1
|
Loe-Sack-Sioe GE, de Vos DW, Visser LG, Jochems SP, Roukens AHE. Pneumococcal vaccine hyporesponsiveness in people living with HIV: A narrative review of immunological mechanisms and insights from minimally invasive lymph node sampling. Hum Vaccin Immunother 2025; 21:2503602. [PMID: 40374620 PMCID: PMC12087491 DOI: 10.1080/21645515.2025.2503602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/11/2025] [Accepted: 05/06/2025] [Indexed: 05/17/2025] Open
Abstract
Despite highly effective antiretroviral therapy, people living with HIV (PLWH) remain at elevated risk for invasive pneumococcal disease. Clinical studies show that, even with high CD4+ counts, PLWH exhibit diminished serological responses and rapid antibody decline following pneumococcal vaccination, plausibly due to underlying immune dysfunction. Germinal centers (GCs), located within lymph nodes, are essential for generating high-affinity antibodies, but are structurally and functionally disrupted in PLWH. These local impairments, combined with systemic immune dysregulation, contribute to vaccine hyporesponsiveness in PLWH. This narrative review links immunological findings from experimental and in vivo studies to clinical pneumococcal vaccine trials, to investigate mechanisms that may be leveraged to strengthen vaccine-induced immunity in PLWH. We also highlight the application of fine needle aspiration (FNA) of the lymph node as a way to study pneumococcal vaccine hyporesponsiveness in the GC and provide potential direction to improve responses for next-generation pneumococcal conjugate vaccines in PLWH.
Collapse
Affiliation(s)
- Giovanni E. Loe-Sack-Sioe
- Center for Infectious Diseases, Subdepartment of Research, Leiden University, Leiden, The Netherlands
| | - Danny W. de Vos
- Center for Infectious Diseases, Subdepartment of Infectious Diseases, Leiden University, Leiden, The Netherlands
| | - Leo G. Visser
- Center for Infectious Diseases, Subdepartment of Infectious Diseases, Leiden University, Leiden, The Netherlands
| | - Simon P. Jochems
- Center for Infectious Diseases, Subdepartment of Research, Leiden University, Leiden, The Netherlands
| | - Anna H. E. Roukens
- Center for Infectious Diseases, Subdepartment of Infectious Diseases, Leiden University, Leiden, The Netherlands
| |
Collapse
|
2
|
Mitchell JL, Buranapraditkun S, Gantner P, Takata H, Dietze K, N'guessan KF, Pollara J, Nohara J, Muir R, Kroon E, Pinyakorn S, Tulmethakaan N, Manasnayakorn S, Chottanapund S, Thantiworasit P, Prueksakaew P, Ratnaratorn N, Puttamaswin S, Nuntapinit B, Fox L, Haddad EK, Paquin-Proulx D, Phanuphak P, Sacdalan CP, Phanuphak N, Ananworanich J, Hsu D, Vasan S, Ferrari G, Chomont N, Trautmann L, on behalf of RV254 and RV304 Study Groups. Activation of CXCR3 + Tfh cells and B cells in lymph nodes during acute HIV-1 infection correlates with HIV-specific antibody development. J Virol 2025; 99:e0153224. [PMID: 39932316 PMCID: PMC11915809 DOI: 10.1128/jvi.01532-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/17/2025] [Indexed: 03/19/2025] Open
Abstract
Lymph node T follicular helper (Tfh) cells and germinal center (GC) B cells are critical to generate potent antibodies but are rarely possible to study in humans. To understand how Tfh/GC B-cell interactions during acute HIV-1 infection (AHI) impact the generation of HIV-specific antibodies, we performed a unique cross-sectional analysis of inguinal lymph node biopsies taken prior to antiretroviral therapy (ART) initiation in AHI. Although total Tfh and GC B cell frequencies did not change during AHI, increased frequencies of proliferating Th1-like CXCR3+ Tfh, CXCR3+ non-GC B cells, and total CXCR3+ GC B cells correlated with gp120-specific IgG antibody levels in AHI. Frequencies of proliferating CXCR3+ Tfh in AHI also correlated with gp120-specific IgG antibody levels after 48 weeks of ART, antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and increased antibody binding to infected cells after ART. Importantly, while beneficial for antibody development, CXCR3+ Tfh cells were also infected by HIV-1 at higher frequencies than their CXCR3- counterparts and may contribute to the initial dissemination of HIV-1 in follicles. Together, these data suggest that activation of CXCR3+ Tfh cells is associated with induction of the germinal center response and subsequent antibody development, making these cells an important target for future therapeutic interventions. IMPORTANCE Early initiation of antiretroviral therapy (ART) is important to limit the seeding of the long-lasting HIV-1 reservoir; however, it also precludes the development of HIV-specific antibodies that can help control the virus if ART is stopped. Antibody development occurs within germinal centers in the lymph node and requires activation of both antigen-specific B cells and T follicular helper cells (Tfh), a specialized CD4+ cell that provides B cell help. To understand how early ART initiation may prohibit antibody development, we analyzed the frequencies and activation status of Tfh and B cells in lymph node biopsies collected in the different stages of acute HIV-1 infection. Our data suggest that decreased antibody development after early ART initiation may be due to limited germinal center development at the time of treatment and that new interventions that target activation of CXCR3+ Tfh may be beneficial to increase long-term HIV-specific antibody levels.
