1
|
Ngo MH, Pankrac J, Ho RCY, Ndashimye E, Pawa R, Ceccacci R, Biru T, Olabode AS, Klein K, Li Y, Kovacs C, Assad R, Jacobson JM, Canaday DH, Tomusange S, Jamiru S, Anok A, Kityamuweesi T, Buule P, Galiwango RM, Reynolds SJ, Quinn TC, Redd AD, Prodger JL, Mann JFS, Arts EJ. Effective and targeted latency reversal in CD4 + T cells from individuals on long term combined antiretroviral therapy initiated during chronic HIV-1 infection. Emerg Microbes Infect 2024; 13:2327371. [PMID: 38444369 DOI: 10.1080/22221751.2024.2327371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
To date, an affordable, effective treatment for an HIV-1 cure remains only a concept with most "latency reversal" agents (LRAs) lacking specificity for the latent HIV-1 reservoir and failing in early clinical trials. We assessed HIV-1 latency reversal using a multivalent HIV-1-derived virus-like particle (HLP) to treat samples from 32 people living with HIV-1 (PLWH) in Uganda, US and Canada who initiated combined antiretroviral therapy (cART) during chronic infection. Even after 5-20 years on stable cART, HLP could target CD4+ T cells harbouring latent HIV-1 reservoir resulting in 100-fold more HIV-1 release into culture supernatant than by common recall antigens, and 1000-fold more than by chemotherapeutic LRAs. HLP induced release of a divergent and replication-competent HIV-1 population from PLWH on cART. These findings suggest HLP provides a targeted approach to reactivate the majority of latent HIV-1 proviruses among individuals infected with HIV-1.
Collapse
Affiliation(s)
- Minh Ha Ngo
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- College of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Joshua Pankrac
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Ryan C Y Ho
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Emmanuel Ndashimye
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Rahul Pawa
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Renata Ceccacci
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Tsigereda Biru
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- Special Immunology Unit and Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Abayomi S Olabode
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Katja Klein
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- Bristol Veterinary School, University of Bristol, Bristol, UK
| | - Yue Li
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Colin Kovacs
- Maple Leaf Medical Clinic and Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Canada
| | - Robert Assad
- Special Immunology Unit and Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jeffrey M Jacobson
- Special Immunology Unit and Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - David H Canaday
- Special Immunology Unit and Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | | | - Aggrey Anok
- Rakai Health Sciences Program, Kalisizo, Uganda
| | | | - Paul Buule
- Rakai Health Sciences Program, Kalisizo, Uganda
| | | | - Steven J Reynolds
- Rakai Health Sciences Program, Kalisizo, Uganda
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Thomas C Quinn
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew D Redd
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jessica L Prodger
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | - Jamie F S Mann
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
- Bristol Veterinary School, University of Bristol, Bristol, UK
| | - Eric J Arts
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| |
Collapse
|
2
|
Cossarini F, Shang J, Krek A, Al-Taie Z, Hou R, Canales-Herrerias P, Tokuyama M, Tankelevich M, Tillowitz A, Jha D, Livanos AE, Leyre L, Uzzan M, Martinez-Delgado G, Taylor MD, Sharma K, Bourgonje AR, Cruz M, Ioannou G, Dawson T, D'Souza D, Kim-Schulze S, Akm A, Aberg JA, Chen BK, Kwon DS, Gnjatic S, Polydorides AD, Cerutti A, Argmann C, Vujkovic-Cvijin I, Suarez-Fariñas M, Petralia F, Faith JJ, Mehandru S. Gastrointestinal germinal center B cell depletion and reduction in IgA + plasma cells in HIV-1 infection. Sci Immunol 2024; 9:eado0090. [PMID: 39454027 DOI: 10.1126/sciimmunol.ado0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 09/25/2024] [Indexed: 10/27/2024]
Abstract
Gastrointestinal (GI) B cells and plasma cells (PCs) are critical to mucosal homeostasis and the host response to HIV-1 infection. Here, high-resolution mapping of human B cells and PCs sampled from the colon and ileum during both viremic and suppressed HIV-1 infection identified a reduction in germinal center (GC) B cells and follicular dendritic cells (FDCs) during HIV-1 viremia. Immunoglobulin A-positive (IgA+) PCs are the major cellular output of intestinal GCs and were significantly reduced during viremic HIV-1 infection. PC-associated transcriptional perturbations, including type I interferon signaling, persisted in antiretroviral therapy (ART)-treated individuals, suggesting ongoing disruption of the intestinal immune milieu during ART. GI humoral immune perturbations were associated with changes in the intestinal microbiome composition and systemic inflammation. These findings highlight a key immune defect in the GI mucosa due to HIV-1 viremia.
Collapse
Affiliation(s)
- Francesca Cossarini
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joan Shang
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Azra Krek
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zainab Al-Taie
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruixue Hou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pablo Canales-Herrerias
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Minami Tokuyama
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Tankelevich
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam Tillowitz
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Divya Jha
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra E Livanos
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Louise Leyre
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mathieu Uzzan
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Gastroenterology Department, Hôpital Henri Mondor, APHP, Créteil, France
| | - Gustavo Martinez-Delgado
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Taylor
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keshav Sharma
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arno R Bourgonje
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Cruz
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giorgio Ioannou
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Travis Dawson
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Darwin D'Souza
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ahmed Akm
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Judith A Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin K Chen
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas S Kwon
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Sacha Gnjatic
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandros D Polydorides
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Cerutti
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Translational Clinical Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ivan Vujkovic-Cvijin
- F. Widjaja IBD Institute, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mayte Suarez-Fariñas
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeremiah J Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
3
|
Cossarini F, Shang J, Krek A, Al-Taie Z, Hou R, Canales-Herrerias P, Tokuyama M, Tankelevich M, Tillowiz A, Jha D, Livanos AE, Leyre L, Uzzan M, Martinez-Delgado G, Taylor MD, Sharma K, Bourgonje AR, Cruz M, Ioannou G, Dawson T, D'Souza D, Kim-Schulze S, Akm A, Aberg JA, Chen BK, Kwon DS, Gnjatic S, Polydorides AD, Cerutti A, Argmann C, Vujkovic-Cvijin I, Suarez-Fariñas M, Petralia F, Faith JJ, Mehandru S. Gastrointestinal germinal center B cell depletion and reduction in IgA + plasma cells in HIV-1 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.590425. [PMID: 38826293 PMCID: PMC11142040 DOI: 10.1101/2024.05.17.590425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Gastrointestinal (GI) B cells and plasma cells (PCs) are critical to mucosal homeostasis and the host response to HIV-1 infection. Here, high resolution mapping of human B cells and PCs sampled from the colon and ileum during both viremic and suppressed HIV-1 infection identified a reduction in germinal center (GC) B cells and follicular dendritic cells (FDCs) during HIV-1 viremia. IgA + PCs are the major cellular output of intestinal GCs and were significantly reduced during viremic HIV-1 infection. PC-associated transcriptional perturbations, including type I interferon signaling, persisted in antiretroviral therapy (ART)-treated individuals, suggesting ongoing disruption of the intestinal immune milieu during ART. GI humoral immune perturbations were associated with changes in the intestinal microbiome composition and systemic inflammation. These findings highlight a key immune defect in the GI mucosa due to HIV-1 viremia. One Sentence Summary Intestinal germinal center B cell reduction in HIV-1 infection linked to reduced IgA + plasma cells and systemic inflammation.
Collapse
|
4
|
Tarasova O, Petrou A, Ivanov SM, Geronikaki A, Poroikov V. Viral Factors in Modulation of Host Immune Response: A Route to Novel Antiviral Agents and New Therapeutic Approaches. Int J Mol Sci 2024; 25:9408. [PMID: 39273355 PMCID: PMC11395507 DOI: 10.3390/ijms25179408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Viruses utilize host cells at all stages of their life cycle, from the transcription of genes and translation of viral proteins to the release of viral copies. The human immune system counteracts viruses through a variety of complex mechanisms, including both innate and adaptive components. Viruses have an ability to evade different components of the immune system and affect them, leading to disruption. This review covers contemporary knowledge about the virus-induced complex interplay of molecular interactions, including regulation of transcription and translation in host cells resulting in the modulation of immune system functions. Thorough investigation of molecular mechanisms and signaling pathways that are involved in modulating of host immune response to viral infections can help to develop novel approaches for antiviral therapy. In this review, we consider new therapeutic approaches for antiviral treatment. Modern therapeutic strategies for the treatment and cure of human immunodeficiency virus (HIV) are considered in detail because HIV is a unique example of a virus that leads to host T lymphocyte deregulation and significant modulation of the host immune response. Furthermore, peculiarities of some promising novel agents for the treatment of various viral infections are described.
Collapse
Affiliation(s)
- Olga Tarasova
- Institute of Biomedical Chemistry, Moscow 119121, Russia
| | - Anthi Petrou
- School of Pharmacy, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | | | - Athina Geronikaki
- School of Pharmacy, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | | |
Collapse
|
5
|
Moretti S, Schietroma I, Sberna G, Maggiorella MT, Sernicola L, Farcomeni S, Giovanetti M, Ciccozzi M, Borsetti A. HIV-1-Host Interaction in Gut-Associated Lymphoid Tissue (GALT): Effects on Local Environment and Comorbidities. Int J Mol Sci 2023; 24:12193. [PMID: 37569570 PMCID: PMC10418605 DOI: 10.3390/ijms241512193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/20/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
HIV-1 replication in the gastrointestinal (GI) tract causes severe CD4+ T-cell depletion and disruption of the protective epithelial barrier in the intestinal mucosa, causing microbial translocation, the main driver of inflammation and immune activation, even in people living with HIV (PLWH) taking antiretroviral drug therapy. The higher levels of HIV DNA in the gut compared to the blood highlight the importance of the gut as a viral reservoir. CD4+ T-cell subsets in the gut differ in phenotypic characteristics and differentiation status from the ones in other tissues or in peripheral blood, and little is still known about the mechanisms by which the persistence of HIV is maintained at this anatomical site. This review aims to describe the interaction with key subsets of CD4+ T cells in the intestinal mucosa targeted by HIV-1 and the role of gut microbiome and its metabolites in HIV-associated systemic inflammation and immune activation that are crucial in the pathogenesis of HIV infection and related comorbidities.
Collapse
Affiliation(s)
- Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00162 Rome, Italy; (S.M.); (I.S.); (G.S.); (M.T.M.); (L.S.); (S.F.)
| | - Ivan Schietroma
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00162 Rome, Italy; (S.M.); (I.S.); (G.S.); (M.T.M.); (L.S.); (S.F.)
| | - Giuseppe Sberna
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00162 Rome, Italy; (S.M.); (I.S.); (G.S.); (M.T.M.); (L.S.); (S.F.)
| | - Maria Teresa Maggiorella
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00162 Rome, Italy; (S.M.); (I.S.); (G.S.); (M.T.M.); (L.S.); (S.F.)
| | - Leonardo Sernicola
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00162 Rome, Italy; (S.M.); (I.S.); (G.S.); (M.T.M.); (L.S.); (S.F.)
| | - Stefania Farcomeni
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00162 Rome, Italy; (S.M.); (I.S.); (G.S.); (M.T.M.); (L.S.); (S.F.)
| | - Marta Giovanetti
- Instituto Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Minas Gerais, Brazil;
- Sciences and Technologies for Sustainable Development and One Health, University Campus Bio-Medico of Rome, 00128 Rome, Italy
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, 00128 Rome, Italy;
| | - Alessandra Borsetti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, 00162 Rome, Italy; (S.M.); (I.S.); (G.S.); (M.T.M.); (L.S.); (S.F.)
| |
Collapse
|
6
|
Association of envelope-specific B-cell differentiation and viral selective pressure signatures in HIV-1 CRF01_AE infection. AIDS 2022; 36:1629-1641. [PMID: 35848590 DOI: 10.1097/qad.0000000000003323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE In HIV type 1 (HIV-1) infection, virus-specific B-cell and neutralizing antibody (NAb) responses are impaired but exert selective pressure on target viral Envelope (Env) resulting in prominent sequence diversification among geographical areas. The basal induction patterns of HIV Env-specific B cells and their interaction with HIV Env awaits clarification. DESIGN We investigated the relationship of Env polymorphisms and Env-specific B-cell responses in treatment-naive HIV-1 CRF01_AE-infected Vietnamese. METHODS Samples of 43 HIV-1 CRF01_AE infection-identified individuals were divided into acute-phase ( n = 12) and chronic-phase ( n = 31) by combined criteria of serological recent-infection assay and clinical parameters. We quantified subcloning-based polymorphic residue site numbers in plasma-derived Env variable region 1-5 (V1-V5)-coding regions within each individual, designating their summation within each region as variant index. Peripheral blood Env gp 140-specific B-cell responses and plasma neutralizing activity of Env pseudoviruses were examined to analyze their relationship with variant index. RESULTS HIV-1 CRF01_AE Env gp140-specific total B-cell and plasma cell (CD19 + IgD - CD27 + CD38 + CD138 + ) responses were determined. In chronic-phase samples, significant correlation of variant index in all Env V1-V5 regions with Env-specific plasma cell responses was shown, and V1-V5 total variant index correlated stronger with Env-specific plasma cell as compared with total Env-specific B-cell responses. Env V5 variant index was significantly higher in chronic-phase cross-neutralizers of V5-polymorphic/VRC01-insensitive CRF01_AE Env. CONCLUSION Results revealed the association between circulating Env-specific plasma cell responses and Env polymorphisms, implicating selective pressure on Env by plasma cell-derived antibodies and conversely suggests that Env-specific B-cell induction alone is insufficient for exerting Env selective pressure in HIV infection.
