007268. [PMID: 30161247 PMCID: PMC6135519 DOI: 10.1371/journal.ppat.1007268] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 09/12/2018] [Accepted: 08/08/2018] [Indexed: 12/12/2022] Open
Abstract
Differences in immune activation were identified as the most significant difference between AIDS-susceptible and resistant species. p38 MAPK, activated in HIV infection, is key to induction of interferon-stimulated genes and cytokine-mediated inflammation and is associated with some of the pathology produced by HIV or SIV infection in AIDS-susceptible primates. As small molecule p38 MAPK inhibitors are being tested in human trials for inflammatory diseases, we evaluated the effects of treating SIV-infected macaques with the p38 MAPK inhibitor PH-797804 in conjunction with ART. PH-797804 had no side effects, did not impact negatively the antiviral immune response and, used alone, had no significant effect on levels of immune activation and did not reduced the viremia. When administered with ART, it significantly reduced numerous immune activation markers compared to ART alone. CD38+/HLA-DR+ and Ki-67+ T-cell percentages in blood, lymph node and rectal CD4+ and CD8+ T cells, PD-1 expression in CD8+ T cells and plasma levels of IFNα, IFNγ, TNFα, IL-6, IP-10, sCD163 and C-reactive protein were all significantly reduced. Significant preservation of CD4+, CD4+ central memory, CD4+/IL-22+ and CD4+/IL-17+ T-cell percentages and improvement of Th17/Treg ratio in blood and rectal mucosa were also observed. Importantly, the addition of PH-797804 to ART initiated during chronic SIV infection reduced immune activation and restored immune system parameters to the levels observed when ART was initiated on week 1 after infection. After ART interruption, viremia rebounded in a similar fashion in all groups, regardless of when ART was initiated. We concluded that the inhibitor PH-797804 significantly reduced, even if did not normalized, the immune activation parameters evaluated during ART treatment, improved preservation of critical populations of the immune system targeted by SIV, and increased the efficacy of ART treatment initiated in chronic infection to levels similar to those observed when initiated in acute infection but did not affect positively or negatively viral reservoirs. The hallmark of Human Immunodeficiency Virus and Simian Immunodeficiency Virus infection in disease-susceptible species is the progressive decline of the CD4+ T cell population and heightened immune activation, which by itself can contribute to CD4+ T-cell death. The cellular pathway regulated by p38 MAPK, which is activated in HIV and SIV infection, can contribute significantly to immune activation. We tested in SIV-infected macaques a p38 MAPK inhibitor in combination with anti-retroviral therapy. This drug is already being evaluated in humans for treatment of immune activation associated with other diseases. We found that, when combined with antiretroviral therapy, the inhibitor PH-797804 significantly reduced a few parameters of SIV-induced immune activation and improved preservation of critical populations of the immune system targeted by SIV, but did not modulate viral reservoirs. Importantly, the addition of the inhibitor to anti-retroviral therapy during the chronic phase of the infection, which is the time when most HIV-infected individuals initiate treatment, permitted a more significant preservation of the immune system compared to antiretroviral therapy alone that was similar to that observed when anti-retroviral therapy was initiated in the acute phase of the infection, which rarely occurs in HIV infection.
Collapse
Affiliation(s)
- Omkar Chaudhary
- Boston Children’s Hospital, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston MA, United States of America
| | - Vivek Narayan
- Boston Children’s Hospital, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston MA, United States of America
| | - Felipe Lelis
- Boston Children’s Hospital, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston MA, United States of America
| | - Brandon Linz
- Boston Children’s Hospital, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston MA, United States of America
| | - Meagan Watkins
- Tulane National Primate Research Center, Division of Comparative Pathology, Covington LA, United States of America
| | - Ronald Veazey
- Tulane National Primate Research Center, Division of Comparative Pathology, Covington LA, United States of America
| | - Anna Aldovini
- Boston Children’s Hospital, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston MA, United States of America
- * E-mail:
| |
Collapse
|
68
|
Liu Y, Sun JK, Qi X, Chen YM, Li J, Chen SY, Liu H. Expression and Significance of Th17 and Treg Cells in Pulmonary Infections with Gram-Negative Bacteria. Immunol Invest 2018; 46:730-741. [PMID: 28872972 DOI: 10.1080/08820139.2017.1360338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The aim of this study was to investigate the expression and significance of T helper type 17 (Th17) and regulatory T (Treg) cells in severe pulmonary infection with gram-negative bacteria (GNB). The peripheral venous blood (PVB) and bronchoalveolar lavage fluid (BALF) were collected from patients receiving mechanical ventilation in the intensive care unit (ICU) owing to: (1) pulmonary GNB infection (group I) and (2) nonpulmonary infection (group NI). Patients from the two groups were matched based on their Acute Physiology and Chronic Health Evaluation II (APACHE II) scores and were recruited in the same period. The levels of Th17 and Treg cells in the PVB and BALF were measured by flow cytometry. (1) The levels of Th17 and Treg cells in the PVB and BALF of the infection group (I) were significantly higher than those of the noninfection group (NI) (p < 0.01), and the levels decreased significantly after treatment (p < 0.01). (2) The Treg/Th17 cell ratio in the PVB and BALF of group I was significantly lower than those of group NI and after treatment (p < 0.01). (3) The levels of Th17 and Treg cells in the PVB and BALF could not predict the 28-day mortality (p > 0.05). The expression of Th17 and Treg cells was abnormal in patients with severe pulmonary GNB infection. Our data suggest an overactive immune response in the early stages of inflammation, but the levels of Treg and Th17 cells failed to predict the 28-day mortality.
Collapse
Affiliation(s)
- Ying Liu
- a Department of Critical Care Medicine , Nanjing First Hospital, Nanjing Medical University , Nanjing , China
| | - Jia-Kui Sun
- a Department of Critical Care Medicine , Nanjing First Hospital, Nanjing Medical University , Nanjing , China
| | - Xiang Qi
- a Department of Critical Care Medicine , Nanjing First Hospital, Nanjing Medical University , Nanjing , China
| | - Yong-Ming Chen
- a Department of Critical Care Medicine , Nanjing First Hospital, Nanjing Medical University , Nanjing , China
| | - Jing Li
- a Department of Critical Care Medicine , Nanjing First Hospital, Nanjing Medical University , Nanjing , China
| | - Shang-Yu Chen
- a Department of Critical Care Medicine , Nanjing First Hospital, Nanjing Medical University , Nanjing , China
| | - Han Liu
- a Department of Critical Care Medicine , Nanjing First Hospital, Nanjing Medical University , Nanjing , China
| |
Collapse
|
69
|
Huot N, Bosinger SE, Paiardini M, Reeves RK, Müller-Trutwin M. Lymph Node Cellular and Viral Dynamics in Natural Hosts and Impact for HIV Cure Strategies. Front Immunol 2018; 9:780. [PMID: 29725327 PMCID: PMC5916971 DOI: 10.3389/fimmu.2018.00780] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 03/28/2018] [Indexed: 01/03/2023] Open
Abstract
Combined antiretroviral therapies (cARTs) efficiently control HIV replication leading to undetectable viremia and drastic increases in lifespan of people living with HIV. However, cART does not cure HIV infection as virus persists in cellular and anatomical reservoirs, from which the virus generally rebounds soon after cART cessation. One major anatomical reservoir are lymph node (LN) follicles, where HIV persists through replication in follicular helper T cells and is also trapped by follicular dendritic cells. Natural hosts of SIV, such as African green monkeys and sooty mangabeys, generally do not progress to disease although displaying persistently high viremia. Strikingly, these hosts mount a strong control of viral replication in LN follicles shortly after peak viremia that lasts throughout infection. Herein, we discuss the potential interplay between viral control in LNs and the resolution of inflammation, which is characteristic for natural hosts. We furthermore detail the differences that exist between non-pathogenic SIV infection in natural hosts and pathogenic HIV/SIV infection in humans and macaques regarding virus target cells and replication dynamics in LNs. Several mechanisms have been proposed to be implicated in the strong control of viral replication in natural host's LNs, such as NK cell-mediated control, that will be reviewed here, together with lessons and limitations of in vivo cell depletion studies that have been performed in natural hosts. Finally, we discuss the impact that these insights on viral dynamics and host responses in LNs of natural hosts have for the development of strategies toward HIV cure.
Collapse
Affiliation(s)
- Nicolas Huot
- HIV Inflammation and Persistence Unit, Institut Pasteur, Paris, France.,Vaccine Research Institute, Créteil, France
| | - Steven E Bosinger
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes Nonhuman Primate Genomics Core, Yerkes National Primate Research Center, Atlanta, GA, United States
| | - Mirko Paiardini
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA, United States
| | - R Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, United States.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Michaela Müller-Trutwin
- HIV Inflammation and Persistence Unit, Institut Pasteur, Paris, France.,Vaccine Research Institute, Créteil, France
| |
Collapse
|
70
|
Cecchinato V, Uguccioni M. Insight on the regulation of chemokine activities. J Leukoc Biol 2018; 104:295-300. [PMID: 29668065 DOI: 10.1002/jlb.3mr0118-014r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/09/2018] [Accepted: 03/09/2018] [Indexed: 01/06/2023] Open
Abstract
The activity of chemokines is regulated by several mechanisms that control the final cellular response. The present review discusses the complexity of the regulation of the chemokine system, and the novel findings describing how in persistent infections, the expression of chemokine receptors on the surface of T cells does not correlate with their homing potential. Thanks to the latest advances in our comprehension of the chemokine system, novel approaches targeting chemokines, chemokine receptors, or protein of their signaling pathway should be considered in order to achieve a personalized therapy.