Collapse
Affiliation(s)
- Julie L. Mitchell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Supranee Buranapraditkun
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Thai Pediatric Gastroenterology, Hepatology and Immunology (TPGHAI) Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pierre Gantner
- Centre de Recherche du CHUM (CRCHUM) and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Québec, Canada
| | - Hiroshi Takata
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Kenneth Dietze
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Kombo F. N'guessan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Justin Pollara
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Junsuke Nohara
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Roshell Muir
- Division of Infectious Diseases & HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Suteeraporn Pinyakorn
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | - Sopark Manasnayakorn
- Department of Surgery, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Pattarawat Thantiworasit
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | | | | | - Bessara Nuntapinit
- Armed Forces Research Institute of Medical Sciences in Bangkok, Bangkok, Thailand
| | - Lawrence Fox
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Elias K. Haddad
- Division of Infectious Diseases & HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Dominic Paquin-Proulx
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | - Carlo P. Sacdalan
- SEARCH Research Foundation, Bangkok, Thailand
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Jintanat Ananworanich
- Department of Global Health, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| | - Denise Hsu
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Sandhya Vasan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Guido Ferrari
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Nicolas Chomont
- Centre de Recherche du CHUM (CRCHUM) and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Québec, Canada
| | - Lydie Trautmann
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - on behalf of RV254 and RV304 Study Groups
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Thai Pediatric Gastroenterology, Hepatology and Immunology (TPGHAI) Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Centre de Recherche du CHUM (CRCHUM) and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Québec, Canada
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Division of Infectious Diseases & HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
- SEARCH Research Foundation, Bangkok, Thailand
- Department of Surgery, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Armed Forces Research Institute of Medical Sciences in Bangkok, Bangkok, Thailand
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Institute of HIV Research and Innovation (IHRI), Bangkok, Thailand
- Department of Global Health, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| |
Collapse
|
3
|
Yamamoto H, Matano T. SIV-specific neutralizing antibody induction following selection of a PI3K drive-attenuated nef variant. eLife 2025; 12:RP88849. [PMID: 40029304 PMCID: PMC11875539 DOI: 10.7554/elife.88849] [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] [Indexed: 03/05/2025] Open
Abstract
HIV and simian immunodeficiency virus (SIV) infections are known for impaired neutralizing antibody (NAb) responses. While sequential virus-host B cell interaction appears to be basally required for NAb induction, driver molecular signatures predisposing to NAb induction still remain largely unknown. Here we describe SIV-specific NAb induction following a virus-host interplay decreasing aberrant viral drive of phosphoinositide 3-kinase (PI3K). Screening of seventy difficult-to-neutralize SIVmac239-infected macaques found nine NAb-inducing animals, with seven selecting for a specific CD8+ T-cell escape mutation in viral nef before NAb induction. This Nef-G63E mutation reduced excess Nef interaction-mediated drive of B-cell maturation-limiting PI3K/mammalian target of rapamycin complex 2 (mTORC2). In vivo imaging cytometry depicted preferential Nef perturbation of cognate Envelope-specific B cells, suggestive of polarized contact-dependent Nef transfer and corroborating cognate B-cell maturation post-mutant selection up to NAb induction. Results collectively exemplify a NAb induction pattern extrinsically reciprocal to human PI3K gain-of-function antibody-dysregulating disease and indicate that harnessing the PI3K/mTORC2 axis may facilitate NAb induction against difficult-to-neutralize viruses including HIV/SIV.
Collapse
Grants
- JP24fk0410066 Japan Agency for Medical Research and Development
- JP21jk0210002 Japan Agency for Medical Research and Development
- 24K21287 Ministry of Education, Culture, Sports, Science and Technology
- 21H02745 Ministry of Education, Culture, Sports, Science and Technology
- JP22wm0325006 Japan Agency for Medical Research and Development
- JP19fm0208017 Japan Agency for Medical Research and Development
- JP20fk0410022 Japan Agency for Medical Research and Development
- JP18fk0410003 Japan Agency for Medical Research and Development
- JP20fk0410011 Japan Agency for Medical Research and Development
- JP20fk0108125 Japan Agency for Medical Research and Development
- JP20jm0110012 Japan Agency for Medical Research and Development
- JP21fk0410035 Japan Agency for Medical Research and Development
- 17H02185 Ministry of Education, Culture, Sports, Science and Technology
- 18K07157 Ministry of Education, Culture, Sports, Science and Technology
- Takeda Science Foundation
- Imai Memorial Trust for AIDS Research
- Mitsui Sumitomo Insurance Welfare Foundation
Collapse
Affiliation(s)
- Hiroyuki Yamamoto
- AIDS Research Center, National Institute of Infectious DiseasesTokyoJapan
- Department of Biomedicine, University Hospital BaselBaselSwitzerland
- Joint Research Center for Human Retrovirus Infection, Kumamoto UniversityKumamotoJapan
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious DiseasesTokyoJapan
- Joint Research Center for Human Retrovirus Infection, Kumamoto UniversityKumamotoJapan
- The Institute of Medical Science, The University of TokyoTokyoJapan
| |
Collapse
|
4
|
Clain JA, Picard M, Rabezanahary H, André S, Boutrais S, Goma Matsetse E, Dewatines J, Dueymes Q, Thiboutot E, Racine G, Soundaramourty C, Mammano F, Corbeau P, Zghidi-Abouzid O, Estaquier J. Immune Alterations and Viral Reservoir Atlas in SIV-Infected Chinese Rhesus Macaques. Infect Dis Rep 2025; 17:12. [PMID: 39997464 PMCID: PMC11855486 DOI: 10.3390/idr17010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Over the last decades, our projects have been dedicated to clarifying immunopathological and virological events associated with Human Immunodeficiency Virus (HIV) infection. METHODS By using non-human primate models of pathogenic and non-pathogenic lentiviral infections, we aimed at identifying the cells and tissues in which the virus persists, despite antiretroviral therapy (ART). Indeed, the eradication of viral reservoirs is a major challenge for HIV cure. RESULTS We present a series of results performed in rhesus macaques of Chinese origin deciphering the virological and immunological events associated with ART that can be of interest for people living with HIV. CONCLUSIONS This model could be of interest for understanding in whole body the clinical alteration that persist despite ART.
Collapse
Affiliation(s)
- Julien A. Clain
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Morgane Picard
- Institut national de la santé et de la recherche médicale (INSERM) U1124, Université Paris Cité, 75006 Paris, France; (M.P.); (S.A.); (C.S.); (F.M.)
| | - Henintsoa Rabezanahary
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Sonia André
- Institut national de la santé et de la recherche médicale (INSERM) U1124, Université Paris Cité, 75006 Paris, France; (M.P.); (S.A.); (C.S.); (F.M.)
| | - Steven Boutrais
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Ella Goma Matsetse
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Juliette Dewatines
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Quentin Dueymes
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Elise Thiboutot
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Gina Racine
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Calaiselvy Soundaramourty
- Institut national de la santé et de la recherche médicale (INSERM) U1124, Université Paris Cité, 75006 Paris, France; (M.P.); (S.A.); (C.S.); (F.M.)
| | - Fabrizio Mammano
- Institut national de la santé et de la recherche médicale (INSERM) U1124, Université Paris Cité, 75006 Paris, France; (M.P.); (S.A.); (C.S.); (F.M.)
- Institut national de la santé et de la recherche médicale (Inserm) U1259 MAVIVHe, Université de Tours, 37032 Tours, France
| | - Pierre Corbeau
- Institut de Génétique Humaine, CNRS-Université de Montpellier UMR9002, 34094 Montpellier, France;
| | - Ouafa Zghidi-Abouzid
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
| | - Jérôme Estaquier
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (H.R.); (S.B.); (E.G.M.); (J.D.); (Q.D.); (E.T.); (G.R.); (O.Z.-A.)