Collapse
|
7
|
Jones ST, Guo K, Cooper EH, Dillon SM, Wood C, Nguyen DH, Shen G, Barrett BS, Frank DN, Kroehl M, Janoff EN, Kechris K, Wilson CC, Santiago ML. Altered Immunoglobulin Repertoire and Decreased IgA Somatic Hypermutation in the Gut during Chronic HIV-1 Infection. J Virol 2022; 96:e0097622. [PMID: 35938870 PMCID: PMC9472609 DOI: 10.1128/jvi.00976-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/21/2022] [Indexed: 11/27/2022] Open
Abstract
Humoral immune perturbations contribute to pathogenic outcomes in persons with HIV-1 infection (PWH). Gut barrier dysfunction in PWH is associated with microbial translocation and alterations in microbial communities (dysbiosis), and IgA, the most abundant immunoglobulin (Ig) isotype in the gut, is involved in gut homeostasis by interacting with the microbiome. We determined the impact of HIV-1 infection on the antibody repertoire in the gastrointestinal tract by comparing Ig gene utilization and somatic hypermutation (SHM) in colon biopsies from PWH (n = 19) versus age and sex-matched controls (n = 13). We correlated these Ig parameters with clinical, immunological, microbiome and virological data. Gene signatures of enhanced B cell activation were accompanied by skewed frequencies of multiple Ig Variable genes in PWH. PWH showed decreased frequencies of SHM in IgA and possibly IgG, with a substantial loss of highly mutated IgA sequences. The decline in IgA SHM in PWH correlated with gut CD4+ T cell loss and inversely correlated with mucosal inflammation and microbial translocation. Diminished gut IgA SHM in PWH was driven by transversion mutations at A or T deoxynucleotides, suggesting a defect not at the AID/APOBEC3 deamination step but at later stages of IgA SHM. These results expand our understanding of humoral immune perturbations in PWH that could have important implications in understanding mucosal immune defects in individuals with chronic HIV-1 infection. IMPORTANCE The gut is a major site of early HIV-1 replication and pathogenesis. Extensive CD4+ T cell depletion in this compartment results in a compromised epithelial barrier that facilitates the translocation of microbes into the underlying lamina propria and systemic circulation, resulting in chronic immune activation. To date, the consequences of microbial translocation on the mucosal humoral immune response (or vice versa) remains poorly integrated into the panoply of mucosal immune defects in PWH. We utilized next-generation sequencing approaches to profile the Ab repertoire and ascertain frequencies of somatic hypermutation in colon biopsies from antiretroviral therapy-naive PWH versus controls. Our findings identify perturbations in the Ab repertoire of PWH that could contribute to development or maintenance of dysbiosis. Moreover, IgA mutations significantly decreased in PWH and this was associated with adverse clinical outcomes. These data may provide insight into the mechanisms underlying impaired Ab-dependent gut homeostasis during chronic HIV-1 infection.
Collapse
Affiliation(s)
- Sean T. Jones
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kejun Guo
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Emily H. Cooper
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Stephanie M. Dillon
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cheyret Wood
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David H. Nguyen
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Guannan Shen
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Bradley S. Barrett
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Daniel N. Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Miranda Kroehl
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Edward N. Janoff
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA
| | - Katerina Kechris
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cara C. Wilson
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mario L. Santiago
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
8
|
Romero-Martín L, Tarrés-Freixas F, Pedreño-López N, de la Concepción MLR, Cunyat F, Hartigan-O'Connor D, Carrillo J, Mothe B, Blanco J, Ruiz-Riol M, Brander C, Olvera A. T-Follicular-Like CD8 + T Cell Responses in Chronic HIV Infection Are Associated With Virus Control and Antibody Isotype Switching to IgG. Front Immunol 2022; 13:928039. [PMID: 35784304 PMCID: PMC9241491 DOI: 10.3389/fimmu.2022.928039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/16/2022] [Indexed: 01/26/2023] Open
Abstract
T cell responses are considered critical for the in vivo control of HIV, but the contribution of different T cell subsets to this control remains unclear. Using a boosted flow cytometric approach that is able to differentiate CD4+ and CD8+ T cell Th1/Tc1, Th2/Tc2, Th17/Tc17, Treg and Tfh/Tfc-like HIV-specific T cell populations, we identified CD8+ Tfc responses that were related to HIV plasma viral loads and associated with rate of antibody isotype class switching to IgG. This favorable balance towards IgG responses positively correlated with increased virus neutralization, higher avidity of neutralizing antibodies and more potent antibody-dependent cell cytotoxicity (ADCC) in PBMCs from HIV controllers compared to non-controllers. Our results identified the CD8+ Tfc-like T-cell response as a component of effective virus control which could possibly be exploited therapeutically.
Collapse
Affiliation(s)
- Luis Romero-Martín
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- Departament de Biologia Cellular, de Fisiologia i d’Immunologia, Universitat Autonoma de Barcelona, Cerdanyola del Valles, Spain
| | - Ferran Tarrés-Freixas
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Núria Pedreño-López
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Maria L. Rodríguez de la Concepción
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Francesc Cunyat
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Dennis Hartigan-O'Connor
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, United States
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- Fundació Lluita contra la Sida, Infectious Disease Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Marta Ruiz-Riol
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- AELIX Therapeutics, Barcelona, Spain
| | - Alex Olvera
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de salud Carlos III, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| |
Collapse
|
9
|
Marginal Zone B-Cell Populations and Their Regulatory Potential in the Context of HIV and Other Chronic Inflammatory Conditions. Int J Mol Sci 2022; 23:ijms23063372. [PMID: 35328792 PMCID: PMC8949885 DOI: 10.3390/ijms23063372] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Inflammation in the context of Human Immunodeficiency Virus (HIV) establishes early and persists beyond antiretroviral therapy (ART). As such, we have shown excess B-cell activating factor (BAFF) in the blood of HIV-infected progressors, as soon as in the acute phase, and despite successful ART. Excess BAFF was associated with deregulation of the B-cell compartment; notably, with increased frequencies of a population sharing features of both transitional immature (TI) and marginal zone (MZ) B-cells, we termed Marginal Zone precursor-like (MZp). We have reported similar observations with HIV-transgenic mice, Simian Immunodeficiency Virus (SIV)-infected macaques, and more recently, with HIV-infected Beninese commercial sex workers, which suggests that excess BAFF and increased frequencies of MZp B-cells are reliable markers of inflammation in the context of HIV. Importantly, we have recently shown that in healthy individuals, MZps present an important regulatory B-cell (Breg) profile and function. Herein, we wish to review our current knowledge on MZ B-cell populations, especially their Breg status, and that of other B-cell populations sharing similar features. BAFF and its analog A Proliferation-Inducing Ligand (APRIL) are important in shaping the MZ B-cell pool; moreover, the impact that excess BAFF—encountered in the context of HIV and several chronic inflammatory conditions—may exert on MZ B-cell populations, Breg and antibody producing capacities is a threat to the self-integrity of their antibody responses and immune surveillance functions. As such, deregulations of MZ B-cell populations contribute to autoimmune manifestations and the development of MZ lymphomas (MZLs) in the context of HIV and other inflammatory diseases. Therefore, further comprehending the mechanisms regulating MZ B-cell populations and their functions could be beneficial to innovative therapeutic avenues that could be deployed to restore MZ B-cell immune competence in the context of chronic inflammation involving excess BAFF.
Collapse
|
10
|
Kleinman AJ, Pandrea I, Apetrei C. So Pathogenic or So What?-A Brief Overview of SIV Pathogenesis with an Emphasis on Cure Research. Viruses 2022; 14:135. [PMID: 35062339 PMCID: PMC8781889 DOI: 10.3390/v14010135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/10/2021] [Accepted: 12/25/2021] [Indexed: 02/07/2023] Open
Abstract
HIV infection requires lifelong antiretroviral therapy (ART) to control disease progression. Although ART has greatly extended the life expectancy of persons living with HIV (PWH), PWH nonetheless suffer from an increase in AIDS-related and non-AIDS related comorbidities resulting from HIV pathogenesis. Thus, an HIV cure is imperative to improve the quality of life of PWH. In this review, we discuss the origins of various SIV strains utilized in cure and comorbidity research as well as their respective animal species used. We briefly detail the life cycle of HIV and describe the pathogenesis of HIV/SIV and the integral role of chronic immune activation and inflammation on disease progression and comorbidities, with comparisons between pathogenic infections and nonpathogenic infections that occur in natural hosts of SIVs. We further discuss the various HIV cure strategies being explored with an emphasis on immunological therapies and "shock and kill".
Collapse
Affiliation(s)
- Adam J. Kleinman
- Division of Infectious Diseases, DOM, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Ivona Pandrea
- Department of Infectious Diseases and Immunology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Cristian Apetrei
- Division of Infectious Diseases, DOM, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Department of Infectious Diseases and Immunology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| |
Collapse
|
11
|
Ding C, Patel D, Ma Y, Mann JFS, Wu J, Gao Y. Employing Broadly Neutralizing Antibodies as a Human Immunodeficiency Virus Prophylactic & Therapeutic Application. Front Immunol 2021; 12:697683. [PMID: 34354709 PMCID: PMC8329590 DOI: 10.3389/fimmu.2021.697683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/05/2021] [Indexed: 11/18/2022] Open
Abstract
Despite the discovery that the human immunodeficiency virus 1 (HIV-1) is the pathogen of acquired immunodeficiency syndrome (AIDS) in 1983, there is still no effective anti-HIV-1 vaccine. The major obstacle to the development of HIV-1 vaccine is the extreme diversity of viral genome sequences. Nonetheless, a number of broadly neutralizing antibodies (bNAbs) against HIV-1 have been made and identified in this area. Novel strategies based on using these bNAbs as an efficacious preventive and/or therapeutic intervention have been applied in clinical. In this review, we summarize the recent development of bNAbs and its application in HIV-1 acquisition prevention as well as discuss the innovative approaches being used to try to convey protection within individuals at risk and being treated for HIV-1 infection.
Collapse
Affiliation(s)
- Chengchao Ding
- The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Darshit Patel
- Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | - Yunjing Ma
- Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | - Jamie F S Mann
- Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| | - Jianjun Wu
- Department of AIDS Research, Anhui Provincial Center for Disease Control and Prevention, Hefei, China
| | - Yong Gao
- The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.,Department of Microbiology and Immunology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
12
|
Duerr R, Crosse KM, Valero-Jimenez AM, Dittmann M. SARS-CoV-2 Portrayed against HIV: Contrary Viral Strategies in Similar Disguise. Microorganisms 2021; 9:1389. [PMID: 34198973 PMCID: PMC8307803 DOI: 10.3390/microorganisms9071389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 11/16/2022] Open
Abstract
SARS-CoV-2 and HIV are zoonotic viruses that rapidly reached pandemic scale, causing global losses and fear. The COVID-19 and AIDS pandemics ignited massive efforts worldwide to develop antiviral strategies and characterize viral architectures, biological and immunological properties, and clinical outcomes. Although both viruses have a comparable appearance as enveloped viruses with positive-stranded RNA and envelope spikes mediating cellular entry, the entry process, downstream biological and immunological pathways, clinical outcomes, and disease courses are strikingly different. This review provides a systemic comparison of both viruses' structural and functional characteristics, delineating their distinct strategies for efficient spread.
Collapse
Affiliation(s)
- Ralf Duerr
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; (K.M.C.); (A.M.V.-J.); (M.D.)
| | | | | | | |
Collapse
|
13
|
Weber MG, Walters-Laird CJ, Kol A, Santos Rocha C, Hirao LA, Mende A, Balan B, Arredondo J, Elizaldi SR, Iyer SS, Tarantal AF, Dandekar S. Gut germinal center regeneration and enhanced antiviral immunity by mesenchymal stem/stromal cells in SIV infection. JCI Insight 2021; 6:149033. [PMID: 34014838 PMCID: PMC8262475 DOI: 10.1172/jci.insight.149033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022] Open
Abstract
Although antiretroviral therapy suppresses HIV replication, it does not eliminate viral reservoirs or restore damaged lymphoid tissue, posing obstacles to HIV eradication. Using the SIV model of AIDS, we investigated the effect of mesenchymal stem/stromal cell (MSC) infusions on gut mucosal recovery, antiviral immunity, and viral suppression and determined associated molecular/metabolic signatures. MSC administration to SIV-infected macaques resulted in viral reduction and heightened virus-specific responses. Marked clearance of SIV-positive cells from gut mucosal effector sites was correlated with robust regeneration of germinal centers, restoration of follicular B cells and T follicular helper (Tfh) cells, and enhanced antigen presentation by viral trapping within the follicular DC network. Gut transcriptomic analyses showed increased antiviral response mediated by pathways of type I/II IFN signaling, viral restriction factors, innate immunity, and B cell proliferation and provided the molecular signature underlying enhanced host immunity. Metabolic analysis revealed strong correlations between B and Tfh cell activation, anti-SIV antibodies, and IL-7 expression with enriched retinol metabolism, which facilitates gut homing of antigen-activated lymphocytes. We identified potentially new MSC functions in modulating antiviral immunity for enhanced viral clearance predominantly through type I/II IFN signaling and B cell signature, providing a road map for multipronged HIV eradication strategies.