Collapse
Affiliation(s)
- Valentina Cecchinato
- Laboratory of "Chemokines in Immunity", Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Mariagrazia Uguccioni
- Laboratory of "Chemokines in Immunity", Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| |
Collapse
|
71
|
Wetzel KS, Yi Y, Yadav A, Bauer AM, Bello EA, Romero DC, Bibollet-Ruche F, Hahn BH, Paiardini M, Silvestri G, Peeters M, Collman RG. Loss of CXCR6 coreceptor usage characterizes pathogenic lentiviruses. PLoS Pathog 2018; 14:e1007003. [PMID: 29659623 PMCID: PMC5919676 DOI: 10.1371/journal.ppat.1007003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 04/26/2018] [Accepted: 04/02/2018] [Indexed: 11/18/2022] Open
Abstract
Pandemic HIV-1 originated from the cross-species transmission of SIVcpz, which infects chimpanzees, while SIVcpz itself emerged following the cross-species transmission and recombination of monkey SIVs, with env contributed by the SIVgsn/mus/mon lineage that infects greater spot-nosed, mustached and mona monkeys. SIVcpz and HIV-1 are pathogenic in their respective hosts, while the phenotype of their SIVgsn/mus/mon ancestors is unknown. However, two well-studied SIV infected natural hosts, sooty mangabeys (SMs) and African green monkeys (AGMs), typically remain healthy despite high viral loads; these species express low levels of the canonical coreceptor CCR5, and recent work shows that CXCR6 is a major coreceptor for SIV in these hosts. It is not known what coreceptors were used by the precursors of SIVcpz, whether coreceptor use changed during emergence of the SIVcpz/HIV-1 lineage, and what T cell subsets express CXCR6 in natural hosts. Using species-matched coreceptors and CD4, we show here that SIVcpz uses only CCR5 for entry and, like HIV-1, cannot use CXCR6. In contrast, SIVmus efficiently uses both CXCR6 and CCR5. Coreceptor selectivity was determined by Env, with CXCR6 use abrogated by Pro326 in the V3 crown, which is absent in monkey SIVs but highly conserved in SIVcpz/HIV-1. To characterize which cells express CXCR6, we generated a novel antibody that recognizes CXCR6 of multiple primate species. Testing lymphocytes from SM, the best-studied natural host, we found that CXCR6 is restricted to CD4+ effector memory cells, and is expressed by a sub-population distinct from those expressing CCR5. Thus, efficient CXCR6 use, previously identified in SM and AGM infection, also characterizes a member of the SIV lineage that gave rise to SIVcpz/HIV-1. Loss of CXCR6 usage by SIVcpz may have altered its cell tropism, shifting virus from CXCR6-expressing cells that may support replication without disrupting immune function or homeostasis, towards CCR5-expressing cells with pathogenic consequences.
Collapse
Affiliation(s)
- Katherine S. Wetzel
- Departments of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Yanjie Yi
- Departments of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Anjana Yadav
- Departments of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Anya M. Bauer
- Departments of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Ezekiel A. Bello
- Departments of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Dino C. Romero
- Departments of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Frederic Bibollet-Ruche
- Departments of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Beatrice H. Hahn
- Departments of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Atlanta, GA, United States of America
| | - Guido Silvestri
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Atlanta, GA, United States of America
| | - Martine Peeters
- UMI233-TransVIHMI/INSERM U1175, Institut de Recherche pour le Développement (IRD) and University of Montpellier, Montpellier, France
| | - Ronald G. Collman
- Departments of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
72
|
Chitre AS, Kattah MG, Rosli YY, Pao M, Deswal M, Deeks SG, Hunt PW, Abdel-Mohsen M, Montaner LJ, Kim CC, Ma A, Somsouk M, McCune JM. A20 upregulation during treated HIV disease is associated with intestinal epithelial cell recovery and function. PLoS Pathog 2018; 14:e1006806. [PMID: 29505600 PMCID: PMC5854440 DOI: 10.1371/journal.ppat.1006806] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 03/15/2018] [Accepted: 12/13/2017] [Indexed: 02/06/2023] Open
Abstract
TRIAL REGISTRATION ClinicalTrials.gov Clinical Trial NCT00594880.
Collapse
Affiliation(s)
- Avantika S. Chitre
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Michael G. Kattah
- Division of Gastroenterology, University of California, San Francisco, San Francisco, CA, United States of America
| | - Yenny Y. Rosli
- Division of Gastroenterology, University of California, San Francisco, San Francisco, CA, United States of America
| | - Montha Pao
- Division of HIV/AIDS, University of California, San Francisco, San Francisco, CA, United States of America
| | - Monika Deswal
- Division of HIV/AIDS, University of California, San Francisco, San Francisco, CA, United States of America
| | - Steven G. Deeks
- Division of HIV/AIDS, University of California, San Francisco, San Francisco, CA, United States of America
| | - Peter W. Hunt
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | | | - Luis J. Montaner
- The Wistar Institute, Philadelphia, PA, United States of America
| | - Charles C. Kim
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Averil Ma
- Division of Gastroenterology, University of California, San Francisco, San Francisco, CA, United States of America
| | - Ma Somsouk
- Division of Gastroenterology, University of California, San Francisco, San Francisco, CA, United States of America
| | - Joseph M. McCune
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| |
Collapse
|
73
|
Pastor L, Urrea V, Carrillo J, Parker E, Fuente-Soro L, Jairoce C, Mandomando I, Naniche D, Blanco J. Dynamics of CD4 and CD8 T-Cell Subsets and Inflammatory Biomarkers during Early and Chronic HIV Infection in Mozambican Adults. Front Immunol 2018; 8:1925. [PMID: 29354131 PMCID: PMC5760549 DOI: 10.3389/fimmu.2017.01925] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/15/2017] [Indexed: 12/13/2022] Open
Abstract
During primary HIV infection (PHI), there is a striking cascade response of inflammatory cytokines and many cells of the immune system show altered frequencies and signs of extensive activation. These changes have been shown to have a relevant role in predicting disease progression; however, the challenges of identifying PHI have resulted in a lack of critical information about the dynamics of early pathogenic events. We studied soluble inflammatory biomarkers and changes in T-cell subsets in individuals at PHI (n = 40), chronic HIV infection (CHI, n = 56), and HIV-uninfected (n = 58) recruited at the Manhiça District Hospital in Mozambique. Plasma levels of 49 biomarkers were determined by Luminex and ELISA. T-cell immunophenotyping was performed by multicolor flow cytometry. Plasma HIV viremia, CD4, and CD8 T cell counts underwent rapid stabilization after PHI. However, several immunological parameters, including Th1-Th17 CD4 T cells and activation or exhaustion of CD8 T cells continued decreasing until more than 9 months postinfection. Importantly, no sign of immunosenescence was observed over the first year of HIV infection. Levels of IP-10, MCP-1, BAFF, sCD14, tumor necrosis factor receptor-2, and TRAIL were significantly overexpressed at the first month of infection and underwent a prompt decrease in the subsequent months while, MIG and CD27 levels began to increase 1 month after infection and remained overexpressed for almost 1 year postinfection. Early levels of soluble biomarkers were significantly associated with subsequently exhausted CD4 T-cells or with CD8 T-cell activation. Despite rapid immune control of virus replication, the stabilization of the T-cell subsets occurs months after viremia and CD4 count plateau, suggesting persistent immune dysfunction and highlighting the potential benefit of early treatment initiation that could limit immunological damage.
Collapse
Affiliation(s)
- Lucía Pastor
- AIDS Research Institute-IrsiCaixa, Hospital Germans Trias i Pujol, Badalona, Spain.,ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain.,Institut Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Universitat Autonoma de Barcelona, Badalona, Spain.,Centro de Investigação em Saúde da Manhiça (CISM), Maputo, Mozambique
| | - Victor Urrea
- AIDS Research Institute-IrsiCaixa, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Jorge Carrillo
- AIDS Research Institute-IrsiCaixa, Hospital Germans Trias i Pujol, Badalona, Spain.,ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Erica Parker
- School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia
| | - Laura Fuente-Soro
- ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde da Manhiça (CISM), Maputo, Mozambique
| | - Chenjerai Jairoce
- Centro de Investigação em Saúde da Manhiça (CISM), Maputo, Mozambique
| | - Inacio Mandomando
- Centro de Investigação em Saúde da Manhiça (CISM), Maputo, Mozambique
| | - Denise Naniche
- ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde da Manhiça (CISM), Maputo, Mozambique
| | - Julià Blanco
- AIDS Research Institute-IrsiCaixa, Hospital Germans Trias i Pujol, Badalona, Spain.,Institut Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Universitat Autonoma de Barcelona, Badalona, Spain.,Universitat de Vic-Universitat Central de Catalunya, Vic, Spain
| |
Collapse
|
74
|
Fernandes SM, Pires AR, Matoso P, Ferreira C, Nunes-Cabaço H, Correia L, Valadas E, Poças J, Pacheco P, Veiga-Fernandes H, Foxall RB, Sousa AE. HIV-2 infection is associated with preserved GALT homeostasis and epithelial integrity despite ongoing mucosal viral replication. Mucosal Immunol 2018; 11:236-248. [PMID: 28513595 DOI: 10.1038/mi.2017.44] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/10/2017] [Indexed: 02/04/2023]
Abstract
The mechanisms that enable preservation of gut mucosal integrity during persistent viral replication and inherent inflammation remain unclear. Here, we investigated, for the first time, gut homeostasis in HIV-2 infection, a naturally occurring form of attenuated HIV disease. We found viral replication in both sigmoid and ileum of asymptomatic HIV-2+ patients (range: 240-851 circulating CD4+T-cells per μl) despite their undetectable viremia, accompanied by interferon-γ-producing CD8 T-cell expansion, irrespective of antiretroviral treatment. Nevertheless, there was no CD4 T-cell depletion, and Foxp3+ and IL-17- or IL-22-producing CD4 T-cell numbers were unaffected. Moreover, IL-22-producing innate lymphoid cells and IL-22-induced antimicrobial peptides and mucins were maintained. In agreement, the epithelium histology was preserved, including tight junction protein zonula occludens (ZO-1) levels. Furthermore, in vitro infection of colon epithelia with primary isolates revealed no HIV-2 impact on ZO-1 expression. Notably, sigmoid transcriptional levels of CCL20 and CCL28 were significantly increased, in direct correlation with GM-CSF, indicating a local response able to enhance CD4 T-cell recruitment. In conclusion, maintenance of mucosal integrity in HIV-2 infection was associated with T-cell recruitment responses, potentially counteracting CD4 T-cell depletion due to HIV-2 replication. These data have unique implications for the design of therapies targeting gut homeostasis in HIV-1 infection and other chronic inflammatory settings.