- Institut national de la santé et de la recherche médicale (INSERM) U1124, Université Paris Cité, 75006 Paris, France; (M.P.); (S.A.); (C.S.); (F.M.)
| |
Collapse
|
5
|
Yong L, Hutchings C, Barnes E, Klenerman P, Provine NM. Distinct Requirements for CD4 + T Cell Help for Immune Responses Induced by mRNA and Adenovirus-Vector SARS-CoV-2 Vaccines. Eur J Immunol 2025; 55:e202451142. [PMID: 39604225 PMCID: PMC11739681 DOI: 10.1002/eji.202451142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
CD4+ T cells have been established as central orchestrators of cellular and humoral immune responses to infection or vaccination. However, the need for CD4+ T cell help to generate primary CD8+ T cell responses is variable depending on the infectious agent or vaccine and yet consistently required for the recall of CD8+ T cell memory responses or antibody responses. Given the deployment of new vaccine platforms such as nucleoside-modified mRNA vaccines, we sought to elucidate the requirement for CD4+ T cell help in the induction of cellular and antibody responses to mRNA and adenovirus (Ad)-vectored vaccines against SARS-CoV-2. Using antibody-mediated depletion of CD4+ T cells in a mouse immunization model, we observed that CD4+ T cell help was dispensable for both primary and secondary CD8+ T cell responses to the BNT162b2 and mRNA-1273 mRNA vaccines but required for the AZD1222 Ad-vectored vaccine. Nonetheless, CD4+ T cell help was needed by both mRNA and Ad-vectored vaccine platforms for the generation of antibodies, demonstrating the centrality of CD4+ T cells in vaccine-induced protective immunity against SARS-CoV-2. Ultimately, this highlights the shared and distinct regulation of humoral and cellular responses induced by these vaccine platforms.
Collapse
Affiliation(s)
- Lyn Yong
- Pandemic Sciences InstituteNuffield Department of MedicineUniversity of OxfordOxfordUK
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine–Experimental MedicineUniversity of OxfordOxfordUK
| | - Claire Hutchings
- Peter Medawar Building for Pathogen Research, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Eleanor Barnes
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine–Experimental MedicineUniversity of OxfordOxfordUK
- Peter Medawar Building for Pathogen Research, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Paul Klenerman
- Pandemic Sciences InstituteNuffield Department of MedicineUniversity of OxfordOxfordUK
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine–Experimental MedicineUniversity of OxfordOxfordUK
- Peter Medawar Building for Pathogen Research, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Nicholas M. Provine
- Pandemic Sciences InstituteNuffield Department of MedicineUniversity of OxfordOxfordUK
- Centre for Human GeneticsNuffield Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
6
|
Hu L, Sun C, Yuan K, Yang P. Expression, regulation, and function of PD-L1 on non-tumor cells in the tumor microenvironment. Drug Discov Today 2024; 29:104181. [PMID: 39278561 DOI: 10.1016/j.drudis.2024.104181] [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: 07/14/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
Antiprogrammed death ligand 1 (PD-L1) therapy is a leading immunotherapy, but only some patients with solid cancers benefit. Overwhelming evidence has revealed that PD-L1 is expressed on various immune cells in the tumor microenvironment (TME), including macrophages, dendritic cells, and regulatory T cells, modulating tumor immunity and influencing tumor progression. PD-L1 can also be located on tumor cell membranes as well as in exosomes and cytoplasm. Accordingly, the dynamic expression and various forms of PD-L1 might explain the therapy's limited efficacy and resistance. Herein a systematic summary of the expression of PD-L1 on different immune cells and their regulatory mechanisms is provided to offer a solid foundation for future studies.
Collapse
Affiliation(s)
- Lingrong Hu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Chengliang Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Kai Yuan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China.
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
7
|
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 PMCID: PMC11557871 DOI: 10.1126/sciimmunol.ado0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
|
8
|
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
|
9
|
Zhang Y, Chen A, Li D, Yuan Q, Zhu A, Deng J, Wang Y, Liu J, Liang C, Li W, Fang Q, Xie J, Zhang X, Zhang X, Zhang Y, Chen R, Pan T, Zhang H, He X. Development of T follicular helper cell-independent nanoparticle vaccines for SARS-CoV-2 or HIV-1 by targeting ICOSL. NPJ Vaccines 2024; 9:176. [PMID: 39341822 PMCID: PMC11438966 DOI: 10.1038/s41541-024-00971-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
T helper cells, particularly T follicular helper (TFH) cells, are essential for the neutralizing antibody production elicited by pathogens or vaccines. However, in immunocompromised individuals, the inefficient support from TFH cells could lead to limited protection after vaccine inoculation. Here we showed that the conjugation of inducible T cell costimulatory (ICOS) onto the nanoparticle, together with immunogen, significantly enhanced the immune response of the vaccines specific for SARS-CoV-2 or human immunodeficiency virus type-1 (HIV-1) in TFH-deficient mice. Further studies indicated that ICOSL on B cells was triggered by ICOS binding, subsequently activated the PKCβ signaling pathway, and enhanced the survival and proliferation of B cells. Our findings revealed that the stimulation of ICOS-ICOSL interaction by adding ICOS on the nanoparticle vaccine significantly substitutes the function of TFH cells to support B cell response, which is significant for the immunocompromised people, such as the elderly or HIV-1-infected individuals.
Collapse
Affiliation(s)
- Yongli Zhang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Achun Chen
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Daiying Li
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Quyu Yuan
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Airu Zhu
- Guangzhou Laboratory, Bio-island, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jieyi Deng
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yalin Wang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jie Liu
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chaofeng Liang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wenjie Li
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qiannan Fang
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Jiatong Xie
- Shenzhen College of International Education, No. 3 Antuoshan 6th Road, Futian District, Shenzhen, China
| | - Xiantao Zhang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xu Zhang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yiwen Zhang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ran Chen
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ting Pan
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Center for Infection and Immunity Study, School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Hui Zhang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangzhou Laboratory, Bio-island, Guangzhou, China
| | - Xin He
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
10
|
Han R, Su L, Cheng L. Advancing Human Vaccine Development Using Humanized Mouse Models. Vaccines (Basel) 2024; 12:1012. [PMID: 39340042 PMCID: PMC11436046 DOI: 10.3390/vaccines12091012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/11/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The development of effective vaccines against infectious diseases remains a critical challenge in global health. Animal models play a crucial role in vaccine development by providing valuable insights into the efficacy, safety, and mechanisms of immune response induction, which guide the design and formulation of vaccines. However, traditional animal models often inadequately recapitulate human immune responses. Humanized mice (hu-mice) models with a functional human immune system have emerged as invaluable tools in bridging the translational gap between preclinical research and clinical trials for human vaccine development. This review summarizes commonly used hu-mice models and advances in optimizing them to improve human immune responses. We review the application of humanized mice for human vaccine development with a focus on HIV-1 vaccines. We also discuss the remaining challenges and improvements needed for the currently available hu-mice models to better facilitate the development and testing of human vaccines for infectious diseases.