Collapse
Affiliation(s)
| | | | - Amir Kol
- Department of Pathology, Microbiology and Immunology, University of California Davis, Davis, California, USA
| | | | | | - Abigail Mende
- Department of Medical Microbiology and Immunology and
| | - Bipin Balan
- Dipartimento di Scienze Agrarie Alimentari Forestali, Università di Palermo, Viale delle Scienze, Palermo, Italy
| | | | | | - Smita S Iyer
- Department of Pathology, Microbiology and Immunology, University of California Davis, Davis, California, USA.,Center for Immunology and Infectious Diseases.,California National Primate Research Center, and
| | - Alice F Tarantal
- California National Primate Research Center, and.,Departments of Pediatrics and Cell Biology and Human Anatomy, University of California Davis, Davis, California, USA
| | - Satya Dandekar
- Department of Medical Microbiology and Immunology and.,California National Primate Research Center, and
| |
Collapse
|
14
|
Onabajo OO, Lewis MG, Mattapallil JJ. GALT CD4 +PD-1 hi T follicular helper (Tfh) cells repopulate after anti-retroviral therapy. Cell Immunol 2021; 366:104396. [PMID: 34157462 DOI: 10.1016/j.cellimm.2021.104396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/15/2021] [Accepted: 06/11/2021] [Indexed: 11/26/2022]
Abstract
Human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) infections are characterized by dramatic alterations in the mucosal CD4 T cell compartment. Though viremia is effectively suppressed, and peripheral CD4 T cell numbers recover to near healthy levels after highly active anti-retroviral therapy (HAART), some of the dysfunctional consequences of HIV infection continue to persist during therapy. We hypothesized that CD4 T follicular helper (Tfh) cell deficiencies may play a role in this process. Using the macaque model we show that SIV infection was associated with a significant loss of Tfh cells in the GALT that drain the mesentery lining the gastrointestinal tract (GIT). Loss of Tfh cells significantly correlated with the depletion of the overall memory CD4 T cell compartment; most Tfh cells in the GALT expressed a CD95+CD28+ memory phenotype suggesting that infection of the memory compartment likely drives the loss of GALT Tfh cells during infection. Continuous anti-retroviral therapy (cART) was accompanied by a significant repopulation of Tfh cells in the GALT to levels similar to those of uninfected animals. Repopulating Tfh cells displayed significantly higher capacity to produce IL-21 as compared to SIV infected animals suggesting that cART fully restores Tfh cells that are functionally capable of supporting GC reactions in the GALT.
Collapse
Affiliation(s)
- Olusegun O Onabajo
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20814, USA
| | | | - Joseph J Mattapallil
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA.
| |
Collapse
|
15
|
Ponnan SM, Vidyavijayan KK, Thiruvengadam K, Hilda J N, Mathayan M, Murugavel KG, Hanna LE. Role of Circulating T Follicular Helper Cells and Stem-Like Memory CD4 + T Cells in the Pathogenesis of HIV-2 Infection and Disease Progression. Front Immunol 2021; 12:666388. [PMID: 33936106 PMCID: PMC8085399 DOI: 10.3389/fimmu.2021.666388] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/23/2021] [Indexed: 12/27/2022] Open
Abstract
CD4+ T cells are critical players in the host adaptive immune response. Emerging evidence suggests that certain CD4+ T cell subsets contribute significantly to the production of neutralizing antibodies and help in the control of virus replication. Circulating T follicular helper cells (Tfh) constitute a key T cell subset that triggers the adaptive immune response and stimulates the production of neutralizing antibodies (NAbs). T cells having stem cell-like property, called stem-like memory T cells (Tscm), constitute another important subset of T cells that play a critical role in slowing the rate of disease progression through the differentiation and expansion of different types of memory cell subsets. However, the role of these immune cell subsets in T cell homeostasis, CD4+ T cell proliferation, and progression of disease, particularly in HIV-2 infection, has not yet been elucidated. The present study involved a detailed evaluation of the different CD4+ T cell subsets in HIV-2 infected persons with a view to understanding the role of these immune cell subsets in the better control of virus replication and delayed disease progression that is characteristic of HIV-2 infection. We observed elevated levels of CD4+ Tfh and CD4+ Tscm cells along with memory and effector T cell abundance in HIV-2 infected individuals. We also found increased frequencies of CXCR5+ CD8+ T cells and CD8+ Tscm cells, as well as memory B cells that are responsible for NAb development in HIV-2 infected persons. Interestingly, we found that the frequency of memory CD4+ T cells as well as memory B cells correlated significantly with neutralizing antibody titers in HIV-2 infected persons. These observations point to a more robust CD4+ T cell response that supports B cell differentiation, antibody production, and CD8+ T cell development in HIV-2 infected persons and contributes to better control of the virus and slower rate of disease progression in these individuals.
Collapse
Affiliation(s)
- Sivasankaran Munusamy Ponnan
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India.,Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - K K Vidyavijayan
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Kannan Thiruvengadam
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Nancy Hilda J
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Manikannan Mathayan
- Centre for Drug Discovery and Development, Sathyabama Institute of Science and Technology, Chennai, India
| | | | - Luke Elizabeth Hanna
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| |
Collapse
|
16
|
Elazab MFA, Younes AM, Gaafar AY, Abu-Bryka AZ, Abdel-Daim MM. Immunosuppressive effect of cyclophosphamide in Nile tilapia (Oreochromis niloticus). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:20784-20793. [PMID: 33405143 DOI: 10.1007/s11356-020-11893-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/30/2020] [Indexed: 06/12/2023]
Abstract
Cyclophosphamide, a common chemotherapeutic and immune suppressor agent, is regularly used in research to weaken the immune system in laboratory animal models. Nile tilapia (Oreochromis niloticus) is a widely used experimental fish model for immune-modulatory research; with the lack of knowledge about the immune-compromised tilapia model, an urgent need is to develop and optimise such a model. Sixty healthy Oreochromis niloticus fish, average weight 50 ± 10 g, were divided into four experimental groups. Fish in group I, negative control group, were injected with phosphate-buffered saline only, and fish in groups II, III and IV were injected with cyclophosphamide (CP) at 100, 200 and 400 mg/kg body weight (BW), respectively, via the intraperitoneal route. Different immune-related parameters were investigated 3 weeks after CP injection. The results have revealed a significant decrease in total red blood cells (RBCs), white blood cells (WBCs) and thrombocyte counts and reduced haemoglobin and haematocrit values in CP-treated fish, especially those injected with 200 mg/kg BW compared with the control group (p < 0.05). Also, significantly lower levels of serum proteins (total protein, albumin, α1- and γ-globulins) were observed in CP-treated fish, especially those injected with 200 mg/kg BW in comparison with the control group (p < 0.05). Furthermore, CP-treated fish showed a reduction in the expression of immune-related genes (interleukin-1, and tumour necrosis factor-α in spleen and tumour necrosis factor-α and transforming growth factor β-1 in head-kidney), especially those injected with 200 mg/kg BW compared with the control group (p < 0.05). In conclusion, the Oreochromis niloticus immune-suppressed model can be induced by intraperitoneal CP injection at 200 mg/kg BW.
Collapse
Affiliation(s)
- Mohamed F A Elazab
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr Elsheikh, 33516, Egypt
| | - Abdelgayed M Younes
- Hydrobiology Department, Veterinary Research Division, National Research Centre, 33 El Buhouth St., Dokki, Giza, 12622, Egypt
| | - Alkhateib Y Gaafar
- Hydrobiology Department, Veterinary Research Division, National Research Centre, 33 El Buhouth St., Dokki, Giza, 12622, Egypt.
| | - Alaa Z Abu-Bryka
- Hydrobiology Department, Veterinary Research Division, National Research Centre, 33 El Buhouth St., Dokki, Giza, 12622, Egypt
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
17
|
Onabajo OO, Mattapallil JJ. Gut Microbiome Homeostasis and the CD4 T- Follicular Helper Cell IgA Axis in Human Immunodeficiency Virus Infection. Front Immunol 2021; 12:657679. [PMID: 33815419 PMCID: PMC8017181 DOI: 10.3389/fimmu.2021.657679] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Human Immunodeficiency Virus (HIV) and Simian Immunodeficiency Virus (SIV) are associated with severe perturbations in the gut mucosal environment characterized by massive viral replication and depletion of CD4 T cells leading to dysbiosis, breakdown of the epithelial barrier, microbial translocation, immune activation and disease progression. Multiple mechanisms play a role in maintaining homeostasis in the gut mucosa and protecting the integrity of the epithelial barrier. Among these are the secretory IgA (sIgA) that are produced daily in vast quantities throughout the mucosa and play a pivotal role in preventing commensal microbes from breaching the epithelial barrier. These microbe specific, high affinity IgA are produced by IgA+ plasma cells that are present within the Peyer’s Patches, mesenteric lymph nodes and the isolated lymphoid follicles that are prevalent in the lamina propria of the gastrointestinal tract (GIT). Differentiation, maturation and class switching to IgA producing plasma cells requires help from T follicular helper (Tfh) cells that are present within these lymphoid tissues. HIV replication and CD4 T cell depletion is accompanied by severe dysregulation of Tfh cell responses that compromises the generation of mucosal IgA that in turn alters barrier integrity leading to commensal bacteria readily breaching the epithelial barrier and causing mucosal pathology. Here we review the effect of HIV infection on Tfh cells and mucosal IgA responses in the GIT and the consequences these have for gut dysbiosis and mucosal immunopathogenesis.
Collapse
Affiliation(s)
- Olusegun O Onabajo
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Joseph J Mattapallil
- F. E. Hebert School of Medicine, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
18
|
Ward AR, Mota TM, Jones RB. Immunological approaches to HIV cure. Semin Immunol 2020; 51:101412. [PMID: 32981836 DOI: 10.1016/j.smim.2020.101412] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
Combination antiretroviral therapy (ART) to treat human immunodeficiency virus (HIV) infection has proven remarkably successful - for those who can access and afford it - yet HIV infection persists indefinitely in a reservoir of cells, despite effective ART and despite host antiviral immune responses. An HIV cure is therefore the next aspirational goal and challenge, though approaches differ in their objectives - with 'functional cures' aiming for durable viral control in the absence of ART, and 'sterilizing cures' aiming for the more difficult to realize objective of complete viral eradication. Mechanisms of HIV persistence, including viral latency, anatomical sequestration, suboptimal immune functioning, reservoir replenishment, target cell-intrinsic immune resistance, and, potentially, target cell distraction of immune effectors, likely need to be overcome in order to achieve a cure. A small fraction of people living with HIV (PLWH) naturally control infection via immune-mediated mechanisms, however, providing both sound rationale and optimism that an immunological approach to cure is possible. Herein we review up to date knowledge and emerging evidence on: the mechanisms contributing to HIV persistence, as well as potential strategies to overcome these barriers; promising immunological approaches to achieve viral control and elimination of reservoir-harboring cells, including harnessing adaptive immune responses to HIV and engineered therapies, as well as enhancers of their functions and of complementary innate immune functioning; and combination strategies that are most likely to succeed. Ultimately, a cure must be safe, effective, durable, and, eventually, scalable in order to be widely acceptable and available.
Collapse
Affiliation(s)
- Adam R Ward
- Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA; Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA; PhD Program in Epidemiology, The George Washington University, Washington, DC, USA
| | - Talia M Mota
- Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - R Brad Jones
- Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA; Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA.
| |
Collapse
|
19
|
Wendel BS, Fu Y, He C, Hernandez SM, Qu M, Zhang Z, Jiang Y, Han X, Xu J, Ding H, Jiang N, Shang H. Rapid HIV Progression Is Associated with Extensive Ongoing Somatic Hypermutation. THE JOURNAL OF IMMUNOLOGY 2020; 205:587-594. [PMID: 32591400 DOI: 10.4049/jimmunol.1901161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 05/15/2020] [Indexed: 12/24/2022]
Abstract
The Ab response to HIV is of great interest, particularly in the context of a protective vaccine and broadly neutralizing Abs, but research is typically geared toward elite controllers because of their ability to successfully control the virus. In this study, we studied the evolution of the Ab repertoire over the first year of HIV infection in people classified as rapid progressors (RP) compared with typical progressors. HIV RPs are an important yet understudied group of HIV patients classified by a rapid decline in CD4 counts and accelerated development of AIDS. We found that the global IgG somatic hypermutation load negatively correlated with disease progression, possibly because of exaggerated isotype switching of unmutated sequences in patients with low CD4 counts. We measured Ab sequence evolution over time using longitudinal samples taken during the early stages of infection and 1 year postinfection. Within clonal lineages spanning both timepoints, visit 2-derived sequences harbored considerably more mutations than their visit 1 relatives. Despite extensive ongoing somatic hypermutation, the initially strong signs of Ag selection pressure observed in visit 1-derived sequences decayed by visit 2. These data suggest that excessive immune activation in RPs leads to a hyperactive B cell response that fails to confer protection.