Collapse
Affiliation(s)
- S M Fernandes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Hospital de Santa Maria, Centro Hospitalar Lisboa Norte-EPE, Lisboa, Portugal
| | - A R Pires
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - P Matoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - C Ferreira
- Hospital de Santa Maria, Centro Hospitalar Lisboa Norte-EPE, Lisboa, Portugal
| | - H Nunes-Cabaço
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - L Correia
- Hospital de Santa Maria, Centro Hospitalar Lisboa Norte-EPE, Lisboa, Portugal
| | - E Valadas
- Hospital de Santa Maria, Centro Hospitalar Lisboa Norte-EPE, Lisboa, Portugal
- Clínica Universitária de Doenças Infecciosas, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - J Poças
- Serviço de Infecciologia, Hospital de S. Bernardo, Setúbal, Portugal
| | - P Pacheco
- Serviço de Infecciologia, Hospital Fernando da Fonseca, Amadora, Portugal
| | - H Veiga-Fernandes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - R B Foxall
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - A E Sousa
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
75
|
Veazey RS, Lackner AA. Nonhuman Primate Models and Understanding the Pathogenesis of HIV Infection and AIDS. ILAR J 2017; 58:160-171. [PMID: 29228218 PMCID: PMC5886333 DOI: 10.1093/ilar/ilx032] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/01/2017] [Accepted: 11/04/2017] [Indexed: 12/16/2022] Open
Abstract
Research using nonhuman primates (NHPs) as models for human immunodeficiency virus (HIV) infection and acquired immunodeficiency syndrome (AIDS) has resulted in tremendous achievements not only in the prevention and treatment of HIV, but also in biomedical research more broadly. Once considered a death sentence, HIV infection is now fairly well controlled with combination antiretroviral treatments, almost all of which were first tested for efficacy and safety in nonhuman primates or other laboratory animals. Research in NHP has led to "dogma changing" discoveries in immunology, infectious disease, and even our own genetics. We now know that many of our genes are retroviral remnants, or developed in response to archaic HIV-like retroviral infections. Early studies involving blood from HIV patients and in experiments in cultured tissues contributed to confusion regarding the cause of AIDS and impeded progress in the development of effective interventions. Research on the many retroviruses of different NHP species have broadened our understanding of human immunology and perhaps even our origins and evolution as a species. In combination with recent advances in molecular biology and computational analytics, research in NHPs has unique potential for discoveries that will directly lead to new cures for old human and animal diseases, including HIV/AIDS.
Collapse
Affiliation(s)
- Ronald S Veazey
- Ronald S. Veazey, DVM, PhD, is chair of the Division of Comparative Pathology at the Tulane National Primate Research Center and professor in the Department of Pathology and Laboratory Medicine at the Tulane University School of Medicine. Dr. Andrew Lackner, DVM, PhD is director of the Tulane National Primate Research Center and professor of the Department of Microbiology and Pathology and Laboratory Medicine at the Tulane University School of Medicine
| | - Andrew A Lackner
- Ronald S. Veazey, DVM, PhD, is chair of the Division of Comparative Pathology at the Tulane National Primate Research Center and professor in the Department of Pathology and Laboratory Medicine at the Tulane University School of Medicine. Dr. Andrew Lackner, DVM, PhD is director of the Tulane National Primate Research Center and professor of the Department of Microbiology and Pathology and Laboratory Medicine at the Tulane University School of Medicine
| |
Collapse
|
76
|
Huang Q, Wang Y, Si C, Zhao D, Wang Y, Duan Y. Interleukin-35 Modulates the Imbalance Between Regulatory T Cells and T Helper 17 Cells in Enterovirus 71-Induced Hand, Foot, and Mouth Disease. J Interferon Cytokine Res 2017; 37:522-530. [PMID: 29172969 DOI: 10.1089/jir.2017.0080] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Interleukin (IL)-35 modulates the imbalance between regulatory T cells (Tregs) and T helper (Th) 17 cells, which played vital roles in the pathogenesis of autoimmune and infectious diseases. However, the role of Tregs/Th17 cell imbalance and the regulatory functions of IL-35 have remained largely unknown in enterovirus 71 (EV71)-induced hand, foot, and mouth disease (HFMD). In this study, a total of 47 HFMD patients (30 with mild HFMD and 17 with severe HFMD) and 13 healthy individuals were enrolled. The frequencies of CD4+CD25+CD127dim/- Tregs and CD4+IL-17+ Th17 cells, as well as IL-35 expression levels, were measured. Cellular proliferation and cytokine production was also determined in purified Tregs following recombinant IL-35 stimulation. An imbalance between Tregs and Th17 cells was observed in children with severe HFMD, which manifested as a reduction in the Tregs population and an elevation in the Th17 population. Serum IL-35 concentrations were also decreased in case of severe HFMD, which correlated with the Tregs:Th17 cell ratios. Recombinant IL-35 stimulation increased the proportion of Tregs, but downregulated that of Th17 cells. Treatment with IL-35 enhanced Tregs suppressive function and IL-35 and IL-10 expression, but reduced IL-22 secretion in both healthy individuals and those with severe HFMD. The Tregs:Th17 cell ratio was increased in the convalescent patients, however, a significant reduction in serum IL-35 was not observed. Our findings indicated that EV71 infection shifted the Tregs:Th17 cell ratio through IL-35 by downregulating inhibitory cytokine production and reducing the cell-to-cell contact inhibition of effector T cells. Regulation of IL-35 as it relates to the Tregs/Th17 balance may play a critical role in the pathogenesis of EV71-associated HFMD.
Collapse
Affiliation(s)
- Qian Huang
- 1 Department of Pediatrics, The First Affiliated Hospital of Xinxiang Medical University , Weihui, China
| | - Yanhua Wang
- 1 Department of Pediatrics, The First Affiliated Hospital of Xinxiang Medical University , Weihui, China
| | - Changyun Si
- 2 Department of Infectious Diseases, The First Affiliated Hospital of Xinxiang Medical University , Weihui, China
| | - De'an Zhao
- 1 Department of Pediatrics, The First Affiliated Hospital of Xinxiang Medical University , Weihui, China
| | - Yanping Wang
- 2 Department of Infectious Diseases, The First Affiliated Hospital of Xinxiang Medical University , Weihui, China
| | - Yuxiu Duan
- 2 Department of Infectious Diseases, The First Affiliated Hospital of Xinxiang Medical University , Weihui, China
| |
Collapse
|
77
|
Kulkarni V, Ruprecht RM. Mucosal IgA Responses: Damaged in Established HIV Infection-Yet, Effective Weapon against HIV Transmission. Front Immunol 2017; 8:1581. [PMID: 29176985 PMCID: PMC5686557 DOI: 10.3389/fimmu.2017.01581] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/02/2017] [Indexed: 12/13/2022] Open
Abstract
HIV infection not only destroys CD4+ T cells but also inflicts serious damage to the B-cell compartment, such as lymphadenopathy, destruction of normal B-cell follicle architecture, polyclonal hypergammaglobulinemia, increased apoptosis of B cells, and irreversible loss of memory B-cell responses with advanced HIV disease. Subepithelial B cells and plasma cells are also affected, which results in loss of mucosal IgG and IgA antibodies. This leaves the mucosal barrier vulnerable to bacterial translocation. The ensuing immune activation in mucosal tissues adds fuel to the fire of local HIV replication. We postulate that compromised mucosal antibody defenses also facilitate superinfection of HIV-positive individuals with new HIV strains. This in turn sets the stage for the generation of circulating recombinant forms of HIV. What can the mucosal B-cell compartment contribute to protect a healthy, uninfected host against mucosal HIV transmission? Here, we discuss proof-of-principle studies we have performed using passive mucosal immunization, i.e., topical administration of preformed anti-HIV monoclonal antibodies (mAbs) as IgG1, dimeric IgA1 (dIgA1), and dIgA2 isotypes, alone or in combination. Our data indicate that mucosally applied anti-HIV envelope mAbs can provide potent protection against mucosal transmission of simian-human immunodeficiency virus. Our review also discusses the induction of mucosal antibody defenses by active vaccination and potential strategies to interrupt the vicious cycle of bacterial translocation, immune activation, and stimulation of HIV replication in individuals with damaged mucosal barriers.
Collapse
Affiliation(s)
- Viraj Kulkarni
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Ruth M Ruprecht
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, United States.,Southwest National Primate Research Center, San Antonio, TX, United States
| |
Collapse
|
78
|
Méndez-Lagares G, Lu D, Merriam D, Baker CA, Villinger F, Van Rompay KKA, McCune JM, Hartigan-O'Connor DJ. IL-21 Therapy Controls Immune Activation and Maintains Antiviral CD8 + T Cell Responses in Acute Simian Immunodeficiency Virus Infection. AIDS Res Hum Retroviruses 2017; 33:S81-S92. [PMID: 29140110 DOI: 10.1089/aid.2017.0160] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) replicate during acute infection in lymphocytes of the gastrointestinal tract, before disseminating systemically. Localized replication and associated loss of gut-resident CD4+ T cells occur regardless of the portal of entry of the virus (e.g., intravenous vs. rectal). Thus, HIV and SIV are tropic for gut tissue, and their pathogenesis requires the special environment of the intestine. T helper 17 (Th17) cells are important contributors to microbial defense in the gut that are vulnerable to HIV infection and whose loss is associated with translocation of microbial products to the systemic circulation, leading to chronic immune activation and disease progression. Interleukin (IL)-21 promotes differentiation and survival of Th17 cells and stimulates CD8+ T cell function. By promoting Th17 cell survival, IL-21 could limit bacterial translocation and immune activation in the setting of acute or rebounding HIV/SIV disease. In this study, we tested the effect of recombinant IL-21-IgFc treatment, given at the time of infection, on SIVmac251 infection. We found that rIL-21-IgFc decreases immune activation and maintains effective antiviral responses by CD8+ T cells in blood, but this maintenance is not associated with lower viral loads. rIL-21-IgFc treatment also did not generally support Th17 cell populations, but Th17 cells remained strongly and independently associated with control of plasma viremia. For example, the single animal exhibiting greatest control over viremia in our study also manifested the highest levels of IL-21 in plasma, Th17 cell maintenance in blood, and Th17 cells in intestinal tissue. These findings provide rationale for further exploration of IL-21 treatment as a support for host CD8+ T cell responses in HIV cure strategies.