Collapse
Affiliation(s)
- Runpeng Han
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
- Center for AIDS Research, Wuhan University, Wuhan 430071, China
| | - Lishan Su
- Laboratory of Viral Pathogenesis and Immunotherapy, Institute of Human Virology, Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 02121, USA
| | - Liang Cheng
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
- Center for AIDS Research, Wuhan University, Wuhan 430071, China
| |
Collapse
|
11
|
Zaman F, Smith ML, Balagopal A, Durand CM, Redd AD, Tobian AAR. Spatial technologies to evaluate the HIV-1 reservoir and its microenvironment in the lymph node. mBio 2024; 15:e0190924. [PMID: 39058091 PMCID: PMC11324018 DOI: 10.1128/mbio.01909-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024] Open
Abstract
The presence of the HIV-1 reservoir, a group of immune cells that contain intact, integrated, and replication-competent proviruses, is a major challenge to cure HIV-1. HIV-1 reservoir cells are largely unaffected by the cytopathic effects of viruses, antiviral immune responses, or antiretroviral therapy (ART). The HIV-1 reservoir is seeded early during HIV-1 infection and augmented during active viral replication. CD4+ T cells are the primary target for HIV-1 infection, and recent studies suggest that memory T follicular helper cells within the lymph node, more precisely in the B cell follicle, harbor integrated provirus, which contribute to viral rebound upon ART discontinuation. The B cell follicle, more specifically the germinal center, possesses a unique environment because of its distinct property of being partly immune privileged, potentially allowing HIV-1-infected cells within the lymph nodes to be protected from CD8+ T cells. This modified immune response in the germinal center of the follicle is potentially explained by the exclusion of CD8+ T cells and the presence of T regulatory cells at the junction of the follicle and extrafollicular region. The proviral makeup of HIV-1-infected cells is similar in lymph nodes and blood, suggesting trafficking between these compartments. Little is known about the cell-to-cell interactions, microenvironment of HIV-1-infected cells in the follicle, and trafficking between the lymph node follicle and other body compartments. Applying a spatiotemporal approach that integrates genomics, transcriptomics, and proteomics to investigate the HIV-1 reservoir and its neighboring cells in the lymph node has promising potential for informing HIV-1 cure efforts.
Collapse
Affiliation(s)
- Fatima Zaman
- Department of
Pathology, Johns Hopkins University School of
Medicine, Baltimore,
Maryland, USA
| | - Melissa L. Smith
- Department of
Biochemistry and Molecular Genetics, University of Louisville School of
Medicine, Louisville,
Kentucky, USA
| | - Ashwin Balagopal
- Division of Infectious
Diseases, Department of Medicine, Johns Hopkins
University, Baltimore,
Maryland, USA
| | - Christine M. Durand
- Division of Infectious
Diseases, Department of Medicine, Johns Hopkins
University, Baltimore,
Maryland, USA
| | - Andrew D. Redd
- Division of Infectious
Diseases, Department of Medicine, Johns Hopkins
University, Baltimore,
Maryland, USA
- Laboratory of
Immunoregulation, National Institute of Allergy and Infectious Diseases,
National Institutes of Health,
Bethesda, Maryland, USA
- Institute of
Infectious Disease and Molecular Medicine, University of Cape
Town, Cape Town,
South Africa
| | - Aaron A. R. Tobian
- Department of
Pathology, Johns Hopkins University School of
Medicine, Baltimore,
Maryland, USA
- Division of Infectious
Diseases, Department of Medicine, Johns Hopkins
University, Baltimore,
Maryland, USA
| |
Collapse
|
12
|
Moysi E, Sharma AA, O’Dell S, Georgakis S, Del Rio Estrada PM, Torres-Ruiz F, Navarro MG, Villalobos YAL, Rios SA, Reyes-Teran G, Beddall MH, Ko SH, Belinky F, Orfanakis M, de Leval L, Enriquez AB, Buckner CM, Moir S, Doria-Rose N, Boritz E, Mascola JR, Sekaly RP, Koup RA, Petrovas C. Neutralization activity in chronic HIV infection is characterized by a distinct programming of follicular helper CD4 T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.605954. [PMID: 39131331 PMCID: PMC11312598 DOI: 10.1101/2024.07.31.605954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
A subset of people living with HIV (PLWH) can produce broadly neutralizing antibodies (bNAbs) against HIV, but the lymph node (LN) dynamics that promote the generation of these antibodies are poorly understood. Here, we explored LN-associated histological, immunological, and virological mechanisms of bNAb generation in a cohort of anti-retroviral therapy (ART)-naïve PLWH. We found that participants who produce bNAbs, termed neutralizers, have a superior LN-associated B cell follicle architecture compared with PLWH who do not. The latter was associated with a significantly higher in situ prevalence of Bcl-6hi follicular helper CD4 T cells (TFH), expressing a molecular program that favors their differentiation and stemness, and significantly reduced IL-10 follicular suppressor CD4 T cells. Furthermore, our data reveal possible molecular targets mediating TFH- B cell interactions in neutralizers. Together, we identify cellular and molecular mechanisms that contribute to the development of bNAbs in PLWH.
Collapse
Affiliation(s)
- Eirini Moysi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Ashish A. Sharma
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sijy O’Dell
- Virology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Spiros Georgakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Perla Mariana Del Rio Estrada
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Fernanda Torres-Ruiz
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Mauricio González Navarro
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico, Subdireccion de Otorrinolaringologia, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”
| | - Yara Andrea Luna Villalobos
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Santiago Avila Rios
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Gustavo Reyes-Teran
- Institutos Nacionales de Salud y Hospitales de Alta Especialidad, Secretaría de Salud de México, Mexico City, Mexico
| | - Margaret H. Beddall
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Sung-Hee Ko
- Virus Persistence and Dynamics Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Frida Belinky
- Virus Persistence and Dynamics Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Michail Orfanakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laurence de Leval
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana B. Enriquez
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Susan Moir
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA
| | - Nicole Doria-Rose
- Virology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Eli Boritz
- Virus Persistence and Dynamics Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - John R. Mascola
- Virology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
- ModeX Therapeutics, Weston, MA, USA
| | - Rafick-Pierre Sekaly
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Richard A. Koup
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
13
|
Tolomeo M, Cascio A. The Complex Dysregulations of CD4 T Cell Subtypes in HIV Infection. Int J Mol Sci 2024; 25:7512. [PMID: 39062756 PMCID: PMC11276885 DOI: 10.3390/ijms25147512] [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: 06/04/2024] [Revised: 07/04/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Human immunodeficiency virus (HIV) infection remains an important global public health problem. About 40 million people are infected with HIV, and this infection caused about 630,000 deaths in 2022. The hallmark of HIV infection is the depletion of CD4+ T helper lymphocytes (Th cells). There are at least seven different Th subtypes, and not all are the main targets of HIV. Moreover, the effect of the virus in a specific subtype can be completely different from that of the others. Although the most compromised Th subtype in HIV infection is Th17, HIV can induce important dysregulations in other subtypes, such as follicular Th (Tfh) cells and regulatory Th cells (Treg cells or Tregs). Several studies have shown that HIV can induce an increase in the immunosuppressive activity of Tregs without causing a significant reduction in their numbers, at least in the early phase of infection. The increased activity of this Th subtype seems to play an important role in determining the immunodeficiency status of HIV-infected patients, and Tregs may represent a new target for innovative anti-HIV therapies, including the so-called "Kick and Kill" therapeutic method whose goal is the complete elimination of the virus and the healing of HIV infection. In this review, we report the most important findings on the effects of HIV on different CD4+ T cell subtypes, the molecular mechanisms by which the virus impairs the functions of these cells, and the implications for new anti-HIV therapeutic strategies.