Collapse
Affiliation(s)
- Ben S Wendel
- McKetta Department of Chemical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 78712
| | - Yajing Fu
- Key Laboratory of AIDS Immunology of National Health Commission (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Chenfeng He
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 78712; and
| | - Stefany M Hernandez
- McKetta Department of Chemical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 78712
| | - Mingjuan Qu
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 78712; and
| | - Zining Zhang
- Key Laboratory of AIDS Immunology of National Health Commission (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Yongjun Jiang
- Key Laboratory of AIDS Immunology of National Health Commission (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Xiaoxu Han
- Key Laboratory of AIDS Immunology of National Health Commission (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Junjie Xu
- Key Laboratory of AIDS Immunology of National Health Commission (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Haibo Ding
- Key Laboratory of AIDS Immunology of National Health Commission (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Ning Jiang
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 78712; and .,Institute for Cellular and Molecular Biology, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712
| | - Hong Shang
- Key Laboratory of AIDS Immunology of National Health Commission (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; .,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
20
|
Planchais C, Kök A, Kanyavuz A, Lorin V, Bruel T, Guivel-Benhassine F, Rollenske T, Prigent J, Hieu T, Prazuck T, Lefrou L, Wardemann H, Schwartz O, Dimitrov JD, Hocqueloux L, Mouquet H. HIV-1 Envelope Recognition by Polyreactive and Cross-Reactive Intestinal B Cells. Cell Rep 2020; 27:572-585.e7. [PMID: 30970259 PMCID: PMC6458971 DOI: 10.1016/j.celrep.2019.03.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 02/19/2019] [Accepted: 03/08/2019] [Indexed: 12/18/2022] Open
Abstract
Mucosal immune responses to HIV-1 involve the recognition of the viral envelope glycoprotein (gp)160 by tissue-resident B cells and subsequent secretion of antibodies. To characterize the B cells “sensing” HIV-1 in the gut of infected individuals, we probed monoclonal antibodies produced from single intestinal B cells binding to recombinant gp140 trimers. A large fraction of mucosal B cell antibodies were polyreactive and showed only low affinity to HIV-1 envelope glycoproteins, particularly the gp41 moiety. A few high-affinity gp140 antibodies were isolated but lacked neutralizing, potent ADCC, and transcytosis-blocking capacities. Instead, they displayed cross-reactivity with defined self-antigens. Specifically, intestinal HIV-1 gp41 antibodies targeting the heptad repeat 2 region (HR2) cluster II cross-reacted with the p38α mitogen-activated protein kinase 14 (MAPK14). Hence, physiologic polyreactivity of intestinal B cells and molecular mimicry-based self-reactivity of HIV-1 antibodies are two independent phenomena, possibly diverting and/or impairing mucosal humoral immunity to HIV-1. Polyreactive B cells in HIV-1+ intestinal mucosa interact with HIV-1 Env proteins High-affinity intestinal HIV-1 gp140 antibodies display poor antiviral activities Antibodies targeting the gp41 cluster II region cross-react with MAPK14
Collapse
Affiliation(s)
- Cyril Planchais
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Ayrin Kök
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Alexia Kanyavuz
- Sorbonne Universités, UPMC Université Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France
| | - Valérie Lorin
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Timothée Bruel
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Paris 75015, France; CNRS URA3015, Paris, 75015, France
| | - Florence Guivel-Benhassine
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Paris 75015, France; CNRS URA3015, Paris, 75015, France
| | - Tim Rollenske
- Division of B Cell Immunology, German Cancer Research Center, Heidelberg 69120, Germany
| | - Julie Prigent
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Thierry Hieu
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France
| | - Thierry Prazuck
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, Orléans 45067, France
| | - Laurent Lefrou
- Service d'Hépato-Gastro-Entérologie, CHR d'Orléans-La Source, Orléans 45067, France
| | - Hedda Wardemann
- Division of B Cell Immunology, German Cancer Research Center, Heidelberg 69120, Germany
| | - Olivier Schwartz
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, Paris 75015, France; CNRS URA3015, Paris, 75015, France
| | - Jordan D Dimitrov
- Sorbonne Universités, UPMC Université Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France; Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris 75006, France
| | - Laurent Hocqueloux
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, Orléans 45067, France
| | - Hugo Mouquet
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris 75015, France; INSERM U1222, Paris 75015, France.
| |
Collapse
|
21
|
Giltiay NV, Giordano D, Clark EA. The Plasticity of Newly Formed B Cells. THE JOURNAL OF IMMUNOLOGY 2020; 203:3095-3104. [PMID: 31818922 DOI: 10.4049/jimmunol.1900928] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 09/26/2019] [Indexed: 12/21/2022]
Abstract
Newly formed B cells (NF-B cells) that emerge from the bone marrow to the periphery have often been referred to as immature or transitional B cells. However, NF-B cells have several striking characteristics, including a distinct BCR repertoire, high expression of AID, high sensitivity to PAMPs, and the ability to produce cytokines. A number of findings do not support their designation as immature because NF-B cells have the potential to become Ab-producing cells and to undergo class-switch recombination. In this review, we provide a fresh perspective on NF-B cell functions and describe some of the signals driving their activation. We summarize growing evidence supporting a role for NF-B cells in protection against infections and as a potential source of autoantibody-producing cells in autoimmune diseases such as systemic lupus erythematosus.
Collapse
Affiliation(s)
- Natalia V Giltiay
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA 98109; and
| | - Daniela Giordano
- Department of Immunology, University of Washington, Seattle, WA 98109
| | - Edward A Clark
- Department of Immunology, University of Washington, Seattle, WA 98109
| |
Collapse
|
22
|
Cooper L, Good-Jacobson KL. Dysregulation of humoral immunity in chronic infection. Immunol Cell Biol 2020; 98:456-466. [PMID: 32275789 DOI: 10.1111/imcb.12338] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/29/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022]
Abstract
Chronic viral infections disrupt the ability of the humoral immune response to produce neutralizing antibody or form effective immune memory, preventing viral clearance and making vaccine design difficult. Multiple components of the B-cell response are affected by pathogens that are not cleared from the host. Changes in the microenvironment shift production of B cells to short-lived plasma cells early in the response. Polyclonal B cells are recruited into both the plasma cell and germinal center compartments, inhibiting the formation of a targeted, high-affinity response. Finally, memory B cells shift toward an "atypical" phenotype, which may in turn result in changes to the functional properties of this population. While similar properties of B-cell dysregulation have been described across different types of persistent infections, key questions about the underlying mechanisms remain. This review will discuss the recent advances in this field, as well as highlight the critical questions about the interplay between viral load, microenvironment, the polyclonal response and atypical memory B cells that are yet to be answered. Design of new preventative treatments will rely on identifying the extrinsic and intrinsic modulators that push B cells toward an ineffective response, and thus identify new ways to guide them back onto the best path for clearance of virus and formation of effective immune memory.
Collapse
Affiliation(s)
- Lucy Cooper
- Infection and Immunity Program, The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Kim L Good-Jacobson
- Infection and Immunity Program, The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| |
Collapse
|
23
|
Butler JE, Sinkora M, Wang G, Stepanova K, Li Y, Cai X. Perturbation of Thymocyte Development Underlies the PRRS Pandemic: A Testable Hypothesis. Front Immunol 2019; 10:1077. [PMID: 31156633 PMCID: PMC6529568 DOI: 10.3389/fimmu.2019.01077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/26/2019] [Indexed: 11/13/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) causes immune dysregulation during the Critical Window of Immunological Development. We hypothesize that thymocyte development is altered by infected thymic antigen presenting cells (TAPCs) in the fetal/neonatal thymus that interact with double-positive thymocytes causing an acute deficiency of T cells that produces "holes" in the T cell repertoire allowing for poor recognition of PRRSV and other neonatal pathogens. The deficiency may be the result of random elimination of PRRSV-specific T cells or the generation of T cells that accept PRRSV epitopes as self-antigens. Loss of helper T cells for virus neutralizing (VN) epitopes can result in the failure of selection for B cells in lymph node germinal centers capable of producing high affinity VN antibodies. Generation of cytotoxic and regulatory T cells may also be impaired. Similar to infections with LDV, LCMV, MCMV, HIV-1 and trypanosomes, the host responds to the deficiency of pathogen-specific T cells and perhaps regulatory T cells, by "last ditch" polyclonal B cell activation. In colostrum-deprived PRRSV-infected isolator piglets, this results in hypergammaglobulinemia, which we believe to be a "red herring" that detracts attention from the thymic atrophy story, but leads to our second independent hypothesis. Since hypergammaglobulinemia has not been reported in PRRSV-infected conventionally-reared piglets, we hypothesize that this is due to the down-regulatory effect of passive maternal IgG and cytokines in porcine colostrum, especially TGFβ which stimulates development of regulatory T cells (Tregs).
Collapse
Affiliation(s)
- John E. Butler
- Carver College of Medicine, University of Iowa, Iowa, IA, United States
| | - Marek Sinkora
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Gang Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Katerina Stepanova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Yuming Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xuehui Cai
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
24
|
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
|
25
|
Mucosal T follicular helper cells in SIV-infected rhesus macaques: contributing role of IL-27. Mucosal Immunol 2019; 12:1038-1054. [PMID: 31114010 PMCID: PMC7746526 DOI: 10.1038/s41385-019-0174-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 04/04/2019] [Accepted: 04/17/2019] [Indexed: 02/04/2023]
Abstract
Mesenteric lymph nodes (MLNs), that drain the large and small intestine, are critical sites for the induction of oral tolerance. Although depletion of CD4 T cells in the intestinal lamina propria is a hallmark of HIV infection, CD4 T cell dynamics in MLNs is less known due to the lack of accessibility to these LNs. We demonstrate the early loss of memory CD4 T cells, including T follicular helper cells (Tfh) and a remodeling of MLN architecture in SIV-infected rhesus macaques (RMs). Along with the loss of Tfh cells, we observe the loss of memory B cells and of germinal center B cells. Tfh cells display a Th1 profile with increased levels of the transcription factors that negatively impact on Tfh differentiation and of Stat5 phosphorylation. MLNs of SIV-infected RMs display lower mRNA transcripts encoding for IL-12, IL-23, and IL-35, whereas those coding for IL-27 are not impaired in MLNs. In vitro, IL-27 negatively impacts on Tfh cells and recapitulates the profile observed in SIV-infected RMs. Therefore, early defects of memory CD4 T cells, as well of Tfh cells in MLNs, which play a central role in regulating the mucosal immune response, may have major implications for Aids.
Collapse
|
26
|
Veazey RS. Intestinal CD4 Depletion in HIV / SIV Infection. CURRENT IMMUNOLOGY REVIEWS 2019; 15:76-91. [PMID: 31431807 PMCID: PMC6701936 DOI: 10.2174/1573395514666180605083448] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/12/2018] [Accepted: 05/18/2018] [Indexed: 12/28/2022]
Abstract
Among the most significant findings in the pathogenesis of HIV infection was the discovery that almost total depletion of intestinal CD4+ T cells occurs rapidly after SIV or HIV infection, regardless of the route of exposure, and long before CD4+ T cell losses occur in blood or lymph nodes. Since these seminal discoveries, we have learned much about mucosal and systemic CD4+ T cells, and found several key differences between the circulating and intestinal CD4+ T cell subsets, both in phenotype, relative proportions, and functional capabilities. Further, specific subsets of CD4+ T cells are selectively targeted and eliminated first, especially cells critically important for initiating primary immune responses, and for maintenance of mucosal integrity (Th1, Th17, and Th22 cells). This simultaneously results in loss of innate immune responses, and loss of mucosal integrity, resulting in mucosal, and systemic immune activation that drives proliferation and activation of new target cells throughout the course of infection. The propensity for the SIV/HIV to infect and efficiently replicate in specific cells also permits viral persistence, as the mucosal and systemic activation that ensues continues to damage mucosal barriers, resulting in continued influx of target cells to maintain viral replication. Finally, infection and elimination of recently activated and proliferating CD4+ T cells, and infection and dysregulation of Tfh and other key CD4+ T cell results in hyperactive, yet non-protective immune responses that support active viral replication and evolution, and thus persistence in host tissue reservoirs, all of which continue to challenge our efforts to design effective vaccine or cure strategies.
Collapse
Affiliation(s)
- Ronald S. Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
27
|
Nicholson LK, Pratap H, Bowers E, Gunzburger E, Bandi SR, Gardner EM, Palmer BE, Wright T, Kittelson J, Janoff EN. Effective B cell activation in vitro during viremic HIV-1 infection with surrogate T cell stimulation. Immunobiology 2018; 223:839-849. [PMID: 30219203 PMCID: PMC6264910 DOI: 10.1016/j.imbio.2018.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/09/2018] [Accepted: 08/19/2018] [Indexed: 02/07/2023]
Abstract
Identifying HIV-1-associated B cell defects and responses to activation may direct interventions to circumvent their impaired antibody responses to infection and vaccines. Among 34 viremic HIV-1-infected and 20 seronegative control adults, we measured baseline frequencies and activation of B and T cell subsets, expression of activation-induced cytidine deaminase (AID), potential determinants of B cell activation in vivo and B and T cell responses in vitro. At baseline, HIV-1 infection was associated with increased IgM memory and decreased anergic cell frequencies, as well as increased activation in all 10 B cell subsets compared with controls. HIV-1 status, TFH activation, and BAFF were significant potential drivers of B cell activation. Despite high baseline activation among HIV-1-infected subjects, stimulation in vitro with combined surrogates for antigen (anti-IgM), cognate (CD40 ligand) and soluble T cell factors (IL-4) elicited comparable B cell activation, transitions from naïve to class-switched memory cells and AID expression in both groups. In summary, viremic HIV-1 infection perturbs circulating B cell subsets and activation at each stage of B cell maturation. However, that appropriate stimulation of B cells elicits effective activation and maturation provides impetus for advancing vaccine development to prevent secondary infections by circumventing early B cell defects.