Collapse
Affiliation(s)
- Gema Méndez-Lagares
- California National Primate Research Center, University of California, Davis, California
- Department of Medical Microbiology and Immunology, University of California, Davis, California
| | - Ding Lu
- California National Primate Research Center, University of California, Davis, California
- Department of Medical Microbiology and Immunology, University of California, Davis, California
| | - David Merriam
- California National Primate Research Center, University of California, Davis, California
- Department of Medical Microbiology and Immunology, University of California, Davis, California
| | - Christopher A. Baker
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California
| | - François Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana
| | - Koen K. A. Van Rompay
- California National Primate Research Center, University of California, Davis, California
| | - Joseph M. McCune
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California
| | - Dennis J. Hartigan-O'Connor
- California National Primate Research Center, University of California, Davis, California
- Department of Medical Microbiology and Immunology, University of California, Davis, California
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California
| |
Collapse
|
79
|
The Th17 Lineage: From Barrier Surfaces Homeostasis to Autoimmunity, Cancer, and HIV-1 Pathogenesis. Viruses 2017; 9:v9100303. [PMID: 29048384 PMCID: PMC5691654 DOI: 10.3390/v9100303] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022] Open
Abstract
The T helper 17 (Th17) cells represent a subset of CD4+ T-cells with unique effector functions, developmental plasticity, and stem-cell features. Th17 cells bridge innate and adaptive immunity against fungal and bacterial infections at skin and mucosal barrier surfaces. Although Th17 cells have been extensively studied in the context of autoimmunity, their role in various other pathologies is underexplored and remains an area of open investigation. This review summarizes the history of Th17 cell discovery and the current knowledge relative to the beneficial role of Th17 cells in maintaining mucosal immunity homeostasis. We further discuss the concept of Th17 pathogenicity in the context of autoimmunity, cancer, and HIV infection, and we review the most recent discoveries on molecular mechanisms regulating HIV replication/persistence in pathogenic Th17 cells. Finally, we stress the need for novel fundamental research discovery-based Th17-specific therapeutic interventions to treat pathogenic conditions associated with Th17 abnormalities, including HIV infection.
Collapse
|
80
|
d'Ettorre G, Rossi G, Scagnolari C, Andreotti M, Giustini N, Serafino S, Schietroma I, Scheri GC, Fard SN, Trinchieri V, Mastromarino P, Selvaggi C, Scarpona S, Fanello G, Fiocca F, Ceccarelli G, Antonelli G, Brenchley JM, Vullo V. Probiotic supplementation promotes a reduction in T-cell activation, an increase in Th17 frequencies, and a recovery of intestinal epithelium integrity and mitochondrial morphology in ART-treated HIV-1-positive patients. Immun Inflamm Dis 2017; 5:244-260. [PMID: 28474815 PMCID: PMC5569369 DOI: 10.1002/iid3.160] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 01/31/2017] [Accepted: 02/16/2017] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION HIV infection is characterized by a persistent immune activation associated to a compromised gut barrier immunity and alterations in the profile of the fecal flora linked with the progression of inflammatory symptoms. The effects of high concentration multistrain probiotic (Vivomixx®, Viale del Policlinico 155, Rome, Italy in EU; Visbiome®, Dupont, Madison, Wisconsin in USA) on several aspects of intestinal immunity in ART-experienced HIV-1 patients was evaluated. METHODS A sub-study of a longitudinal pilot study was performed in HIV-1 patients who received the probiotic supplement twice a day for 6 months (T6). T-cell activation and CD4+ and CD8+ T-cell subsets expressing IFNγ (Th1, Tc1) or IL-17A (Th17, Tc17) were stained by cytoflorimetric analysis. Histological and immunohistochemical analyses were performed on intestinal biopsies while enterocytes apoptosis index was determined by TUNEL assay. RESULTS A reduction in the frequencies of CD4+ and CD8+ T-cell subsets, expressing CD38+ , HLA-DR+ , or both, and an increase in the percentage of Th17 cell subsets, especially those with central or effector memory phenotype, was recorded in the peripheral blood and in gut-associated lymphoid tissue (GALT) after probiotic intervention. Conversely, Tc1 and Tc17 levels remained substantially unchanged at T6, while Th1 cell subsets increase in the GALT. Probiotic supplementation was also associated to a recovery of the integrity of the gut epithelial barrier, a reduction of both intraepithelial lymphocytes density and enterocyte apoptosis and, an improvement of mitochondrial morphology sustained in part by a modulation of heat shock protein 60. CONCLUSIONS These findings highlight the potential beneficial effects of probiotic supplementation for the reconstitution of physical and immunological integrity of the mucosal intestinal barrier in ART-treated HIV-1-positive patients.
Collapse
Affiliation(s)
- Gabriella d'Ettorre
- Department of Public Health and Infectious DiseasesAzienda Policlinico Umberto I of RomeRomeItaly
| | - Giacomo Rossi
- School of BiosciencesVeterinary Medicine University of CamerinoMatelicaItaly
| | - Carolina Scagnolari
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci BolognettiDepartment of Molecular MedicineSapienza University of RomeRomeItaly
| | - Mauro Andreotti
- Department of Therapeutic Research and Medicines EvaluationItalian Institute of HealthRomeItaly
| | - Noemi Giustini
- Department of Public Health and Infectious DiseasesSapienza University of RomeRomeItaly
| | - Sara Serafino
- Department of Public Health and Infectious DiseasesSapienza University of RomeRomeItaly
| | - Ivan Schietroma
- Department of Public Health and Infectious DiseasesSapienza University of RomeRomeItaly
| | | | - Saeid Najafi Fard
- Department of Public Health and Infectious DiseasesSapienza University of RomeRomeItaly
| | - Vito Trinchieri
- Department of Public Health and Infectious DiseasesSapienza University of RomeRomeItaly
| | - Paola Mastromarino
- Section of MicrobiologyDepartment of Public Health and Infectious DiseasesSapienza University of RomeRomeItaly
| | - Carla Selvaggi
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci BolognettiDepartment of Molecular MedicineSapienza University of RomeRomeItaly
| | - Silvia Scarpona
- School of BiosciencesVeterinary Medicine University of CamerinoMatelicaItaly
| | - Gianfranco Fanello
- Department of Emergency Surgery—Emergency Endoscopic UnitPoliclinico Umberto ISapienza University of RomeRomeItaly
| | - Fausto Fiocca
- Department of Emergency Surgery—Emergency Endoscopic UnitPoliclinico Umberto ISapienza University of RomeRomeItaly
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious DiseasesAzienda Policlinico Umberto I of RomeRomeItaly
| | - Guido Antonelli
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci BolognettiDepartment of Molecular MedicineSapienza University of RomeRomeItaly
| | - Jason M. Brenchley
- Laboratory of Parasitic DiseasesNational Institute of Allergy and Infectious Diseases, NIHBethesdaMarylandUSA
| | - Vincenzo Vullo
- Department of Public Health and Infectious DiseasesSapienza University of RomeRomeItaly
| |
Collapse
|
81
|
Garg H, Joshi A. Host and Viral Factors in HIV-Mediated Bystander Apoptosis. Viruses 2017; 9:v9080237. [PMID: 28829402 PMCID: PMC5579491 DOI: 10.3390/v9080237] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/14/2017] [Accepted: 08/16/2017] [Indexed: 02/07/2023] Open
Abstract
Human immunodeficiency virus (HIV) infections lead to a progressive loss of CD4 T cells primarily via the process of apoptosis. With a limited number of infected cells and vastly disproportionate apoptosis in HIV infected patients, it is believed that apoptosis of uninfected bystander cells plays a significant role in this process. Disease progression in HIV infected individuals is highly variable suggesting that both host and viral factors may influence HIV mediated apoptosis. Amongst the viral factors, the role of Envelope (Env) glycoprotein in bystander apoptosis is well documented. Recent evidence on the variability in apoptosis induction by primary patient derived Envs underscores the role of Env glycoprotein in HIV disease. Amongst the host factors, the role of C-C Chemokine Receptor type 5 (CCR5), a coreceptor for HIV Env, is also becoming increasingly evident. Polymorphisms in the CCR5 gene and promoter affect CCR5 cell surface expression and correlate with both apoptosis and CD4 loss. Finally, chronic immune activation in HIV infections induces multiple defects in the immune system and has recently been shown to accelerate HIV Env mediated CD4 apoptosis. Consequently, those factors that affect CCR5 expression and/or immune activation in turn indirectly regulate HIV mediated apoptosis making this phenomenon both complex and multifactorial. This review explores the complex role of various host and viral factors in determining HIV mediated bystander apoptosis.