Collapse
Affiliation(s)
- Manlio Tolomeo
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy;
- Department of Infectious Diseases, A.O.U.P. Palermo, 90127 Palermo, Italy
| | - Antonio Cascio
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy;
- Department of Infectious Diseases, A.O.U.P. Palermo, 90127 Palermo, Italy
| |
Collapse
|
14
|
Kögl T, Chang HF, Staniek J, Chiang SC, Thoulass G, Lao J, Weißert K, Dettmer-Monaco V, Geiger K, Manna PT, Beziat V, Momenilandi M, Tu SM, Keppler SJ, Pattu V, Wolf P, Kupferschmid L, Tholen S, Covill LE, Ebert K, Straub T, Groß M, Gather R, Engel H, Salzer U, Schell C, Maier S, Lehmberg K, Cornu TI, Pircher H, Shahrooei M, Parvaneh N, Elling R, Rizzi M, Bryceson YT, Ehl S, Aichele P, Ammann S. Patients and mice with deficiency in the SNARE protein SYNTAXIN-11 have a secondary B cell defect. J Exp Med 2024; 221:e20221122. [PMID: 38722309 PMCID: PMC11082451 DOI: 10.1084/jem.20221122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 03/08/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
SYNTAXIN-11 (STX11) is a SNARE protein that mediates the fusion of cytotoxic granules with the plasma membrane at the immunological synapses of CD8 T or NK cells. Autosomal recessive inheritance of deleterious STX11 variants impairs cytotoxic granule exocytosis, causing familial hemophagocytic lymphohistiocytosis type 4 (FHL-4). In several FHL-4 patients, we also observed hypogammaglobulinemia, elevated frequencies of naive B cells, and increased double-negative DN2:DN1 B cell ratios, indicating a hitherto unrecognized role of STX11 in humoral immunity. Detailed analysis of Stx11-deficient mice revealed impaired CD4 T cell help for B cells, associated with disrupted germinal center formation, reduced isotype class switching, and low antibody avidity. Mechanistically, Stx11-/- CD4 T cells exhibit impaired membrane fusion leading to reduced CD107a and CD40L surface mobilization and diminished IL-2 and IL-10 secretion. Our findings highlight a critical role of STX11 in SNARE-mediated membrane trafficking and vesicle exocytosis in CD4 T cells, important for successful CD4 T cell-B cell interactions. Deficiency in STX11 impairs CD4 T cell-dependent B cell differentiation and humoral responses.
Collapse
Affiliation(s)
- Tamara Kögl
- Institute for Immunology, Center for Microbiology and Hygiene, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Hsin-Fang Chang
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Julian Staniek
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center— University of Freiburg, Freiburg, Germany
| | - Samuel C.C. Chiang
- Division of Bone Marrow Transplantation and Immune Deficiency, and Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
- Department of Medicine, Center for Hematology and Regenerative Medicine Huddinge, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Gudrun Thoulass
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Jessica Lao
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Kristoffer Weißert
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Viviane Dettmer-Monaco
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Institute for Transfusion Medicine and Gene Therapy—University of Freiburg, Freiburg, Germany
| | - Kerstin Geiger
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Institute for Transfusion Medicine and Gene Therapy—University of Freiburg, Freiburg, Germany
| | - Paul T. Manna
- Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Vivien Beziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris-Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Mana Momenilandi
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris-Cité, Paris, France
| | - Szu-Min Tu
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Selina J. Keppler
- Division of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Varsha Pattu
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Philipp Wolf
- Department of Urology, Faculty of Medicine, Medical Center—University of Freiburg, Freiburg, Germany
| | - Laurence Kupferschmid
- Institute of Medical Microbiology and Hygiene, University Medical Center, Freiburg, Germany
| | - Stefan Tholen
- Department of Pathology, Institute of Surgical Pathology, University Medical Center, University of Freiburg, Freiburg, Germany
| | - Laura E. Covill
- Department of Medicine, Center for Hematology and Regenerative Medicine Huddinge, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Karolina Ebert
- Institute for Immunology, Center for Microbiology and Hygiene, Medical Center—University of Freiburg, Freiburg, Germany
| | - Tobias Straub
- Institute for Immunology, Center for Microbiology and Hygiene, Medical Center—University of Freiburg, Freiburg, Germany
| | - Miriam Groß
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Ruth Gather
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Helena Engel
- Institute for Immunology, Center for Microbiology and Hygiene, Medical Center—University of Freiburg, Freiburg, Germany
| | - Ulrich Salzer
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center— University of Freiburg, Freiburg, Germany
| | - Christoph Schell
- Department of Pathology, Institute of Surgical Pathology, University Medical Center, University of Freiburg, Freiburg, Germany
| | - Sarah Maier
- Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Lehmberg
- Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tatjana I. Cornu
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Institute for Transfusion Medicine and Gene Therapy—University of Freiburg, Freiburg, Germany
| | - Hanspeter Pircher
- Institute for Immunology, Center for Microbiology and Hygiene, Medical Center—University of Freiburg, Freiburg, Germany
| | - Mohammad Shahrooei
- Department of Microbiology, Immunology, and Transplantation, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium
- Dr. Shahrooei Laboratory, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Parvaneh
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| | - Roland Elling
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty for Medicine, Center for Pediatrics and Adolescent Medicine, Medical Center—University of Freiburg, Freiburg, Germany
| | - Marta Rizzi
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center— University of Freiburg, Freiburg, Germany
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Clinical Immunology, Medical Center—University of Freiburg, Freiburg, Germany
| | - Yenan T. Bryceson
- Department of Medicine, Center for Hematology and Regenerative Medicine Huddinge, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Division of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Broegelmann Laboratory, Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Stephan Ehl
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Peter Aichele
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Sandra Ammann
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| |
Collapse
|
15
|