Collapse
Affiliation(s)
- Lindsay K Nicholson
- Mucosal and Vaccine Research Program Colorado (MAVRC), United States; Departments of Medicine, University of Colorado Denver, Aurora, CO, United States; Denver Veterans Affairs Medical Center, Denver, CO, United States
| | - Harsh Pratap
- Mucosal and Vaccine Research Program Colorado (MAVRC), United States; Departments of Medicine, University of Colorado Denver, Aurora, CO, United States; Denver Veterans Affairs Medical Center, Denver, CO, United States
| | - Elisabeth Bowers
- Mucosal and Vaccine Research Program Colorado (MAVRC), United States; Departments of Medicine, University of Colorado Denver, Aurora, CO, United States; Denver Veterans Affairs Medical Center, Denver, CO, United States
| | - Elise Gunzburger
- Departments of Biostatistics, University of Colorado Denver, Aurora, CO, United States
| | - Srinivasa R Bandi
- Mucosal and Vaccine Research Program Colorado (MAVRC), United States; Departments of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Edward M Gardner
- Departments of Medicine, University of Colorado Denver, Aurora, CO, United States; Denver Health and Hospital Authority, Denver, CO, United States
| | - Brent E Palmer
- Mucosal and Vaccine Research Program Colorado (MAVRC), United States; Departments of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Timothy Wright
- Denver Health and Hospital Authority, Denver, CO, United States
| | - John Kittelson
- Mucosal and Vaccine Research Program Colorado (MAVRC), United States; Departments of Biostatistics, University of Colorado Denver, Aurora, CO, United States
| | - Edward N Janoff
- Mucosal and Vaccine Research Program Colorado (MAVRC), United States; Departments of Medicine, University of Colorado Denver, Aurora, CO, United States; Denver Veterans Affairs Medical Center, Denver, CO, United States.
| |
Collapse
|
28
|
Xu W, Luo Z, Alekseyenko AV, Martin L, Wan Z, Ling B, Qin Z, Heath SL, Maas K, Cong X, Jiang W. Distinct systemic microbiome and microbial translocation are associated with plasma level of anti-CD4 autoantibody in HIV infection. Sci Rep 2018; 8:12863. [PMID: 30150778 PMCID: PMC6110826 DOI: 10.1038/s41598-018-31116-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 08/03/2018] [Indexed: 12/13/2022] Open
Abstract
Microbial signals have been linked to autoantibody induction. Recently, we found that purified anti-CD4 autoantibodies from the plasma of chronic HIV-1-infected patients under viral-suppressed antiretroviral therapy (ART) play a pathologic role in poor CD4+ T cell recovery. The purpose of the study was to investigate the association of systemic microbiome and anti-CD4 autoantibody production in HIV. Plasma microbiome from 12 healthy controls and 22 HIV-infected subjects under viral-suppressed ART were analyzed by MiSeq sequencing. Plasma level of autoantibodies and microbial translocation (LPS, total bacterial 16S rDNA, soluble CD14, and LPS binding protein) were analyzed by ELISA, limulus amebocyte assay, and qPCR. We found that plasma level of anti-CD4 IgGs but not anti-CD8 IgGs was increased in HIV+ subjects compared to healthy controls. HIV+ subjects with plasma anti-CD4 IgG > 50 ng/mL (high) had reduced microbial diversity compared to HIV+ subjects with anti-CD4 IgG ≤ 50 ng/mL (low). Moreover, plasma anti-CD4 IgG level was associated with elevated microbial translocation and reduced microbial diversity in HIV+ subjects. The Alphaproteobacteria class was significantly enriched in HIV+ subjects with low anti-CD4 IgG compared to patients with high anti-CD4 IgG even after controlling for false discovery rate (FDR). The microbial components were different from the phylum to genus level in HIV+ subjects with high anti-CD4 IgGs compared to the other two groups, but these differences were not significant after controlling for FDR. These results suggest that systemic microbial translocation and microbiome may associate with anti-CD4 autoantibody production in ART-treated HIV disease.
Collapse
Affiliation(s)
- Wanli Xu
- University of Connecticut School of Nursing, Storrs, Connecticut, 06269, USA
| | - Zhenwu Luo
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Alexander V Alekseyenko
- Program for Human Microbiome Research, Biomedical Informatics Center, Department of Public Health Sciences, Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Lisa Martin
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Zhuang Wan
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Binhua Ling
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Tulane National Primate Research Center, New Orleans, LA, 70433, USA
| | - Zhiqiang Qin
- Departments of Genetics, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, 1700 Tulane Ave., New Orleans, LA, 70112, USA
| | - Sonya L Heath
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Kendra Maas
- Microbial Analysis, Resources, and Services, University of Connecticut, Storrs, CT, 06269, USA
| | - Xiaomei Cong
- University of Connecticut School of Nursing, Storrs, Connecticut, 06269, USA.
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
29
|
Greczmiel U, Oxenius A. The Janus Face of Follicular T Helper Cells in Chronic Viral Infections. Front Immunol 2018; 9:1162. [PMID: 29887868 PMCID: PMC5982684 DOI: 10.3389/fimmu.2018.01162] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/09/2018] [Indexed: 12/28/2022] Open
Abstract
Chronic infections with non-cytopathic viruses constitutively expose virus-specific adaptive immune cells to cognate antigen, requiring their numeric and functional adaptation. Virus-specific CD8 T cells are compromised by various means in their effector functions, collectively termed T cell exhaustion. Alike CD8 T cells, virus-specific CD4 Th1 cell responses are gradually downregulated but instead, follicular T helper (TFH) cell differentiation and maintenance is strongly promoted during chronic infection. Thereby, the immune system promotes antibody responses, which bear less immune-pathological risk compared to cytotoxic and pro-inflammatory T cell responses. This emphasis on TFH cells contributes to tolerance of the chronic infection and is pivotal for the continued maturation and adaptation of the antibody response, leading eventually to the emergence of virus-neutralizing antibodies, which possess the potential to control the established chronic infection. However, sustained high levels of TFH cells can also result in a less stringent B cell selection process in active germinal center reactions, leading to the activation of virus-unspecific B cells, including self-reactive B cells, and to hypergammaglobulinemia. This dispersal of B cell help comes at the expense of a stringently selected virus-specific antibody response, thereby contributing to its delayed maturation. Here, we discuss these opposing facets of TFH cells in chronic viral infections.
Collapse
Affiliation(s)
- Ute Greczmiel
- Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | | |
Collapse
|
30
|
Abstract
OBJECTIVE To characterize the effect of the HIV-1 infection and antiretroviral treatment (ART) in the human memory B (MEB)-cell compartment. DESIGN A cross-sectional study was designed to analyze MEB cells of HIV-1 ART treated and ART-naive study participants, and uninfected individuals. METHODS Frequency and absolute counts of MEB cell subsets in blood were determined by multicolor flow cytometry. Spontaneous cell death and B-cell proliferative capacity was evaluated in vitro by cell culture and flow cytometry. Splenic function was determined by pitted erythrocytes quantification in HIV-1 ART-treated study participants. RESULTS HIV-1 ART-treated individuals did not show functional hyposplenism despite the lack of recovery IgMIgDCD27 marginal zone-like B cells. Moreover, two germinal center-dependent MEB cells subsets were also dysregulated in HIV-1 individuals: IgMIgDCD27 (IgM only) cells were increased, whereas the switched subset (IgMIgD) was reduced in viremic individuals. Althought ART restored the numbers of these populations; the switched MEB cells were enriched in CD27 cells, which showed the highest susceptibility to spontaneous cell death ex vivo. In addition, B cells from viremic individuals showed a poor response to B-cell receptor and toll-like receptor 9 stimulation that was circumvented when both stimuli were used simultaneously. CONCLUSION B cells from HIV-1 study participants show a poor stimulation capacity, that may be bypassed by the proper combination of stimuli, and a dysregulated MEB cell pool that suggest an affectation of the germinal center reaction, only partially normalized by ART. Interestingly, hyposplenism does not explain the lack of recovery of the marginal zone-like B cells in ART-treated HIV-1 individuals.
Collapse
|
31
|
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
|
32
|
Luo Z, Li Z, Martin L, Wan Z, Meissner EG, Espinosa E, Wu H, Yu X, Fu P, Julia Westerink MA, Kilby JM, Wu J, Huang L, Heath SL, Li Z, Jiang W. Pathological Role of Anti-CD4 Antibodies in HIV-Infected Immunologic Nonresponders Receiving Virus-Suppressive Antiretroviral Therapy. J Infect Dis 2017; 216:82-91. [PMID: 28498953 DOI: 10.1093/infdis/jix223] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 05/08/2017] [Indexed: 12/22/2022] Open
Abstract
Increased mortality and morbidity occur among human immunodeficiency virus (HIV)-infected patients in whom CD4+ T-cell counts do not increase despite viral suppression with antiretroviral therapy (ART). Here we identified an underlying mechanism. Significantly elevated plasma levels of anti-CD4 immunoglobulin G (IgG) were found in HIV-positive immunologic nonresponders (ie, HIV-positive individuals with CD4+ T-cell counts of ≤350 cells/μL), compared with levels in HIV-positive immunologic responders (ie, HIV-positive individuals with CD4+ T-cell counts of ≥500 cells/μL) and healthy controls. Higher plasma level of anti-CD4 IgG correlated with blunted CD4+ T-cell recovery. Furthermore, purified anti-CD4 IgG from HIV-positive immunologic nonresponders induced natural killer (NK) cell-dependent CD4+ T-cell cytolysis and apoptosis through antibody-dependent cell-mediated cytotoxicity (ADCC) in vitro. We also found that anti-CD4 IgG-mediated ADCC exerts greater apoptosis of naive CD4+ T cells relative to memory CD4+ T cells. Consistently, increased frequencies of CD107a+ NK cells and profound decreases of naive CD4+ T cells were observed in immunologic nonresponders as compared to responders and healthy controls ex vivo. These data indicate that autoreactive anti-CD4 IgG may play an important role in blunted CD4+ T-cell reconstitution despite effective ART.
Collapse
Affiliation(s)
- Zhenwu Luo
- Department of Microbiology and Immunology
| | - Zhen Li
- Department of Microbiology and Immunology.,Beijing You'an Hospital, Capital Medical University
| | - Lisa Martin
- Divison of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston
| | - Zhuang Wan
- Department of Microbiology and Immunology
| | - Eric G Meissner
- Department of Microbiology and Immunology.,Divison of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston
| | - Enrique Espinosa
- Department of Integrative in Immunology, National Institute for Respiratory Diseases, Mexico City, Mexico
| | - Hao Wu
- Beijing You'an Hospital, Capital Medical University
| | - Xiaocong Yu
- Department of Medicine, University of Massachusetts Medical School, Worcester
| | - Pingfu Fu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio
| | - Maria Anna Julia Westerink
- Divison of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston
| | - J Michael Kilby
- Divison of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston
| | | | - Lei Huang
- Treatment and Research Center for Infectious Diseases, 302nd Hospital of the PLA, Beijing, China
| | - Sonya L Heath
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham
| | - Zihai Li
- Department of Microbiology and Immunology
| | - Wei Jiang
- Department of Microbiology and Immunology.,Divison of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston
| |
Collapse
|
33
|
Mabuka JM, Dugast AS, Muema DM, Reddy T, Ramlakhan Y, Euler Z, Ismail N, Moodley A, Dong KL, Morris L, Walker BD, Alter G, Ndung’u T. Plasma CXCL13 but Not B Cell Frequencies in Acute HIV Infection Predicts Emergence of Cross-Neutralizing Antibodies. Front Immunol 2017; 8:1104. [PMID: 28943879 PMCID: PMC5596076 DOI: 10.3389/fimmu.2017.01104] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/23/2017] [Indexed: 01/12/2023] Open
Abstract
Immunological events in acute HIV-1 infection before peak viremia (hyperacute phase) may contribute to the development of broadly cross-neutralizing antibodies. Here, we used pre-infection and acute-infection peripheral blood mononuclear cells and plasma samples from 22 women, including 10 who initiated antiretroviral treatment in Fiebig stages I-V of acute infection to study B cell subsets and B-cell associated cytokines (BAFF and CXCL13) kinetics for up to ~90 days post detection of plasma viremia. Frequencies of B cell subsets were defined by flow cytometry while plasma cytokine levels were measured by ELISA. We observed a rapid but transient increase in exhausted tissue-like memory, activated memory, and plasmablast B cells accompanied by decline in resting memory cells in untreated, but not treated women. B cell subset frequencies in untreated women positively correlated with viral loads but did not predict emergence of cross-neutralizing antibodies measured 12 months post detection of plasma viremia. Plasma BAFF and CXCL13 levels increased only in untreated women, but their levels did not correlate with viral loads. Importantly, early CXCL13 but not BAFF levels predicted the later emergence of detectable cross-neutralizing antibodies at 12 months post detection of plasma viremia. Thus, hyperacute HIV-1 infection is associated with B cell subset changes, which do not predict emergence of cross-neutralizing antibodies. However, plasma CXCL13 levels during hyperacute infection predicted the subsequent emergence of cross-neutralizing antibodies, providing a potential biomarker for the evaluation of vaccines designed to elicit cross-neutralizing activity or for natural infection studies to explore mechanisms underlying development of neutralizing antibodies.