Collapse
Affiliation(s)
- Himanshu Garg
- Center of Emphasis in Infectious Diseases, Department of Biomedical Sciences, Texas Tech University Health Sciences Center, 5001 El Paso Dr., El Paso, TX 79905, USA.
| | - Anjali Joshi
- Center of Emphasis in Infectious Diseases, Department of Biomedical Sciences, Texas Tech University Health Sciences Center, 5001 El Paso Dr., El Paso, TX 79905, USA.
| |
Collapse
|
82
|
Modeling aging in HIV infection in nonhuman primates to address an emerging challenge of the post-ART era. Curr Opin Virol 2017; 25:66-75. [PMID: 28803049 DOI: 10.1016/j.coviro.2017.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 12/22/2022]
Abstract
The advent of antiretroviral therapy (ART) has dramatically improved both quality and length of life for subjects infected with human immunodeficiency virus (HIV), delaying or preventing progression to acquired immunodeficiency syndrome (AIDS). However, the virus induces aging-related changes to the immune system which confound treatment. Additionally, the normal physiologic events that occur during aging lead to deficiencies in immunity which not only exacerbate HIV pathogenesis but also trigger a variety of comorbidities. Here, the synergistic linkage between aging and HIV infection is examined in regard to the immunological and pathological mechanisms that drive both senescence and disease progression. The use of NHPs to investigate potential therapeutic strategies to control the deleterious consequences of aging with HIV infection is also reviewed.
Collapse
|
83
|
McGary CS, Alvarez X, Harrington S, Cervasi B, Ryan ES, Iriele RI, Paganini S, Harper J, Easley K, Silvestri G, Ansari AA, Lichterfeld M, Micci L, Paiardini M. The loss of CCR6 + and CD161 + CD4 + T-cell homeostasis contributes to disease progression in SIV-infected rhesus macaques. Mucosal Immunol 2017; 10:1082-1096. [PMID: 28051083 PMCID: PMC5474141 DOI: 10.1038/mi.2016.116] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/10/2016] [Indexed: 02/04/2023]
Abstract
Although previous studies have shown that CD4+ T cells expressing CCR6 and CD161 are depleted from blood during HIV infection, the mechanisms underlying their loss remain unclear. In this study, we investigated how the homeostasis of CCR6+ and CD161+ CD4+ T cells contributes to SIV disease progression and the mechanisms responsible for their loss from circulation. By comparing SIV infection in rhesus macaques (RMs) and natural host sooty mangabeys (SMs), we found that the loss of CCR6+ and CD161+ CD4+ T cells from circulation is a distinguishing feature of progressive SIV infection in RMs. Furthermore, while viral infection critically contributes to the loss of CD161+CCR6-CD4+ T cells, a redistribution of CCR6+CD161- and CCR6+CD161+CD4+ T cells from the blood to the rectal mucosa is a chief mechanism for their loss during SIV infection. Finally, we provide evidence that the accumulation of CCR6+CD4+ T cells in the mucosa is damaging to the host by demonstrating their reduction from this site following initiation of antiretroviral therapy in SIV-infected RMs and their lack of accumulation in SIV-infected SMs. These data emphasize the importance of maintaining CCR6+ and CD161+ CD4+ T-cell homeostasis, particularly in the mucosa, to prevent disease progression during pathogenic HIV/SIV infection.
Collapse
Affiliation(s)
- Colleen S. McGary
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Xavier Alvarez
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA
| | - Sean Harrington
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, MA
| | - Barbara Cervasi
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Emily S. Ryan
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Robin I. Iriele
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Sara Paganini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Justin Harper
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Kirk Easley
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Atlanta, GA
| | - Guido Silvestri
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Aftab A. Ansari
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Mathias Lichterfeld
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, MA
| | - Luca Micci
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
84
|
Cheng L, Yu H, Li G, Li F, Ma J, Li J, Chi L, Zhang L, Su L. Type I interferons suppress viral replication but contribute to T cell depletion and dysfunction during chronic HIV-1 infection. JCI Insight 2017; 2:94366. [PMID: 28614789 PMCID: PMC5470878 DOI: 10.1172/jci.insight.94366] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/10/2017] [Indexed: 12/17/2022] Open
Abstract
The direct link between sustained type I interferon (IFN-I) signaling and HIV-1-induced immunopathogenesis during chronic infection remains unclear. Here we report studies using a monoclonal antibody to block IFN-α/β receptor 1 (IFNAR1) signaling during persistent HIV-1 infection in humanized mice (hu-mice). We discovered that, during chronic HIV-1 infection, IFNAR blockade increased viral replication, which was correlated with elevated T cell activation. Thus, IFN-Is suppress HIV-1 replication during the chronic phase but are not essential for HIV-1-induced aberrant immune activation. Surprisingly, IFNAR blockade rescued both total human T cell and HIV-specific T cell numbers despite elevated HIV-1 replication and immune activation. We showed that IFNAR blockade reduced HIV-1-induced apoptosis of CD4+ T cells. Importantly, IFNAR blockade also rescued the function of human T cells, including HIV-1-specific CD8+ and CD4+ T cells. We conclude that during persistent HIV-1 infection, IFN-Is suppress HIV-1 replication, but contribute to depletion and dysfunction of T cells.
Collapse
Affiliation(s)
- Liang Cheng
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Haisheng Yu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Guangming Li
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Feng Li
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jianping Ma
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jingyun Li
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Liqun Chi
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Liguo Zhang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Lishan Su
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
85
|
Sehrawat S, Rouse BT. Interplay of Regulatory T Cell and Th17 Cells during Infectious Diseases in Humans and Animals. Front Immunol 2017; 8:341. [PMID: 28421070 PMCID: PMC5377923 DOI: 10.3389/fimmu.2017.00341] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/09/2017] [Indexed: 12/14/2022] Open
Abstract
It is now clear that the outcome of an inflammatory process caused by infections depends on the balance of responses by several components of the immune system. Of particular relevance is the interplay between regulatory T cells (Tregs) and CD4+ T cells that produce IL-17 (Th17 cells) during immunoinflammatory events. In addition to discussing studies done in mice to highlight some unresolved issues in the biology of these cells, we emphasize the need to include outbred animals and humans in analyses. Achieving a balance between Treg and Th17 cells responses represents a powerful approach to control events during immunity and immunopathology.
Collapse
Affiliation(s)
- Sharvan Sehrawat
- Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Barry T Rouse
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
86
|
Nixon CC, Mavigner M, Silvestri G, Garcia JV. In Vivo Models of Human Immunodeficiency Virus Persistence and Cure Strategies. J Infect Dis 2017; 215:S142-S151. [PMID: 28520967 PMCID: PMC5410984 DOI: 10.1093/infdis/jiw637] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Current HIV therapy is not curative regardless of how soon after infection it is initiated or how long it is administered, and therapy interruption almost invariably results in robust viral rebound. Human immunodeficiency virus persistence is therefore the major obstacle to a cure for AIDS. The testing and implementation of novel yet unproven approaches to HIV eradication that could compromise the health status of HIV-infected individuals might not be ethically warranted. Therefore, adequate in vitro and in vivo evidence of efficacy is needed to facilitate the clinical implementation of promising strategies for an HIV cure. Animal models of HIV infection have a strong and well-documented history of bridging the gap between laboratory discoveries and eventual clinical implementation. More recently, animal models have been developed and implemented for the in vivo evaluation of novel HIV cure strategies. In this article, we review the recent progress in this rapidly moving area of research, focusing on the two most promising model systems: humanized mice and nonhuman primates.
Collapse
Affiliation(s)
- Christopher C Nixon
- Division of Infectious Diseases, Center for AIDS Research, University of North Carolina at Chapel Hill School of Medicine
| | - Maud Mavigner
- Department of Pediatrics, Emory University School of Medicine, and
| | - Guido Silvestri
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - J Victor Garcia
- Division of Infectious Diseases, Center for AIDS Research, University of North Carolina at Chapel Hill School of Medicine
| |
Collapse
|
87
|
CXCR6-Mediated Simian Immunodeficiency Virus SIVagmSab Entry into Sabaeus African Green Monkey Lymphocytes Implicates Widespread Use of Non-CCR5 Pathways in Natural Host Infections. J Virol 2017; 91:JVI.01626-16. [PMID: 27903799 DOI: 10.1128/jvi.01626-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/22/2016] [Indexed: 12/14/2022] Open
Abstract
African green monkeys (AGM) and sooty mangabeys (SM) are well-studied natural hosts of simian immunodeficiency virus (SIV) that do not progress to AIDS when infected with their species-specific viruses. Natural hosts of SIV express very low levels of the canonical entry coreceptor CCR5, and recent studies have shown that CCR5 is dispensable for SIV infection of SM in vivo and that blocking of CCR5 does not prevent ex vivo infection of peripheral blood mononuclear cells (PBMC) from SM or vervet AGM. In both hosts, CXCR6 is an efficient entry pathway in vitro Here we investigated the use of species-matched CXCR6 and other alternative coreceptors by SIVagmSab, which infects sabaeus AGM. We cloned sabaeus CD4 and 10 candidate coreceptors. Species-matched CXCR6, CCR5, and GPR15 mediated robust entry into transfected cells by pseudotypes carrying SIVagmSab92018ivTF Env, with lower-level entry through GPR1 and APJ. We cloned genetically divergent env genes from the plasma of two wild-infected sabaeus AGM and found similar patterns of coreceptor use. Titration experiments showed that CXCR6 and CCR5 were more efficient than other coreceptors when tested at limiting CD4/coreceptor levels. Finally, blocking of CXCR6 with its ligand CXCL16 significantly inhibited SIVagmSab replication in sabaeus PBMC and had a greater impact than did the CCR5 blocker maraviroc, confirming the use of CXCR6 in primary lymphocyte infection. These data suggest a new paradigm for SIV infection of natural host species, whereby a shared outcome of virus-host coevolution is the use of CXCR6 or other alternative coreceptors for entry, which may direct SIV toward CD4+ T cell subsets and anatomical sites that support viral replication without disrupting immune homeostasis and function. IMPORTANCE Natural hosts of SIV do not progress to AIDS, in stark contrast to pathogenic human immunodeficiency virus type 1 (HIV-1)-human and SIVmac-macaque infections. Identifying how natural hosts avoid immunodeficiency can elucidate key mechanisms of pathogenesis. It is known that despite high viral loads, natural hosts have a low frequency of CD4+ cells expressing the SIV coreceptor CCR5. In this study, we demonstrate the efficient use of the coreceptor CXCR6 by SIVagmSab to infect sabaeus African green monkey lymphocytes. In conjunction with studies of SIVsmm, which infects sooty mangabeys, and SIVagmVer, which infects vervet monkeys, our data suggest a unifying model whereby in natural hosts, in which the CCR5 expression level is low, the use of CXCR6 or other coreceptors to mediate infection may target SIV toward distinct cell populations that are able to support high-level viral replication without causing a loss of CD4+ T cell homeostasis and lymphoid tissue damage that lead to AIDS in HIV-1 and SIVmac infections.