Qi R, Fu R, Lei X, He J, Jiang Y, Zhang L, Wu Y, Wang S, Guo X, Chen F, Nie M, Yang M, Chen Y, Zeng J, Xu J, Xiong H, Fang M, Que Y, Yao Y, Wang Y, Cao J, Ye H, Zhang Y, Zheng Z, Cheng T, Zhang J, Lin X, Yuan Q, Zhang T, Xia N. Therapeutic vaccine-induced plasma cell differentiation is defective in the presence of persistently high HBsAg levels. J Hepatol 2024; 80:714-729. [PMID: 38336348 DOI: 10.1016/j.jhep.2023.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 12/15/2023] [Accepted: 12/29/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND & AIMS Mechanisms behind the impaired response of antigen-specific B cells to therapeutic vaccination in chronic hepatitis B virus (HBV) infection remain unclear. The development of vaccines or strategies to overcome this obstacle is vital for advancing the management of chronic hepatitis B. METHODS A mouse model, denominated as E6F6-B, was engineered to feature a knock-in of a B-cell receptor (BCR) that specifically recognizes HBsAg. This model served as a valuable tool for investigating the temporal and spatial dynamics of humoral responses following therapeutic vaccination under continuous antigen exposure. Using a suite of immunological techniques, we elucidated the differentiation trajectory of HBsAg-specific B cells post-therapeutic vaccination in HBV carrier mice. RESULTS Utilizing the E6F6-B transfer model, we observed a marked decline in antibody-secreting cells 2 weeks after vaccination. A dysfunctional and atypical pre-plasma cell population (BLIMP-1+ IRF4+ CD40- CD138- BCMA-) emerged, manifested by sustained BCR signaling. By deploying an antibody to purge persistent HBsAg, we effectively prompted the therapeutic vaccine to provoke conventional plasma cell differentiation. This resulted in an enhanced anti-HBs antibody response and facilitated HBsAg clearance. CONCLUSIONS Sustained high levels of HBsAg limit the ability of therapeutic hepatitis B vaccines to induce the canonical plasma cell differentiation necessary for anti-HBs antibody production. Employing a strategy combining antibodies with vaccines can surmount this altered humoral response associated with atypical pre-plasma cells, leading to improved therapeutic efficacy in HBV carrier mice. IMPACT AND IMPLICATIONS Therapeutic vaccines aimed at combatting HBV encounter suboptimal humoral responses in clinical settings, and the mechanisms impeding their effectiveness have remained obscure. Our research, utilizing the innovative E6F6-B mouse transfer model, reveals that the persistence of HBsAg can lead to the emergence of an atypical pre-plasma cell population, which proves to be relevant to the potency of therapeutic HBV vaccines. Targeting the aberrant differentiation process of these atypical pre-plasma cells stands out as a critical strategy to amplify the humoral response elicited by HBV therapeutic vaccines in carrier mouse models. This discovery suggests a compelling avenue for further study in the context of human chronic hepatitis B. Encouragingly, our findings indicate that synergistic therapy combining HBV-specific antibodies with vaccines offers a promising approach that could significantly advance the pursuit of a functional cure for HBV.
Collapse
Affiliation(s)
- Ruoyao Qi
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Rao Fu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Xing Lei
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Jinhang He
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Yao Jiang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Liang Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Yangtao Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Siling Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Xueran Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Feng Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Meifeng Nie
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Man Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Yiyi Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Jing Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China; Department of clinical laboratory, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Jingjing Xu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Hualong Xiong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Mujin Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Yuqiong Que
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Youliang Yao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Yingbin Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Jiali Cao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China; Department of clinical laboratory, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Huiming Ye
- Department of clinical laboratory, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Yali Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Zizheng Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Xu Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
| | - Quan Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China.
| | - Tianying Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China.
| |
Collapse
|
16
|
Höft MA, Burgers WA, Riou C. The immune response to SARS-CoV-2 in people with HIV. Cell Mol Immunol 2024; 21:184-196. [PMID: 37821620 PMCID: PMC10806256 DOI: 10.1038/s41423-023-01087-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023] Open
Abstract
This review examines the intersection of the HIV and SARS-CoV-2 pandemics. People with HIV (PWH) are a heterogeneous group that differ in their degree of immune suppression, immune reconstitution, and viral control. While COVID-19 in those with well-controlled HIV infection poses no greater risk than that for HIV-uninfected individuals, people with advanced HIV disease are more vulnerable to poor COVID-19 outcomes. COVID-19 vaccines are effective and well tolerated in the majority of PWH, though reduced vaccine efficacy, breakthrough infections and faster waning of vaccine effectiveness have been demonstrated in PWH. This is likely a result of suboptimal humoral and cellular immune responses after vaccination. People with advanced HIV may also experience prolonged infection that may give rise to new epidemiologically significant variants, but initiation or resumption of antiretroviral therapy (ART) can effectively clear persistent infection. COVID-19 vaccine guidelines reflect these increased risks and recommend prioritization for vaccination and additional booster doses for PWH who are moderately to severely immunocompromised. We recommend continued research and monitoring of PWH with SARS-CoV-2 infection, especially in areas with a high HIV burden.
Collapse
Affiliation(s)
- Maxine A Höft
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Wendy A Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa.
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
17
|
Xie L, Fang J, Yu J, Zhang W, He Z, Ye L, Wang H. The role of CD4 + T cells in tumor and chronic viral immune responses. MedComm (Beijing) 2023; 4:e390. [PMID: 37829505 PMCID: PMC10565399 DOI: 10.1002/mco2.390] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
Immunotherapies are mainly aimed to promote a CD8+ T cell response rather than a CD4+ T cell response as cytotoxic T lymphocytes (CTLs) can directly kill target cells. Recently, CD4+ T cells have received more attention due to their diverse roles in tumors and chronic viral infections. In antitumor and antichronic viral responses, CD4+ T cells relay help signals through dendritic cells to indirectly regulate CD8+ T cell response, interact with B cells or macrophages to indirectly modulate humoral immunity or macrophage polarization, and inhibit tumor blood vessel formation. Additionally, CD4+ T cells can also exhibit direct cytotoxicity toward target cells. However, regulatory T cells exhibit immunosuppression and CD4+ T cells become exhausted, which promote tumor progression and chronic viral persistence. Finally, we also outline immunotherapies based on CD4+ T cells, including adoptive cell transfer, vaccines, and immune checkpoint blockade. Overall, this review summarizes diverse roles of CD4+ T cells in the antitumor or protumor and chronic viral responses, and also highlights the immunotherapies based on CD4+ T cells, giving a better understanding of their roles in tumors and chronic viral infections.