Collapse
Affiliation(s)
- Jenniffer M. Mabuka
- Africa Health Research Institute, Durban, South Africa
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, 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
| | - Anne-Sophie Dugast
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
| | - Daniel M. Muema
- Africa Health Research Institute, Durban, South Africa
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Tarylee Reddy
- Biostatistics Unit, Medical Research Council, Durban, South Africa
| | - Yathisha Ramlakhan
- Africa Health Research Institute, Durban, South Africa
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Zelda Euler
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
| | - Nasreen Ismail
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Amber Moodley
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, 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
| | - Krista L. Dong
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, 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
| | - Lynn Morris
- National Institute for Communicable Diseases, Johannesburg, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Bruce D. Walker
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, 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
- Howard Hughes Medical Institute, Chevy Chase, MD, United States
- Institute for Medical and Engineering Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
| | - Thumbi Ndung’u
- Africa Health Research Institute, Durban, South Africa
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, 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
| |
Collapse
|
34
|
Borrow P, Moody MA. Immunologic characteristics of HIV-infected individuals who make broadly neutralizing antibodies. Immunol Rev 2017; 275:62-78. [PMID: 28133804 PMCID: PMC5299500 DOI: 10.1111/imr.12504] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Induction of broadly neutralizing antibodies (bnAbs) capable of inhibiting infection with diverse variants of human immunodeficiency virus type 1 (HIV‐1) is a key, as‐yet‐unachieved goal of prophylactic HIV‐1 vaccine strategies. However, some HIV‐infected individuals develop bnAbs after approximately 2‐4 years of infection, enabling analysis of features of these antibodies and the immunological environment that enables their induction. Distinct subsets of CD4+ T cells play opposing roles in the regulation of humoral responses: T follicular helper (Tfh) cells support germinal center formation and provide help for affinity maturation and the development of memory B cells and plasma cells, while regulatory CD4+ (Treg) cells including T follicular regulatory (Tfr) cells inhibit the germinal center reaction to limit autoantibody production. BnAbs exhibit high somatic mutation frequencies, long third heavy‐chain complementarity determining regions, and/or autoreactivity, suggesting that bnAb generation is likely to be highly dependent on the activity of CD4+ Tfh cells, and may be constrained by host tolerance controls. This review discusses what is known about the immunological environment during HIV‐1 infection, in particular alterations in CD4+ Tfh, Treg, and Tfr populations and autoantibody generation, and how this is related to bnAb development, and considers the implications for HIV‐1 vaccine design.
Collapse
Affiliation(s)
- Persephone Borrow
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - M Anthony Moody
- Duke University Human Vaccine Institute and Departments of Pediatrics and Immunology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
35
|
Verkoczy L, Alt FW, Tian M. Human Ig knockin mice to study the development and regulation of HIV-1 broadly neutralizing antibodies. Immunol Rev 2017; 275:89-107. [PMID: 28133799 DOI: 10.1111/imr.12505] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A major challenge for HIV-1 vaccine research is developing a successful immunization approach for inducing broadly neutralizing antibodies (bnAbs). A key shortcoming in meeting this challenge has been the lack of animal models capable of identifying impediments limiting bnAb induction and ranking vaccine strategies for their ability to promote bnAb development. Since 2010, immunoglobulin knockin (KI) technology, involving inserting functional rearranged human variable exons into the mouse IgH and IgL loci has been used to express bnAbs in mice. This approach has allowed immune tolerance mechanisms limiting bnAb production to be elucidated and strategies to overcome such limitations to be evaluated. From these studies, along with the wealth of knowledge afforded by analyses of recombinant Ig-based bnAb structures, it became apparent that key functional features of bnAbs often are problematic for their elicitation in mice by classic vaccine paradigms, necessitating more iterative testing of new vaccine concepts. In this regard, bnAb KI models expressing deduced precursor V(D)J rearrangements of mature bnAbs or unrearranged germline V, D, J segments (that can be assembled into variable region exons that encode bnAb precursors), have been engineered to evaluate novel immunogens/regimens for effectiveness in driving bnAb responses. One promising approach emerging from such studies is the ability of sequentially administered, modified immunogens (designed to bind progressively more mature bnAb precursors) to initiate affinity maturation. Here, we review insights gained from bnAb KI studies regarding the regulation and induction of bnAbs, and discuss new Ig KI methodologies to manipulate the production and/or expression of bnAbs in vivo, to further facilitate vaccine-guided bnAb induction studies.
Collapse
Affiliation(s)
- Laurent Verkoczy
- Departments of Medicine and Pathology, Duke University Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Frederick W Alt
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Ming Tian
- Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Genetics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
36
|
French MA, Tjiam MC, Abudulai LN, Fernandez S. Antiviral Functions of Human Immunodeficiency Virus Type 1 (HIV-1)-Specific IgG Antibodies: Effects of Antiretroviral Therapy and Implications for Therapeutic HIV-1 Vaccine Design. Front Immunol 2017; 8:780. [PMID: 28725225 PMCID: PMC5495868 DOI: 10.3389/fimmu.2017.00780] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 06/19/2017] [Indexed: 12/24/2022] Open
Abstract
Contemporary antiretroviral therapy (ART) is effective and tolerable for long periods of time but cannot eradicate human immunodeficiency virus type 1 (HIV-1) infection by either elimination of viral reservoirs or enhancement of HIV-1-specific immune responses. Boosting "protective" HIV-1-specific immune responses by active or passive immunization will therefore be necessary to control or eradicate HIV-1 infection and is currently the topic of intense investigation. Recently reported studies conducted in HIV patients and non-human primate (NHP) models of HIV-1 infection suggest that HIV-1-specific IgG antibody responses may contribute to the control of HIV-1 infection. However, production of IgG antibodies with virus neutralizing activity by vaccination remains problematic and while vaccine-induced natural killer cell-activating IgG antibodies have been shown to prevent the acquisition of HIV-1 infection, they may not be sufficient to control or eradicate established HIV-1 infection. It is, therefore, important to consider other functional characteristics of IgG antibody responses. IgG antibodies to viruses also mediate opsonophagocytic antibody responses against virions and capsids that enhance the function of phagocytic cells playing critical roles in antiviral immune responses, particularly conventional dendritic cells and plasmacytoid dendritic cells. Emerging evidence suggests that these antibody functions might contribute to the control of HIV-1 infection. In addition, IgG antibodies contribute to the intracellular degradation of viruses via binding to the cytosolic fragment crystallizable (Fc) receptor tripartite motif containing-21 (TRIM21). The functional activity of an IgG antibody response is influenced by the IgG subclass content, which affects binding to antigens and to Fcγ receptors on phagocytic cells and to TRIM21. The IgG subclass content and avidity of IgG antibodies is determined by germinal center (GC) reactions in follicles of lymphoid tissue. As HIV-1 infects cells in GCs and induces GC dysfunction, which may persist during ART, strategies for boosting HIV-1-specific IgG antibody responses should include early commencement of ART and possibly the use of particular antiretroviral drugs to optimize drug levels in lymphoid follicles. Finally, enhancing particular functions of HIV-1-specific IgG antibody responses by using adjuvants or cytokines to modulate the IgG subclass content of the antibody response might be investigated in NHP models of HIV-1 infection and during trials of therapeutic vaccines in HIV patients.
Collapse
Affiliation(s)
- Martyn A. French
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- Medical School, University of Western Australia, Perth, WA, Australia
- Department of Clinical Immunology, Royal Perth Hospital and PathWest Laboratory Medicine, Perth, WA, Australia
| | - M. Christian Tjiam
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Laila N. Abudulai
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Sonia Fernandez
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
37
|
Zaunders J, Xu Y, Kent SJ, Koelsch KK, Kelleher AD. Divergent Expression of CXCR5 and CCR5 on CD4 + T Cells and the Paradoxical Accumulation of T Follicular Helper Cells during HIV Infection. Front Immunol 2017; 8:495. [PMID: 28553284 PMCID: PMC5427074 DOI: 10.3389/fimmu.2017.00495] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 04/10/2017] [Indexed: 12/23/2022] Open
Abstract
Viral infection sets in motion a cascade of immune responses, including both CXCR5+CD4+ T follicular helper (Tfh) cells that regulate humoral immunity and CCR5+CD4+ T cells that mediate cell-mediated immunity. In peripheral blood mononuclear cells, the majority of memory CD4+ T cells appear to fall into either of these two lineages, CCR5−CXCR5+ or CCR5+CXCR5−. Very high titers of anti-HIV IgG antibodies are a hallmark of infection, strongly suggesting that there is significant HIV-specific CD4+ T cell help to HIV-specific B cells. We now know that characteristic increases in germinal centers (GC) in lymphoid tissue (LT) during SIV and HIV-1 infections are associated with an increase in CXCR5+PD-1high Tfh, which expand to a large proportion of memory CD4+ T cells in LT, and are presumably specific for SIV or HIV epitopes. Macaque Tfh normally express very little CCR5, yet are infected by CCR5-using SIV, which may occur mainly through infection of a subset of PD-1intermediateCCR5+Bcl-6+ pre-Tfh cells. In contrast, in human LT, a subset of PD-1high Tfh appears to express low levels of CCR5, as measured by flow cytometry, and this may also contribute to the high rate of infection of Tfh. Also, we have found, by assessing fine-needle biopsies of LT, that increases in Tfh and GC B cells in HIV infection are not completely normalized by antiretroviral therapy (ART), suggesting a possible long-lasting reservoir of infected Tfh. In contrast to the increase of CXCR5+ Tfh, there is no accumulation of proliferating CCR5+ CD4 T HIV Gag-specific cells in peripheral blood that make IFN-γ. Altogether, CXCR5+CCR5− CD4 T cells that regulate humoral immunity are allowed greater freedom to operate and expand during HIV-1 infection, but at the same time can contain HIV DNA at levels at least as high as in other CD4 subsets. We argue that early ART including a CCR5 blocker may directly reduce the infected Tfh reservoir in LT and also interrupt cycles of antibody pressure driving virus mutation and additional GC responses to resulting neoantigens.
Collapse
Affiliation(s)
- John Zaunders
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia.,The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Yin Xu
- The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia.,Department of Infectious Diseases, Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Kersten K Koelsch
- The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Anthony D Kelleher
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia.,The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
38
|
Production of IgG antibodies to pneumococcal polysaccharides is associated with expansion of ICOS+ circulating memory T follicular-helper cells which is impaired by HIV infection. PLoS One 2017; 12:e0176641. [PMID: 28463977 PMCID: PMC5413043 DOI: 10.1371/journal.pone.0176641] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/13/2017] [Indexed: 11/26/2022] Open
Abstract
Dysfunction of T follicular-helper (TFH) cells is a possible cause of impaired germinal centre (GC) and IgG antibody responses in individuals with human immunodeficiency virus-1 (HIV-1) infection and might contribute to decreased magnitude and isotype diversification of IgG antibodies to pneumococcal polysaccharides (PcPs). We examined the production of IgG1 and IgG2 antibodies to PcPs 4, 6B, 9V and 14 by enumerating antibody secreting cells (ASCs) at day (D) 7 and determining fold-increase in serum antibody levels at D28 after vaccination with unconjugated PcPs in HIV seronegative subjects (n = 20) and in HIV patients who were receiving antiretroviral therapy (ART) (n = 28) or who were ART-naive (n = 11) and determined their association with ICOS+ and ICOS- circulating memory TFH (cmTFH) cells (CD4+CD45RA-CD27+CXCR5+PD-1+) and short lived plasmablasts (SPBs) at D7, and with PcP-specific and total IgM+ and IgG+ memory B cells at D0. In HIV seronegative subjects, production of IgG1+ and IgG2+ ASCs was consistently associated with the frequency of ICOS+ cmTFH cells but not ICOS- cmTFH cells or memory B cells. In contrast, post-vaccination ASCs in HIV patients, regardless of ART status, were lower than in HIV seronegative subjects and not associated with ICOS+ cmTFH cells, the expansion of which was absent (ART-naive patients) or much lower than in HIV seronegative subjects (ART-treated patients). Production of SPBs was also lower in ART-naive patients. Fold-increase in IgG2 antibodies at D28 also correlated with ICOS+ cmTFH cells at D7 in HIV seronegative subjects but not in HIV patients. These novel findings provide evidence that ICOS+ cmTFH cells contribute to the regulation of PcP-specific IgG antibody responses, including isotype diversification, and that TFH cell dysfunction may be a cause of impaired PcP-specific IgG antibody responses and increased susceptibility to pneumococcal disease in HIV patients.