Collapse
|
88
|
Influence of Plasma Cell Niche Factors on the Recruitment and Maintenance of IRF4hi Plasma Cells and Plasmablasts in Vaccinated, Simian Immunodeficiency Virus-Infected Rhesus Macaques with Low and High Viremia. J Virol 2017; 91:JVI.01727-16. [PMID: 27928009 DOI: 10.1128/jvi.01727-16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/29/2016] [Indexed: 12/30/2022] Open
Abstract
In a recent study, we found that protection following simian immunodeficiency virus (SIV) exposure correlated with rectal plasma cell frequency in vaccinated female rhesus macaques. We sought to determine if the same macaques maintained high mucosal plasma cell frequencies postinfection and if this translated to reduced viremia. Although delayed SIV acquisition did not predict subsequent viral control, alterations existed in the distribution of plasma cells and plasmablasts between macaques that exhibited high or low viremia. Flow cytometric analysis of cells from rectal biopsy specimens, bone marrow, and mesenteric lymph nodes of vaccinated infected, unvaccinated infected, and uninfected macaques identified two main IRF4hi subsets of interest: CD138+ plasma cells, and CD138- plasmablasts. In rectal tissue, plasma cell frequency positively correlated with plasma viremia and unvaccinated macaques had increased plasma cells and plasmablasts compared to vaccinated animals. Likewise, plasmablast frequency in the mesenteric lymph node correlated with viremia. However, in bone marrow, plasmablast frequency negatively correlated with viremia. Accordingly, low-viremic macaques had a higher frequency of both bone marrow IRF4hi subsets than did animals with high viremia. Significant reciprocal relationships between rectal and bone marrow plasmablasts suggested that efficient trafficking to the bone marrow as opposed to the rectal mucosa was linked to viral control. mRNA expression analysis of proteins involved in establishment of plasma cell niches in sorted bone marrow and rectal cell populations further supported this model and revealed differential mRNA expression patterns in these tissues. IMPORTANCE As key antibody producers, plasma cells and plasmablasts are critical components of vaccine-induced immunity to human immunodeficiency virus type 1 (HIV-1) in humans and SIV in the macaque model; however, few have attempted to examine the role of these cells in viral suppression postinfection. Our results suggest that plasmablast trafficking to and retention in the bone marrow play a previously unappreciated role in viral control and contrast the potential contribution of mucosal plasma cells to mediate protection at sites of infection with that of bone marrow plasmablasts and plasma cells to control viremia during chronic infection. Manipulation of niche factors influencing the distribution and maintenance of these critical antibody-secreting cells may serve as potential therapeutic targets to enhance antiviral responses postvaccination and postinfection.
Collapse
|
89
|
Gosselin A, Wiche Salinas TR, Planas D, Wacleche VS, Zhang Y, Fromentin R, Chomont N, Cohen ÉA, Shacklett B, Mehraj V, Ghali MP, Routy JP, Ancuta P. HIV persists in CCR6+CD4+ T cells from colon and blood during antiretroviral therapy. AIDS 2017; 31:35-48. [PMID: 27835617 PMCID: PMC5131694 DOI: 10.1097/qad.0000000000001309] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/07/2016] [Accepted: 10/18/2016] [Indexed: 12/19/2022]
Abstract
OBJECTIVES The objective of this article is to investigate the contribution of colon and blood CD4 T-cell subsets expressing the chemokine receptor CCR6 to HIV persistence during antiretroviral therapy. DESIGN Matched sigmoid biopsies and blood samples (n = 13) as well as leukapheresis (n = 20) were collected from chronically HIV-infected individuals receiving antiretroviral therapy. Subsets of CD4 T cells with distinct differentiation/polarization profiles were identified using surface markers as follows: memory (TM, CD45RA), central memory (TCM; CD45RACCR7), effector (TEM/TM; CD45RACCR7), Th17 (CCR6CCR4), Th1Th17 (CCR6CXCR3), Th1 (CCR6CXCR3), and Th2 (CCR6CCR4). METHODS We used polychromatic flow cytometry for cell sorting, nested real-time PCR for HIV DNA quantification, ELISA and flow cytometry for HIV p24 quantification. HIV reactivation was induced by TCR triggering in the presence/absence of all-trans retinoic acid. RESULTS Compared with blood, the frequency of CCR6 TM was higher in the colon. In both colon and blood compartments, CCR6 TM were significantly enriched in HIV DNA when compared with their CCR6 counterparts (n = 13). In blood, integrated HIV DNA levels were significantly enriched in CCR6 versus CCR6 TCM of four of five individuals and CCR6 versus CCR6 TEM of three of five individuals. Among blood TCM, Th17 and Th1Th17 contributed the most to the pool of cells harboring integrated HIV DNA despite their reduced frequency compared with Th2, which were infected the least. HIV reactivation was induced by TCR triggering and/or retinoic acid exposure at higher levels in CCR6 versus CCR6 TM, TCM, and TEM. CONCLUSION CCR6 is a marker for colon and blood CD4 T cells enriched for replication-competent HIV DNA. Novel eradication strategies should target HIV persistence in CCR6CD4 T cells from various anatomic sites.
Collapse
Affiliation(s)
| | - Tomas Raul Wiche Salinas
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Delphine Planas
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Vanessa S. Wacleche
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Yuwei Zhang
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | | | - Nicolas Chomont
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| | - Éric A. Cohen
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
- Institut de Recherche Clinique de Montréal, Montréal, Québec, Canada
| | | | - Vikram Mehraj
- Chronic Viral Illness Service and Research Institute
| | | | - Jean-Pierre Routy
- Chronic Viral Illness Service and Research Institute
- Division of Hematology, McGill University Health Centre, Montreal, Québec, Canada
| | - Petronela Ancuta
- CHUM-Research Centre
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal
| |
Collapse
|
90
|
Presti R, Pantaleo G. The Immunopathogenesis of HIV-1 Infection. Infect Dis (Lond) 2017. [DOI: 10.1016/b978-0-7020-6285-8.00092-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
91
|
Cheng L, Ma J, Li J, Li D, Li G, Li F, Zhang Q, Yu H, Yasui F, Ye C, Tsao LC, Hu Z, Su L, Zhang L. Blocking type I interferon signaling enhances T cell recovery and reduces HIV-1 reservoirs. J Clin Invest 2016; 127:269-279. [PMID: 27941247 DOI: 10.1172/jci90745] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 10/27/2016] [Indexed: 12/27/2022] Open
Abstract
Despite the efficient suppression of HIV-1 replication that can be achieved with combined antiretroviral therapy (cART), low levels of type I interferon (IFN-I) signaling persist in some individuals. This sustained signaling may impede immune recovery and foster viral persistence. Here we report studies using a monoclonal antibody to block IFN-α/β receptor (IFNAR) signaling in humanized mice (hu-mice) that were persistently infected with HIV-1. We discovered that effective cART restored the number of human immune cells in HIV-1-infected hu-mice but did not rescue their immune hyperactivation and dysfunction. IFNAR blockade fully reversed HIV-1-induced immune hyperactivation and rescued anti-HIV-1 immune responses in T cells from HIV-1-infected hu-mice. Finally, we found that IFNAR blockade in the presence of cART reduced the size of HIV-1 reservoirs in lymphoid tissues and delayed HIV-1 rebound after cART cessation in the HIV-1-infected hu-mice. We conclude that low levels of IFN-I signaling contribute to HIV-1-associated immune dysfunction and foster HIV-1 persistence in cART-treated hosts. Our results suggest that blocking IFNAR may provide a potential strategy to enhance immune recovery and reduce HIV-1 reservoirs in individuals with sustained elevations in IFN-I signaling during suppressive cART.
Collapse
|
92
|
Lu X, Li Z, Li Q, Jiao Y, Ji Y, Zhang H, Liu Z, Li W, Wu H. Preferential loss of gut-homing α4β7 CD4 + T cells and their circulating functional subsets in acute HIV-1 infection. Cell Mol Immunol 2016; 13:776-784. [PMID: 26277899 PMCID: PMC5101442 DOI: 10.1038/cmi.2015.60] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 01/10/2023] Open
Abstract
Preferential infection and depletion of gut-homing α4β7 CD4+ T cells in the blood are observed in chronic HIV/SIV infection. The dynamic change in gut-homing α4β7 CD4+ T cells and their functional subsets during the acute stages of HIV-1 infection are less documented. Therefore, we conducted a cohort study to investigate whether acute HIV-1 infection induced abnormalities in gut-homing α4β7 CD4+ T cells and their functional subsets. We examined the frequency, absolute number, and functionality of gut-homing α4β7 CD4+ T cells in 26 acute HIV-1-infected patients compared with 20 healthy individuals. We found that circulating gut-homing α4β7 CD4+ T cells were preferentially depleted during acute HIV-1 infection and were positively correlated with absolute CD4+ T-cell count in blood. Notably, Th17 and Th1 cell subsets of gut-homing CD4+ T cells were also decreased, which resulted in an imbalance of T helper cells (Th1):regulatory T cells (Treg) and Treg:Th17 ratios. Gut-homing Th17 and Th1 cells were also positively correlated with the absolute number of total CD4+ T cells and gut-homing CD4+ T cells. The gut-homing Treg:Th17 ratio was inversely correlated with the CD4+ T-cell count. Taken together, the analyses of our acute HIV-1 cohort demonstrate that gut-homing α4β7 CD4+ T cells and their functional subsets were profoundly depleted during acute HIV-1 infection, which may have resulted in the persistent loss of circulating CD4+ T cells and an imbalance of Th1:Treg and Treg:Th17 ratios and contribute to HIV-1 disease pathogenesis.