Collapse
Affiliation(s)
- Luoyingzi Xie
- Institute of Hepatopancreatobiliary SurgeryChongqing General HospitalChongqingChina
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Jingyi Fang
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Juncheng Yu
- Department of Thoracic SurgeryXinqiao Hospital Third Military Medical University (Army Medical University)ChongqingChina
| | - Weinan Zhang
- Department of Plastic & Cosmetic SurgeryArmy Medical Center of PLAAmy Medical UniversityChongqingChina
| | - Zhiqiang He
- Department of Plastic & Cosmetic SurgeryArmy Medical Center of PLAAmy Medical UniversityChongqingChina
| | - Lilin Ye
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary SurgeryChongqing General HospitalChongqingChina
| |
Collapse
|
18
|
Govindaraj S, Babu H, Kannanganat S, Vaccari M, Petrovas C, Velu V. Editorial: CD4+ T cells in HIV: A Friend or a Foe? Front Immunol 2023; 14:1203531. [PMID: 37497218 PMCID: PMC10367341 DOI: 10.3389/fimmu.2023.1203531] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/26/2023] [Indexed: 07/28/2023] Open
Affiliation(s)
- Sakthivel Govindaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Hemalatha Babu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Sunil Kannanganat
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital Research Institute, Houston, TX, United States
| | - Monica Vaccari
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, United States
- Department of Microbiology and Immunology, Tulane School of Medicine, New Orleans, LA, United States
| | - Constantinos Petrovas
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Vijayakumar Velu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, United States
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW This review summarizes recent studies reporting the induction of vaccinal effects by human immunodeficiency virus (HIV-1) antibody therapy. It also puts into perspective preclinical studies that have identified mechanisms involved in the immunomodulatory properties of antiviral antibodies. Finally, it discusses potential therapeutic interventions to enhance host adaptive immune responses in people living with HIV (PLWH) treated with broadly neutralizing antibodies (bNAbs). RECENT FINDINGS Recent studies in promising clinical trials have shown that, in addition to controlling viremia, anti-HIV-1 bNAbs are able to enhance the host's humoral and cellular immune response. Such vaccinal effects, in particular the induction of HIV-1-specific CD8 + T-cell responses, have been observed upon treatment with two potent bNAbs (3BNC117 and 10-1074) alone or in combination with latency-reversing agents (LRA). While these studies reinforce the idea that bNAbs can induce protective immunity, the induction of vaccinal effects is not systematic and might depend on both the virological status of the patient as well as the therapeutic strategy chosen. SUMMARY HIV-1 bNAbs can enhance adaptive host immune responses in PLWH. The challenge now is to exploit these immunomodulatory properties to design optimized therapeutic interventions to promote and enhance the induction of protective immunity against HIV-1 infection during bNAbs therapy.
Collapse
|
20
|
Ge M, Yang C, Li T, Du T, Zhang P, Li X, Dou Y, Duan R. Circulating CXCR5 + natural killer cells are expanded in patients with myasthenia gravis. Clin Transl Immunology 2023; 12:e1450. [PMID: 37223338 PMCID: PMC10202622 DOI: 10.1002/cti2.1450] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/18/2023] [Accepted: 04/18/2023] [Indexed: 05/25/2023] Open
Abstract
Objectives Myasthenia gravis (MG) is a classic autoantibody-mediated disease in which pathogenic antibodies target postsynaptic membrane components, causing fluctuating skeletal muscle weakness and fatigue. Natural killer (NK) cells are heterogeneous lymphocytes that have gained increasing attention owing to their potential roles in autoimmune disorders. This study will investigate the relationship between the distinct NK cell subsets and MG pathogenesis. Methods A total of 33 MG patients and 19 healthy controls were enrolled in the present study. Circulating NK cells, their subtypes and follicular helper T cells were analysed by flow cytometry. Serum acetylcholine receptor (AChR) antibody levels were determined by ELISA. The role of NK cells in the regulation of B cells was verified using a co-culture assay. Results Myasthenia gravis patients with acute exacerbations had a reduced number of total NK cells, CD56dim NK cells and IFN-γ-secreting NK cells in the peripheral blood, while CXCR5+ NK cells were significantly elevated. CXCR5+ NK cells expressed a higher level of ICOS and PD-1 and a lower level of IFN-γ than those in CXCR5- NK cells and were positively correlated with Tfh cell and AChR antibody levels. In vitro experiments demonstrated that NK cells suppressed plasmablast differentiation while promoting CD80 and PD-L1 expression on B cells in an IFN-γ-dependent manner. Furthermore, CXCR5- NK cells inhibited plasmablast differentiation, while CXCR5+ NK cells could more efficiently promote B cell proliferation. Conclusion These results reveal that CXCR5+ NK cells exhibit distinct phenotypes and functions compared with CXCR5- NK cells and might participate in the pathogenesis of MG.