Collapse
|
39
|
Frossi B, Tripodo C, Guarnotta C, Carroccio A, De Carli M, De Carli S, Marino M, Calabrò A, Pucillo CE. Mast cells are associated with the onset and progression of celiac disease. J Allergy Clin Immunol 2017; 139:1266-1274.e1. [DOI: 10.1016/j.jaci.2016.08.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 07/19/2016] [Accepted: 08/08/2016] [Indexed: 02/08/2023]
|
40
|
Jaworski JP, Bryk P, Brower Z, Zheng B, Hessell AJ, Rosenberg AF, Wu TT, Sanz I, Keefer MC, Haigwood NL, Kobie JJ. Pre-existing neutralizing antibody mitigates B cell dysregulation and enhances the Env-specific antibody response in SHIV-infected rhesus macaques. PLoS One 2017; 12:e0172524. [PMID: 28222180 PMCID: PMC5319772 DOI: 10.1371/journal.pone.0172524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 02/06/2017] [Indexed: 01/03/2023] Open
Abstract
Our central hypothesis is that protection against HIV infection will be powerfully influenced by the magnitude and quality of the B cell response. Although sterilizing immunity, mediated by pre-formed abundant and potent antibodies is the ultimate goal for B cell-targeted HIV vaccine strategies, scenarios that fall short of this may still confer beneficial defenses against viremia and disease progression. We evaluated the impact of sub-sterilizing pre-existing neutralizing antibody on the B cell response to SHIV infection. Adult male rhesus macaques received passive transfer of a sub-sterilizing amount of polyclonal neutralizing immunoglobulin (Ig) purified from previously infected animals (SHIVIG) or control Ig prior to intra-rectal challenge with SHIVSF162P4 and extensive longitudinal sampling was performed. SHIVIG treated animals exhibited significantly reduced viral load and increased de novo Env-specific plasma antibody. Dysregulation of the B cell profile was grossly apparent soon after infection in untreated animals; exemplified by a ≈50% decrease in total B cells in the blood evident 2-3 weeks post-infection which was not apparent in SHIVIG treated animals. IgD+CD5+CD21+ B cells phenotypically similar to marginal zone-like B cells were highly sensitive to SHIV infection, becoming significantly decreased as early as 3 days post-infection in control animals, while being maintained in SHIVIG treated animals, and were highly correlated with the induction of Env-specific plasma antibody. These results suggest that B cell dysregulation during the early stages of infection likely contributes to suboptimal Env-specific B cell and antibody responses, and strategies that limit this dysregulation may enhance the host's ability to eliminate HIV.
Collapse
Affiliation(s)
- Juan Pablo Jaworski
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Peter Bryk
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Zachary Brower
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Bo Zheng
- Division of Infectious Diseases, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Ann J. Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Alexander F. Rosenberg
- Divsion of Allergy, Immunology & Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Tong Tong Wu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Ignacio Sanz
- Lowance Center for Human Immunology and Division of Rheumatology, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Michael C. Keefer
- Division of Infectious Diseases, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - James J. Kobie
- Division of Infectious Diseases, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
41
|
García M, Iglesias A, Landoni VI, Bellomo C, Bruno A, Córdoba MT, Balboa L, Fernández GC, Sasiain MDC, Martínez VP, Schierloh P. Massive plasmablast response elicited in the acute phase of hantavirus pulmonary syndrome. Immunology 2017; 151:122-135. [PMID: 28106253 DOI: 10.1111/imm.12713] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 12/22/2016] [Accepted: 01/11/2017] [Indexed: 12/26/2022] Open
Abstract
Beside its key diagnostic value, the humoral immune response is thought to play a protective role in hantavirus pulmonary syndrome. However, little is known about the cell source of these antibodies during ongoing human infection. Herein we characterized B-cell subsets circulating in Andes-virus-infected patients. A notable potent plasmablast (PB) response that increased 100-fold over the baseline levels was observed around 1 week after the onset of symptoms. These PB present a CD3neg CD19low CD20neg CD38hi CD27hi CD138+/- IgA+/- surface phenotype together with the presence of cytoplasmic functional immunoglobulins. They are large lymphocytes (lymphoblasts) morphologically coincident with the 'immunoblast-like' cells that have been previously described during blood cytology examinations of hantavirus-infected patients. Immunoreactivity analysis of white blood cell lysates suggests that some circulating PB are virus-specific but we also observed a significant increase of reactivity against virus-unrelated antigens, which suggests a possible bystander effect by polyclonal B-cell activation. The presence of this large and transient PB response raises the question as to whether these cells might have a protective or pathological role during the ongoing hantavirus pulmonary syndrome and suggest their practical application as a diagnostic/prognostic biomarker.
Collapse
Affiliation(s)
- Marina García
- Laboratorio de Inmunología de Enfermedades Respiratorias, Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Ciudad autónoma de Buenos Aires, Argentina
| | - Ayelén Iglesias
- Laboratorio Nacional de Referencia para Hantavirus, Servicio de Biología Molecular, Instituto Nacional de Enfermedades Infecciosas, ANLIS -'Dr. Carlos G. Malbrán', Ciudad autónoma de Buenos Aires, Argentina
| | - Verónica I Landoni
- Laboratorio de Fisiología de procesos Inflamatorios, IMEX-CONICET-ANM, Ciudad autónoma de Buenos Aires, Argentina
| | - Carla Bellomo
- Laboratorio Nacional de Referencia para Hantavirus, Servicio de Biología Molecular, Instituto Nacional de Enfermedades Infecciosas, ANLIS -'Dr. Carlos G. Malbrán', Ciudad autónoma de Buenos Aires, Argentina
| | - Agostina Bruno
- Laboratorio de Enfermedades Tropicales, Hospital San Vicente de Paúl, Orán, Salta, Argentina
| | - María Teresa Córdoba
- Laboratorio de Enfermedades Tropicales, Hospital San Vicente de Paúl, Orán, Salta, Argentina
| | - Luciana Balboa
- Laboratorio de Inmunología de Enfermedades Respiratorias, Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Ciudad autónoma de Buenos Aires, Argentina
| | - Gabriela C Fernández
- Laboratorio de Fisiología de procesos Inflamatorios, IMEX-CONICET-ANM, Ciudad autónoma de Buenos Aires, Argentina
| | - María Del Carmen Sasiain
- Laboratorio de Inmunología de Enfermedades Respiratorias, Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Ciudad autónoma de Buenos Aires, Argentina
| | - Valeria P Martínez
- Laboratorio Nacional de Referencia para Hantavirus, Servicio de Biología Molecular, Instituto Nacional de Enfermedades Infecciosas, ANLIS -'Dr. Carlos G. Malbrán', Ciudad autónoma de Buenos Aires, Argentina
| | - Pablo Schierloh
- Laboratorio de Inmunología de Enfermedades Respiratorias, Instituto de Medicina Experimental (IMEX)-CONICET-Academia Nacional de Medicina (ANM), Ciudad autónoma de Buenos Aires, Argentina
| |
Collapse
|
42
|
Williams LD, Ofek G, Schätzle S, McDaniel JR, Lu X, Nicely NI, Wu L, Lougheed CS, Bradley T, Louder MK, McKee K, Bailer RT, O'Dell S, Georgiev IS, Seaman MS, Parks RJ, Marshall DJ, Anasti K, Yang G, Nie X, Tumba NL, Wiehe K, Wagh K, Korber B, Kepler TB, Munir Alam S, Morris L, Kamanga G, Cohen MS, Bonsignori M, Xia SM, Montefiori DC, Kelsoe G, Gao F, Mascola JR, Moody MA, Saunders KO, Liao HX, Tomaras GD, Georgiou G, Haynes BF. Potent and broad HIV-neutralizing antibodies in memory B cells and plasma. Sci Immunol 2017; 2:2/7/eaal2200. [PMID: 28783671 DOI: 10.1126/sciimmunol.aal2200] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/20/2016] [Indexed: 12/13/2022]
Abstract
Induction of broadly neutralizing antibodies (bnAbs) is a goal of HIV-1 vaccine development. Antibody 10E8, reactive with the distal portion of the membrane-proximal external region (MPER) of HIV-1 gp41, is broadly neutralizing. However, the ontogeny of distal MPER antibodies and the relationship of memory B cell to plasma bnAbs are poorly understood. HIV-1-specific memory B cell flow sorting and proteomic identification of anti-MPER plasma antibodies from an HIV-1-infected individual were used to isolate broadly neutralizing distal MPER bnAbs of the same B cell clonal lineage. Structural analysis demonstrated that antibodies from memory B cells and plasma recognized the envelope gp41 bnAb epitope in a distinct orientation compared with other distal MPER bnAbs. The unmutated common ancestor of this distal MPER bnAb was autoreactive, suggesting lineage immune tolerance control. Construction of chimeric antibodies of memory B cell and plasma antibodies yielded a bnAb that potently neutralized most HIV-1 strains.
Collapse
Affiliation(s)
- LaTonya D Williams
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gilad Ofek
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Sebastian Schätzle
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Jonathan R McDaniel
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Xiaozhi Lu
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Nathan I Nicely
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Liming Wu
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Caleb S Lougheed
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Todd Bradley
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mark K Louder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| | - Krisha McKee
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| | - Robert T Bailer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| | - Sijy O'Dell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| | - Ivelin S Georgiev
- Vanderbilt Vaccine Center and Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Michael S Seaman
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Robert J Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Dawn J Marshall
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kara Anasti
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Guang Yang
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaoyan Nie
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Nancy L Tumba
- Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg 2131, South Africa.,Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Congella 4013, South Africa
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kshitij Wagh
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Bette Korber
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Thomas B Kepler
- Departments of Microbiology and Mathematics & Statistics, Boston University School of Medicine, Boston, MA 02118, USA
| | - S Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lynn Morris
- Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg 2131, South Africa.,Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Congella 4013, South Africa
| | - Gift Kamanga
- University of North Carolina Project-Malawi, Kamuzu Central Hospital, Lilongwe, Malawi
| | - Myron S Cohen
- Departments of Medicine, Epidemiology, and Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Mattia Bonsignori
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Shi-Mao Xia
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - David C Montefiori
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Garnett Kelsoe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Feng Gao
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hua-Xin Liao
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA. .,Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA. .,Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
43
|
Jaworski JP, Vendrell A, Chiavenna SM. Neutralizing Monoclonal Antibodies to Fight HIV-1: On the Threshold of Success. Front Immunol 2017; 7:661. [PMID: 28123384 PMCID: PMC5225137 DOI: 10.3389/fimmu.2016.00661] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 12/16/2016] [Indexed: 12/21/2022] Open
Abstract
Anti-human immunodeficiency virus type-1 (anti-HIV-1) neutralizing monoclonal antibodies are broadening the spectrum of pre- and post-exposure treatment against HIV-1. A better understanding of how these antibodies develop and interact with particular regions of the viral envelope protein is guiding a more rational structure-based immunogen design. The aim of this article is to review the most recent advances in the field, from the development of these particular antibodies during natural HIV-1 infection, to their role preventing infection, boosting endogenous immune responses and clearing both free viral particles and persistently infected cells.
Collapse
Affiliation(s)
- Juan Pablo Jaworski
- National Scientific and Technical Research Council, Buenos Aires, Argentina; Institute of Virology, National Institute of Agricultural Technology, Castelar, Buenos Aires, Argentina
| | - Alejandrina Vendrell
- Pharmacological and Botanical Study Center, School of Medicine, University of Buenos Aires , Buenos Aires , Argentina
| | | |
Collapse
|
44
|
Abstract
The induction of neutralizing antibodies directed against the human immunodeficiency virus (HIV) has received considerable attention in recent years, in part driven by renewed interest and opportunities for antibody-based strategies for prevention such as passive transfer of antibodies and the development of preventive vaccines, as well as immune-based therapeutic interventions. Advances in the ability to screen, isolate, and characterize HIV-specific antibodies have led to the identification of a new generation of potent broadly neutralizing antibodies (bNAbs). The majority of these antibodies have been isolated from B cells of chronically HIV-infected individuals with detectable viremia. In this review, we provide insight into the phenotypic and functional attributes of human B cells, with a focus on HIV-specific memory B cells and plasmablasts/cells that are responsible for sustaining humoral immune responses against HIV. We discuss the abnormalities in B cells that occur in HIV infection both in the peripheral blood and lymphoid tissues, especially in the setting of persisting viremia. Finally, we consider the opportunities and drawbacks of intensively interrogating antibodies isolated from HIV-infected individuals to guide strategies aimed at developing effective antibody-based vaccine and therapeutic interventions for HIV.