Collapse
Affiliation(s)
- Xiaofan Lu
- STD/HIV Research Laboratory, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing 100069, People's Republic of China
| | - Zhen Li
- STD/HIV Research Laboratory, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing 100069, People's Republic of China
| | - Qunhui Li
- Center for Infectious Diseases, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
| | - Yanmei Jiao
- STD/HIV Research Laboratory, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing 100069, People's Republic of China
| | - Yunxia Ji
- STD/HIV Research Laboratory, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing 100069, People's Republic of China
| | - Hongwei Zhang
- Center for Infectious Diseases, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
| | - Zhuoming Liu
- Case Comprehensive Cancer Center and Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Wei Li
- STD/HIV Research Laboratory, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing 100069, People's Republic of China
- Center for Infectious Diseases, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
| | - Hao Wu
- Center for Infectious Diseases, Beijing You-An Hospital, Capital Medical University, Beijing 100069, People's Republic of China
| |
Collapse
|
93
|
Immune activation in HIV infection: what can the natural hosts of simian immunodeficiency virus teach us? Curr Opin HIV AIDS 2016; 11:201-8. [PMID: 26845673 DOI: 10.1097/coh.0000000000000238] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW The review summarizes studies in natural hosts, with a particular focus on the control of immune activation and new insights into viral reservoirs. We discuss why these findings are relevant for HIV research today. RECENT FINDINGS AIDS resistance in natural hosts is characterized by a rapid control of inflammatory processes in response to simian immunodeficiency virus infection despite persistent viremia. Although CD4 T cells are dramatically depleted in the intestine in primary infection, interleukin 17-producing T helper cells (Th17) are preserved and natural hosts lack microbial translocation. Thus, viral replication in the gut is not sufficient to explain mucosal damage, but additional factors are necessary. Natural hosts also display a lower infection rate of stem-cell memory, central memory and follicular helper T cells. The follicles are characterized by a lack of viral trapping and the viral replication in secondary lymphoid organs is rapidly controlled. Hence, the healthy status of natural hosts is associated with preserved lymphoid environments. SUMMARY Understanding the underlying mechanisms of preservation of Th17 and of the low contribution of stem-cell memory, central memory and follicular helper T cells to viral reservoirs could benefit the search for preventive and curative approaches of HIV. Altogether, the complementarity of the model helps to identify strategies aiming at restoring full capacity of the immune system and decreasing the size of the viral reservoirs.
Collapse
|
94
|
CCR6(-) regulatory T cells blunt the restoration of gut Th17 cells along the CCR6-CCL20 axis in treated HIV-1-infected individuals. Mucosal Immunol 2016; 9:1137-50. [PMID: 26883727 DOI: 10.1038/mi.2016.7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/10/2016] [Indexed: 02/07/2023]
Abstract
The gut CD4(+) T cells, particularly the T helper type 17 (Th17) subset, are not completely restored in most HIV-1-infected individuals despite combined antiretroviral therapy, when initiated at the chronic phase of infection. We show here that the CCR6-CCL20 chemotactic axis is altered, with reduced CCL20 production by small intestine epithelial cells in treated HIV-1-infected individuals. This leads to impaired CCR6(+)CD4(+) T-cell homing, particularly Th17 cells, to the small intestine mucosa. In contrast, the frequency of gut FoxP3(+) T regulatory (Treg) cells, specifically the CCR6(-) subset, was increased. The resulting imbalance in the Th17/CCR6(-) Treg ratio and the associated shift from interleukin (IL)-17 to IL-10 and transforming growth factor-β (TGF-β) blunts CCL20 production by enterocytes, perpetuating a negative feedback for the recruitment of CCR6(+)CD4(+) T cells to the small intestine in treated HIV-1-infected individuals.
Collapse
|
95
|
Verstrepen BE, Nieuwenhuis IG, Mooij P, Verschoor EJ, Fagrouch ZC, Kondova I, Boonstra A, Koopman G. Role of microbial translocation in soluble CD14 up-regulation in HIV-, but not in HCV-, infected chimpanzees. J Gen Virol 2016; 97:2599-2607. [PMID: 27534537 DOI: 10.1099/jgv.0.000577] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
During human immunodeficiency virus (HIV) infection, soluble CD14 (sCD14) is up-regulated as a consequence of pathological disruption of the gut epithelial barrier, and subsequent increased microbial translocation. Also in hepatitis C virus (HCV)-infected patients with advanced liver fibrosis, increased levels of sCD14 have been reported. Since the liver plays an important role in clearance of translocated bacterial products, hepatic fibrosis may negatively affect clearance and thus contribute to higher sCD14 levels. Chimpanzees (Pan troglodytes) infected with HCV typically show no signs of liver fibrosis. Here, we have tested the hypothesis that increased levels of sCD14 occur in the absence of hepatic fibrosis or microbial translocation in chimpanzees chronically infected with HCV. sCD14 was up-regulated in both HIV/simian immunodeficiency virus (SIV)- and HCV-infected chimpanzees. In HIV/SIV-infected chimpanzees, intestinal fatty acid-binding protein, a marker for gut perturbation, lipopolysaccharide (LPS)-binding-protein and LPS core antibodies, confirm that sCD14 up-regulation was caused by increased microbial translocation. In HCV-infected chimpanzees, no evidence was found for increased microbial translocation despite up-regulation of sCD14. Additionally, the impact of liver fibrosis on microbial translocation was addressed by direct comparison of chimpanzees with a high HCV load and human patients with advanced fibrosis. These data suggest that only in a small minority of HCV patients, hepatic fibrosis corroborates microbial translocation.
Collapse
Affiliation(s)
- Babs E Verstrepen
- Department of Virology, Biomedical Primate Research Centre, 2280 GH Rijswijk, The Netherlands
| | - Ivonne G Nieuwenhuis
- Department of Virology, Biomedical Primate Research Centre, 2280 GH Rijswijk, The Netherlands
| | - Petra Mooij
- Department of Virology, Biomedical Primate Research Centre, 2280 GH Rijswijk, The Netherlands
| | - Ernst J Verschoor
- Department of Virology, Biomedical Primate Research Centre, 2280 GH Rijswijk, The Netherlands
| | - Zahra C Fagrouch
- Department of Virology, Biomedical Primate Research Centre, 2280 GH Rijswijk, The Netherlands
| | - Ivanela Kondova
- Division of Pathology and Microbiology, Biomedical Primate Research Centre, 2280 GH Rijswijk, The Netherlands
| | - André Boonstra
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Gerrit Koopman
- Department of Virology, Biomedical Primate Research Centre, 2280 GH Rijswijk, The Netherlands
| |
Collapse
|
96
|
Elevated Basal Pre-infection CXCL10 in Plasma and in the Small Intestine after Infection Are Associated with More Rapid HIV/SIV Disease Onset. PLoS Pathog 2016; 12:e1005774. [PMID: 27509048 PMCID: PMC4980058 DOI: 10.1371/journal.ppat.1005774] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/27/2016] [Indexed: 12/02/2022] Open
Abstract
Elevated blood CXCL10/IP-10 levels during primary HIV-1 infection (PHI) were described as an independent marker of rapid disease onset, more robust than peak viremia or CD4 cell nadir. IP-10 enhances the recruitment of CXCR3+ cells, which include major HIV-target cells, raising the question if it promotes the establishment of viral reservoirs. We analyzed data from four cohorts of HIV+ patients, allowing us to study IP-10 levels before infection (Amsterdam cohort), as well as during controlled and uncontrolled viremia (ANRS cohorts). We also addressed IP-10 expression levels with regards to lymphoid tissues (LT) and blood viral reservoirs in patients and non-human primates. Pre-existing elevated IP-10 levels but not sCD63 associated with rapid CD4 T-cell loss upon HIV-1 infection. During PHI, IP-10 levels and to a lesser level IL-18 correlated with cell-associated HIV DNA, while 26 other inflammatory soluble markers did not. IP-10 levels tended to differ between HIV controllers with detectable and undetectable viremia. IP-10 was increased in SIV-exposed aviremic macaques with detectable SIV DNA in tissues. IP-10 mRNA was produced at higher levels in the small intestine than in colon or rectum. Jejunal IP-10+ cells corresponded to numerous small and round CD68neg cells as well as to macrophages. Blood IP-10 response negatively correlated with RORC (Th17 marker) gene expression in the small intestine. CXCR3 expression was higher on memory CD4+ T cells than any other immune cells. CD4 T cells from chronically infected animals expressed extremely high levels of intra-cellular CXCR3 suggesting internalization after ligand recognition. Elevated systemic IP-10 levels before infection associated with rapid disease progression. Systemic IP-10 during PHI correlated with HIV DNA. IP-10 production was regionalized in the intestine during early SIV infection and CD68+ and CD68neg haematopoietic cells in the small intestine appeared to be the major source of IP-10. Chronic immune activation is a hallmark of HIV infection and contributes in multiple ways to HIV persistence. Here, we gained insights on the association between a pro-inflammatory chemokine, CXCL10/IP-10 and HIV infection in four cohorts of HIV+ individuals, studied at distinct stages of infection (before, primary and chronic stage with spontaneous- and treatment-controlled infection). We further analyzed pathogenic and non-pathogenic SIV infections to address IP-10 levels and the presence of infected cells in tissues (lymph nodes, small and large intestine). We found that elevated systemic IP-10 levels before HIV-1 infection associate with a more rapid disease progression. During primary infection, IP-10 in blood strongly correlated with the amount of infected cells in blood. The animal model showed that IP-10 expression was regionalized in the intestine and highest in the small intestine. Studies of aviremic animals suggest that high IP-10 is indicative of viral replication in lymphoid tissues. Haematopoietic cells rather than epithelial/endothelial cells mainly contributed to the IP-10 production in small intestine (jejunum). The receptor of IP-10 was highly expressed on memory CD4+ T cells, i.e. major target cells for the virus. This study contributes to our understanding of the establishment of HIV reservoirs and why IP-10 associates with HIV/AIDS.
Collapse
|
97
|
Kupz A, Zedler U, Stäber M, Kaufmann SHE. A Mouse Model of Latent Tuberculosis Infection to Study Intervention Strategies to Prevent Reactivation. PLoS One 2016; 11:e0158849. [PMID: 27391012 PMCID: PMC4938611 DOI: 10.1371/journal.pone.0158849] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/22/2016] [Indexed: 02/07/2023] Open
Abstract
Infection with Mycobacterium tuberculosis (Mtb) is the leading cause of death in human immunodeficiency virus (HIV)+ individuals, particularly in Sub-Saharan Africa. Management of this deadly co-infection is a significant global health challenge that is exacerbated by the lack of efficient vaccines against both Mtb and HIV, as well as the lack of reliable and robust animal models for Mtb/HIV co-infection. Here we describe a tractable and reproducible mouse model to study the reactivation dynamics of latent Mtb infection following the loss of CD4+ T cells as it occurs in HIV-co-infected individuals. Whereas intradermally (i.d.) infected C57BL/6 mice contained Mtb within the local draining lymph nodes, depletion of CD4+ cells led to progressive systemic spread of the bacteria and induction of lung pathology. To interrogate whether reactivation of Mtb after CD4+ T cell depletion can be reversed, we employed interleukin (IL)-2/anti-IL-2 complex-mediated cell boost approaches. Although populations of non-CD4 lymphocytes, such as CD8+ memory T cells, natural killer (NK) cells and double-negative (DN) T cells significantly expanded after IL-2/anti-IL-2 complex treatment, progressive development of bacteremia and pathologic lung alterations could not be prevented. These data suggest that the failure to reverse Mtb reactivation is likely not due to anergy of the expanded cell subsets and rather indicates a limited potential for IL-2-complex-based therapies in the management of Mtb/HIV co-infection.
Collapse
Affiliation(s)
- Andreas Kupz
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- * E-mail: (SHEK); (AK)
| | - Ulrike Zedler
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Manuela Stäber
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Stefan H. E. Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
- * E-mail: (SHEK); (AK)
| |
Collapse
|
98
|
The well-tempered SIV infection: Pathogenesis of SIV infection in natural hosts in the wild, with emphasis on virus transmission and early events post-infection that may contribute to protection from disease progression. INFECTION GENETICS AND EVOLUTION 2016; 46:308-323. [PMID: 27394696 DOI: 10.1016/j.meegid.2016.07.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/04/2016] [Accepted: 07/05/2016] [Indexed: 12/25/2022]
Abstract
African NHPs are infected by over 40 different simian immunodeficiency viruses. These viruses have coevolved with their hosts for long periods of time and, unlike HIV in humans, infection does not generally lead to disease progression. Chronic viral replication is maintained for the natural lifespan of the host, without loss of overall immune function. Lack of disease progression is not correlated with transmission, as SIV infection is highly prevalent in many African NHP species in the wild. The exact mechanisms by which these natural hosts of SIV avoid disease progression are still unclear, but a number of factors might play a role, including: (i) avoidance of microbial translocation from the gut lumen by preventing or repairing damage to the gut epithelium; (ii) control of immune activation and apoptosis following infection; (iii) establishment of an anti-inflammatory response that resolves chronic inflammation; (iv) maintenance of homeostasis of various immune cell populations, including NK cells, monocytes/macrophages, dendritic cells, Tregs, Th17 T-cells, and γδ T-cells; (v) restriction of CCR5 availability at mucosal sites; (vi) preservation of T-cell function associated with down-regulation of CD4 receptor. Some of these mechanisms might also be involved in protection of natural hosts from mother-to-infant SIV transmission during breastfeeding. The difficulty of performing invasive studies in the wild has prohibited investigation of the exact events surrounding transmission in natural hosts. Increased understanding of the mechanisms of SIV transmission in natural hosts, and of the early events post-transmission which may contribute to avoidance of disease progression, along with better comprehension of the factors involved in protection from SIV breastfeeding transmission in the natural hosts, could prove invaluable for the development of new prevention strategies for HIV.
Collapse
|
99
|
Depincé-Berger AE, Vergnon-Miszczycha D, Girard A, Frésard A, Botelho-Nevers E, Lambert C, Del Tedesco E, Genin C, Pozzetto B, Lucht F, Roblin X, Bourlet T, Paul S. Major influence of CD4 count at the initiation of cART on viral and immunological reservoir constitution in HIV-1 infected patients. Retrovirology 2016; 13:44. [PMID: 27363286 PMCID: PMC4929778 DOI: 10.1186/s12977-016-0278-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/20/2016] [Indexed: 02/01/2023] Open
Abstract
Background A persistent immune activation is observed in gut during HIV-1 infection, which is not completely reversed by a combined antiretroviral therapy (cART). The impact of the time of cART initiation may highly influence the size of the viral reservoir and the ratio of CD4+/CD8+ T cells in the gut. In this study, we analyzed the characteristics of HIV rectal reservoir of long-term treated patients, regarding their blood CD4+ T cells count at the time of cART initiation. Results Twenty-four consenting men were enrolled: 9 exhibiting a CD4+ T cells count >350/mm3 (“high-level CD4 group”) and 15 < 350/mm3 (“low-level CD4 group”) in blood, at the start of cART. An immunophenotypical analysis of T and B cells subpopulations was performed in blood and rectal biopsies. HIV cell-associated DNA loads and qualitative intra-cellular RNA were determined in both compartments. The ratio of CD4+/CD8+ T cells was significantly decreased in the blood but not in the rectum of the “low-level CD4 group” of patients. The alteration in β7+ CD4+ T cells homing was higher in this group and was correlated to a low ratio of CD4+/CD8+ T cells in blood. An initiation of cART in men exhibiting a low-level CD4 count was also associated with an alteration of B cells maturation. HIV blood and gut DNA reservoirs were significantly lower in the “high-level CD4 group” of men. A high HIV DNA level was associated to a detectable intracellular HIV RNA in rectum. Conclusions An early initiation of cART could significantly preserve gut immunity and limit the viral reservoir constitution. Electronic supplementary material The online version of this article (doi:10.1186/s12977-016-0278-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne-Emmanuelle Depincé-Berger
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Laboratoire d'Immunologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Delphine Vergnon-Miszczycha
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Service de Maladies Infectieuses et Tropicales, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Alexandre Girard
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France
| | - Anne Frésard
- Service de Maladies Infectieuses et Tropicales, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Elisabeth Botelho-Nevers
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Service de Maladies Infectieuses et Tropicales, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Claude Lambert
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Laboratoire d'Immunologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Emilie Del Tedesco
- Service d'Hépato-Gastroentérologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Christian Genin
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Laboratoire d'Immunologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Bruno Pozzetto
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Service des Agents Infectieux et d'Hygiène, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Frédéric Lucht
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Service de Maladies Infectieuses et Tropicales, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Xavier Roblin
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Service d'Hépato-Gastroentérologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| | - Thomas Bourlet
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France. .,Service des Agents Infectieux et d'Hygiène, Centre Hospitalo-Universitaire, Saint-Étienne, France.
| | - Stéphane Paul
- Groupe Immunité des Muqueuses et Agents Pathogènes - GIMAP EA 3064, CIC 1408, Faculté de Médecine J. Lisfranc, Université de Saint-Etienne, Université de Lyon, 42023, Saint-Étienne Cedex 02, France.,Laboratoire d'Immunologie, Centre Hospitalo-Universitaire, Saint-Étienne, France
| |
Collapse
|
100
|
Coexpression Network Analysis of Benign and Malignant Phenotypes of SIV-Infected Sooty Mangabey and Rhesus Macaque. PLoS One 2016; 11:e0156170. [PMID: 27280726 PMCID: PMC4900581 DOI: 10.1371/journal.pone.0156170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/10/2016] [Indexed: 02/02/2023] Open
Abstract
To explore the differences between the extreme SIV infection phenotypes, nonprogression (BEN: benign) to AIDS in sooty mangabeys (SMs) and progression to AIDS (MAL: malignant) in rhesus macaques (RMs), we performed an integrated dual positive-negative connectivity (DPNC) analysis of gene coexpression networks (GCN) based on publicly available big data sets in the GEO database of NCBI. The microarray-based gene expression data sets were generated, respectively, from the peripheral blood of SMs and RMs at several time points of SIV infection. Significant differences of GCN changes in DPNC values were observed in SIV-infected SMs and RMs. There are three groups of enriched genes or pathways (EGPs) that are associated with three SIV infection phenotypes (BEN+, MAL+ and mixed BEN+/MAL+). The MAL+ phenotype in SIV-infected RMs is specifically associated with eight EGPs, including the protein ubiquitin proteasome system, p53, granzyme A, gramzyme B, polo-like kinase, Glucocorticoid receptor, oxidative phosyphorylation and mitochondrial signaling. Mitochondrial (endosymbiotic) dysfunction is solely present in RMs. Specific BEN+ pattern changes in four EGPs are identified in SIV-infected SMs, including the pathways contributing to interferon signaling, BRCA1/DNA damage response, PKR/INF induction and LGALS8. There are three enriched pathways (PRR-activated IRF signaling, RIG1-like receptor and PRR pathway) contributing to the mixed (BEN+/MAL+) phenotypes of SIV infections in RMs and SMs, suggesting that these pathways play a dual role in the host defense against viral infections. Further analysis of Hub genes in these GCNs revealed that the genes LGALS8 and IL-17RA, which positively regulate the barrier function of the gut mucosa and the immune homeostasis with the gut microbiota (exosymbiosis), were significantly differentially expressed in RMs and SMs. Our data suggest that there exists an exo- (dysbiosis of the gut microbiota) and endo- (mitochondrial dysfunction) symbiotic imbalance (EESI) in HIV/SIV infections. Dissecting the mechanisms of the exo-endo symbiotic balance (EESB) that maintains immune homeostasis and the EESI problems in HIV/SIV infections may lead to a better understanding of the pathogenesis of AIDS and the development of novel interventions for the rational control of this disease.
Collapse
|