Collapse
Affiliation(s)
- Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| | - Anthony S. Fauci
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, 20892
| |
Collapse
|
45
|
Verkoczy L. Humanized Immunoglobulin Mice: Models for HIV Vaccine Testing and Studying the Broadly Neutralizing Antibody Problem. Adv Immunol 2017; 134:235-352. [PMID: 28413022 PMCID: PMC5914178 DOI: 10.1016/bs.ai.2017.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A vaccine that can effectively prevent HIV-1 transmission remains paramount to ending the HIV pandemic, but to do so, will likely need to induce broadly neutralizing antibody (bnAb) responses. A major technical hurdle toward achieving this goal has been a shortage of animal models with the ability to systematically pinpoint roadblocks to bnAb induction and to rank vaccine strategies based on their ability to stimulate bnAb development. Over the past 6 years, immunoglobulin (Ig) knock-in (KI) technology has been leveraged to express bnAbs in mice, an approach that has enabled elucidation of various B-cell tolerance mechanisms limiting bnAb production and evaluation of strategies to circumvent such processes. From these studies, in conjunction with the wealth of information recently obtained regarding the evolutionary pathways and paratopes/epitopes of multiple bnAbs, it has become clear that the very features of bnAbs desired for their function will be problematic to elicit by traditional vaccine paradigms, necessitating more iterative testing of new vaccine concepts. To meet this need, novel bnAb KI models have now been engineered to express either inferred prerearranged V(D)J exons (or unrearranged germline V, D, or J segments that can be assembled into functional rearranged V(D)J exons) encoding predecessors of mature bnAbs. One encouraging approach that has materialized from studies using such newer models is sequential administration of immunogens designed to bind progressively more mature bnAb predecessors. In this review, insights into the regulation and induction of bnAbs based on the use of KI models will be discussed, as will new Ig KI approaches for higher-throughput production and/or altering expression of bnAbs in vivo, so as to further enable vaccine-guided bnAb induction studies.
Collapse
Affiliation(s)
- Laurent Verkoczy
- Duke University Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States.
| |
Collapse
|
46
|
Hel Z, Xu J, Denning WL, Helton ES, Huijbregts RPH, Heath SL, Overton ET, Christmann BS, Elson CO, Goepfert PA, Mestecky J. Dysregulation of Systemic and Mucosal Humoral Responses to Microbial and Food Antigens as a Factor Contributing to Microbial Translocation and Chronic Inflammation in HIV-1 Infection. PLoS Pathog 2017; 13:e1006087. [PMID: 28125732 PMCID: PMC5268400 DOI: 10.1371/journal.ppat.1006087] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 11/23/2016] [Indexed: 01/25/2023] Open
Abstract
HIV-1 infection is associated with an early and profound depletion of mucosal memory CD4+ T cells, a population that plays an indispensable role in the regulation of isotype switching and transepithelial transport of antibodies. In this study, we addressed whether the depletion of CD4+ T cell in HIV-1-infected individuals results in altered humoral responses specific to antigens encountered at mucosal surfaces. Comprehensive protein microarray of systemic humoral responses to intestinal microbiota demonstrated reduced IgG responses to antigens derived from Proteobacteria and Firmicutes but not Bacteroidetes. Importantly, intestinal secretions of antiretroviral therapy-treated HIV-1-infected individuals exhibited a significant elevation of IgM levels and decreased IgA/IgM and IgG/IgM ratios of antibodies specific to a variety of microbial and food antigens. The presented findings indicate reduced competence of mucosal B cells for class switch recombination from IgM to other isotypes limiting their capacity to react to changing antigenic variety in the gut lumen. Decreased availability of microbiota-specific IgA and IgG may be an important factor contributing to the translocation of microbial antigens across the intestinal mucosal barrier and their systemic dissemination that drives chronic inflammation in HIV-1-infected individuals.
Collapse
Affiliation(s)
- Zdenek Hel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jun Xu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Warren L. Denning
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - E. Scott Helton
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Richard P. H. Huijbregts
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Sonya L. Heath
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - E. Turner Overton
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Benjamin S. Christmann
- Department of Natural Science and Mathematics, Lee University, Cleveland, Tennessee, United States of America
| | - Charles O. Elson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Paul A. Goepfert
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jiri Mestecky
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Institute of Immunology and Microbiology, 1 School of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
47
|
Tanko RF, Soares AP, Müller TL, Garrett NJ, Samsunder N, Abdool Karim Q, Abdool Karim SS, Riou C, Burgers WA. Effect of Antiretroviral Therapy on the Memory and Activation Profiles of B Cells in HIV-Infected African Women. THE JOURNAL OF IMMUNOLOGY 2016; 198:1220-1228. [PMID: 28039305 DOI: 10.4049/jimmunol.1601560] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/30/2016] [Indexed: 12/25/2022]
Abstract
Human immunodeficiency virus infection induces a wide range of effects in B cells, including skewed memory cell differentiation, compromised B cell function, and hypergammaglobulinemia. However, data on the extent to which these B cell abnormalities can be reversed by antiretroviral therapy (ART) are limited. To investigate the effect of ART on B cells, the activation (CD86) and differentiation (IgD, CD27, and CD38) profiles of B cells were measured longitudinally in 19 HIV-infected individuals before (median, 2 mo) and after ART initiation (median, 12 mo) and compared with 19 age-matched HIV-uninfected individuals using flow cytometry. Twelve months of ART restored the typical distribution of B cell subsets, increasing the proportion of naive B cells (CD27-IgD+CD38-) and concomitantly decreasing the immature transitional (CD27-IgD+CD38+), unswitched memory (CD27+IgD+CD38-), switched memory (CD27+IgD-CD38- or CD27-IgD-CD38-), and plasmablast (CD27+IgD-CD38high) subsets. However, B cell activation was only partially normalized post-ART, with the frequency of activated B cells (CD86+CD40+) reduced compared with pre-ART levels (p = 0.0001), but remaining significantly higher compared with HIV-uninfected individuals (p = 0.0001). Interestingly, unlike for T cell activation profiles, the extent of B cell activation prior to ART did not correlate with HIV plasma viral load, but positively associated with plasma sCD14 levels (p = 0.01, r = 0.58). Overall, ART partially normalizes the skewed B cell profiles induced by HIV, with some activation persisting. Understanding the effects of HIV on B cell dysfunction and restoration following ART may provide important insights into the mechanisms of HIV pathogenesis.
Collapse
Affiliation(s)
- Ramla F Tanko
- Department of Pathology, Division of Medical Virology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Andreia P Soares
- Department of Pathology, Division of Medical Virology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Tracey L Müller
- Department of Pathology, Division of Medical Virology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Nigel J Garrett
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban 4013, South Africa
| | - Natasha Samsunder
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban 4013, South Africa
| | - Quarraisha Abdool Karim
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban 4013, South Africa.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY 10032; and
| | - Salim S Abdool Karim
- Centre for the AIDS Program of Research in South Africa, University of KwaZulu-Natal, Durban 4013, South Africa.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY 10032; and
| | - Catherine Riou
- Department of Pathology, Division of Medical Virology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa
| | - Wendy A Burgers
- Department of Pathology, Division of Medical Virology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa; .,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|
48
|
Control of early HIV-1 infection associates with plasmacytoid dendritic cell-reactive opsonophagocytic IgG antibodies to HIV-1 p24. AIDS 2016; 30:2757-2765. [PMID: 27603291 DOI: 10.1097/qad.0000000000001242] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES We have previously demonstrated that HIV-1 p24-specific plasmacytoid dendritic cell-reactive opsonophagocytic antibody (PROAb) responses associate with control of chronic HIV infection. Here, we examined whether HIV-1 p24-specific PROAbs associate with control of early HIV infection and their relationship with HIV-1 p24-specific IgG subclasses. METHODS Plasma collected at 8 and 52 weeks following primary HIV-1 infection was obtained from antiretroviral therapy-naïve patients who were classified as 'good' (plasma HIV-1 RNA < 5000 copies/ml; n = 17) or 'poor' (HIV-1 RNA > 50 000 copies/ml; n = 15) controllers at week 52. HIV-1 p24-specific PROAb responses were assayed using a plasmacytoid dendritic cell line (Gen2.2), and HIV-1 p24-specific IgG3, IgG1 and IgG2 levels were assayed by ELISA. RESULTS HIV-1 p24-specific PROAb responses increased in 'good controllers' (P = 0.01) but remained unchanged in 'poor controllers' between weeks 8 and 52. Of the HIV-1 p24-specific IgG subclasses measured, only IgG1 increased over this time period in 'good controllers' alone (P = 0.003), which correlated with the increase in HIV-1 p24-specific PROAb responses (r = 0.83, P < 0.0001). Depletion of IgG1 from IgG preparations of 'good controllers' resulted in the inhibition of HIV-1 p24-specific PROAb responses. In the total patient cohort, plasma HIV-1 RNA levels at week 52 correlated inversely with changes in HIV-1 p24-specific PROAb responses (r = -0.37, P = 0.04) and IgG1 (r = -0.51, P = 0.003) levels between weeks 8 and 52. CONCLUSION Control of early HIV-1 infection was associated with an increase in HIV-1 p24-specific PROAb responses, which was mediated by HIV-1 p24-specific IgG1 antibodies. These findings provide further evidence that antibodies to HIV core proteins may contribute to control of HIV-1 infection.
Collapse
|
49
|
Yamamoto H, Matano T. Patterns of HIV/SIV Prevention and Control by Passive Antibody Immunization. Front Microbiol 2016; 7:1739. [PMID: 27853456 PMCID: PMC5089984 DOI: 10.3389/fmicb.2016.01739] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 10/18/2016] [Indexed: 01/04/2023] Open
Abstract
Neutralizing antibody (NAb) responses are promising immune effectors for control of human immunodeficiency virus (HIV) infection. Protective activity and mechanisms of immunodeficiency virus-specific NAbs have been increasingly scrutinized in animals infected with simian immunodeficiency virus (SIV), chimeric simian/human immunodeficiency virus (SHIV) and related viruses. Studies on such models have unraveled a previously underscored protective potential against in vivo immunodeficiency virus replication. Pre-challenge NAb titers feasibly provide sterile protection from SIV/SHIV infection by purging the earliest onset of viral replication and likely modulate innate immune cell responses. Sufficient sub-sterile NAb titers after established infection also confer dose-dependent reduction of viremia, and in certain earlier time frames augment adaptive immune cell responses and even provide rebound-free viral control. Here, we provide an overview of the obtained patterns of SIV/SHIV protection and viral control by various types of NAb passive immunizations and discuss how these notions may be extrapolated to NAb-based clinical control of HIV infection.
Collapse
Affiliation(s)
- Hiroyuki Yamamoto
- AIDS Research Center, National Institute of Infectious Diseases Tokyo, Japan
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious DiseasesTokyo, Japan; Department of AIDS Vaccine, The Institute of Medical Science, The University of TokyoTokyo, Japan
| |
Collapse
|
50
|
Karris MY, Umlauf A, Vaida F, Richman D, Little S, Smith D. A randomized controlled clinical trial on the impact of CCR5 blockade with maraviroc in early infection on T-cell dynamics. Medicine (Baltimore) 2016; 95:e5315. [PMID: 27858912 PMCID: PMC5591160 DOI: 10.1097/md.0000000000005315] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Initiation of antiretroviral therapy (ART) in early HIV infection demonstrates clinical benefits including enhanced CD4 T-lymphocyte recovery and minimization of the latent HIV reservoir. Whether ART intensification with CCR5 blockade provides additional benefits is unknown. TRIAL DESIGN This randomized controlled trial evaluated the impact of maraviroc (MVC) intensification in persons starting ART in acute and early HIV (AEH, within 3 months of estimated date of infection). METHODS Twenty persons in AEH in San Diego underwent double-blind randomization to receive either standard of care (SOC) ART or SOC + MVC to evaluate the hypothesis that early CCR5 blockage with a CCR5-containing ART regimen may provide immunologic and clinical benefit. The primary outcome of this study was the difference from baseline to week 48 in the proportion of CCR5 CD4 memory T cells. Blood was drawn at baseline and weeks 12, 24, and 48 to evaluate CCR5 CD4 and CD8 T-cell dynamics using multicolor flow cytometry. RESULTS MVC intensification (n = 10) did not significantly alter CCR5 T-cell dynamics at week 48 of study compared to SOC (n = 9) in this fully recruited study (mean 1.12 vs 0.63, t = 0.36, df = 16, P = 0.727). Exploratory analyses of additional T-cell subsets suggest that MVC intensification in AEH trended to early greater increases in naïve and activated and proliferating CD4 T cells (P = 0.11, 0.19), and greater decreases in senescent memory CD4 T cells (P = 0.06), but these differences did not remain by week 48. CD8 T-cell evaluations did demonstrate trends to differences at week 48 with slower increases in naïve cells and slower decreases in activated memory cells (P = 0.16, 0.09). There were no reported harms or significantly different adverse events. CONCLUSIONS We did observe a few trends, but did not find compelling evidence that MVC intensification during AEH significantly impacts CD4 and CD8 T-cell dynamics. Diagnosing and starting persons in AEH on ART may be of greater clinical importance than the regimen initiated.
Collapse
Affiliation(s)
- Maile Y. Karris
- University of California San Diego
- Correspondence: Maile Y. Karris, University of California San Diego, 200 West Arbor Drive #8208, San Diego, CA 92103 (e-mail: )
| | | | | | - Douglas Richman
- University of California San Diego
- Veterans Affairs Medical Center, San Diego, CA
| | | | - Davey Smith
- University of California San Diego
- Veterans Affairs Medical Center, San Diego, CA
| |
Collapse
|