Collapse
Affiliation(s)
- Meng‐Ru Ge
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan HospitalJinanChina
- Department of Neurology, Shandong Provincial Qianfoshan HospitalCheeloo College of Medicine, Shandong UniversityJinanChina
| | - Chun‐Lin Yang
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan HospitalJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
- Shandong Provincial Medicine and Health Key Laboratory of NeuroimmunologyJinanChina
| | - Tao Li
- Department of Neurology, Shandong Provincial Qianfoshan HospitalCheeloo College of Medicine, Shandong UniversityJinanChina
| | - Tong Du
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan HospitalJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
- Shandong Provincial Medicine and Health Key Laboratory of NeuroimmunologyJinanChina
| | - Peng Zhang
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan HospitalJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
- Shandong Provincial Medicine and Health Key Laboratory of NeuroimmunologyJinanChina
| | - Xiao‐Li Li
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan HospitalJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
- Shandong Provincial Medicine and Health Key Laboratory of NeuroimmunologyJinanChina
| | - Ying‐Chun Dou
- College of Basic Medical Sciences, Shandong University of Traditional Chinese MedicineJinanChina
| | - Rui‐Sheng Duan
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan HospitalJinanChina
- Department of Neurology, Shandong Provincial Qianfoshan HospitalCheeloo College of Medicine, Shandong UniversityJinanChina
- Shandong Institute of NeuroimmunologyJinanChina
- Shandong Provincial Medicine and Health Key Laboratory of NeuroimmunologyJinanChina
| |
Collapse
|
21
|
Schnittman SR, Jung W, Fitch KV, Zanni MV, McCallum S, Lee JSL, Shin S, Davis BJ, Fulda ES, Diggs MR, Giguel F, Chinchay R, Sheth AN, Fichtenbaum CJ, Malvestutto C, Aberg JA, Currier J, Lauffenburger DA, Douglas PS, Ribaudo HJ, Alter G, Grinspoon SK. Effect of host factors and COVID-19 infection on the humoral immune repertoire in treated HIV. JCI Insight 2023; 8:e166848. [PMID: 36805331 PMCID: PMC10077482 DOI: 10.1172/jci.insight.166848] [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: 11/07/2022] [Accepted: 01/27/2023] [Indexed: 02/22/2023] Open
Abstract
People with HIV (PWH) appear to be at higher risk for suboptimal pathogen responses and for worse COVID-19 outcomes, but the effects of host factors and COVID-19 on the humoral repertoire remain unclear. We assessed the antibody isotype/subclass and Fc-receptor binding Luminex arrays of non-SARS-CoV-2 and SARS-CoV-2 humoral responses among antiretroviral therapy-treated (ART-treated) PWH. Among the entire cohort, COVID-19 infection was associated with higher cytomegalovirus (CMV) responses (vs. the COVID- cohort ), potentially signifying increased susceptibility or a consequence of persistent inflammation. Among the COVID+ participants, (a) higher BMI was associated with a striking amplification of SARS-CoV-2 responses, suggesting exaggerated inflammatory responses, and (b) lower nadir CD4 was associated with higher SARS-CoV-2 IgM and FcγRIIB binding capacity, indicating poorly functioning extrafollicular and inhibitory responses. Among the COVID-19- participants, female sex, older age, and lower nadir CD4 were associated with unique repertoire shifts. In this first comprehensive assessment of the humoral repertoire in a global cohort of PWH, we identify distinct SARS-CoV-2-specific humoral immune profiles among PWH with obesity or lower nadir CD4+ T cell count, underlining plausible mechanisms associated with worse COVID-19-related outcomes in this setting. Host factors associated with the humoral repertoire in the COVID-19- cohort enhance our understanding of these important shifts among PWH.
Collapse
Affiliation(s)
- Samuel R. Schnittman
- Division of Infectious Diseases, Department of Medicine, and
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Wonyeong Jung
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Kathleen V. Fitch
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Markella V. Zanni
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sara McCallum
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Sally Shin
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Brandon J. Davis
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Evelynne S. Fulda
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marissa R. Diggs
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Francoise Giguel
- AIDS Clinical Trials Group Lab 01, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Romina Chinchay
- Houston AIDS Research Team, University of Texas Health Science Center Houston, Houston, Texas, USA
| | - Anandi N. Sheth
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Carl J. Fichtenbaum
- Division of Infectious Diseases, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Carlos Malvestutto
- Division of Infectious Diseases, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Judith A. Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Judith Currier
- Division of Infectious Diseases, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Pamela S. Douglas
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | | | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | | |
Collapse
|
22
|
Chi X, Gu J, Ma X. Characteristics and Roles of T Follicular Helper Cells in SARS-CoV-2 Vaccine Response. Vaccines (Basel) 2022; 10:vaccines10101623. [PMID: 36298488 PMCID: PMC9611968 DOI: 10.3390/vaccines10101623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination is critical to controlling the coronavirus disease 2019 (COVID-19) pandemic. However, a weak response to the vaccine and insufficient persistence of specific antibodies may threaten the global impact of mass vaccination campaigns. This study summarizes the internal factors of the body that affect the effectiveness of the SARS-CoV-2 vaccine. T follicular helper (Tfh) cells support germinal center B cells to produce vaccine-specific immunoglobulins. A reduction in the Tfh cell number and a shift in the subset phenotypes caused by multiple factors may impair the production and persistence of high-affinity antibodies. Besides efficacy differences caused by the different types of vaccines, the factors that affect vaccine effectiveness by intervening in the Tfh cell response also include age-related defects, the polarity of the body microenvironment, repeated immunization, immunodeficiency, and immunosuppressive treatments. Assessing the phenotypic distribution and activation levels of Tfh cell subsets after vaccination is helpful in predicting vaccine responses and may identify potential targets for improving vaccine effectiveness.
Collapse
Affiliation(s)
- Xuyang Chi
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jia Gu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaoxue Ma
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang 110001, China
- Department of Microbiology & Immunology and Pediatrics, Dalhousie University, and Canadian Center for Vaccinology, IWK Health Centre, Halifax, NS B3K 6R8, Canada
- Correspondence: ; Tel.: +86-024-83282527
| |
Collapse
|
23
|
Noto A, Suffiotti M, Joo V, Mancarella A, Procopio FA, Cavet G, Leung Y, Corpataux JM, Cavassini M, Riva A, Stamatatos L, Gottardo R, McDermott AB, Koup RA, Fenwick C, Perreau M, Pantaleo G. The deficiency in Th2-like Tfh cells affects the maturation and quality of HIV-specific B cell response in viremic infection. Front Immunol 2022; 13:960120. [PMID: 36091040 PMCID: PMC9450063 DOI: 10.3389/fimmu.2022.960120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Optimal T follicular helper (Tfh) cells function is important to promote the development of germinal centers and maturation of high affinity antigen-specific B cells. We have found that the expression of CXCR3 defines distinct Tfh subsets: CXCR3+ Th1-like Tfh cells mainly producing single IFN-γ and dual IL-21/IFN-γ and CXCR3- Th2-like Tfh cells mainly producing single IL-4 and dual IL-21/IL-4 cytokines. CXCR3- Th2-like Tfhs are significantly reduced during ongoing HIV replication. While the percentage of Th2-like Tfh cells correlates with that of total and cycling HIV-specific B cells, the percentage of CXCR3+ Th1-like Tfhs correlates with HIV-specific B cells expressing T-bet and CXCR3. Of note, only IL-4 and IL-21 cytokines boosted efficient maturation of HIV-specific B cells while IFN-γ induced expression of T-bet and CXCR3 in B cells. Interestingly, total and HIV-specific CXCR3+ B cells showed lower rate of somatic hypermutation, as compared to CXCR3- B cells. Therefore, the imbalance in Th2/Th1-like Tfhs affects B cell responses in viremic HIV infection.
Collapse
Affiliation(s)
- Alessandra Noto
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Madeleine Suffiotti
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Victor Joo
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Antonio Mancarella
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Francesco A. Procopio
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Guy Cavet
- Atreca, Redwood City, CA, United States
| | | | - Jean-Marc Corpataux
- Service of Vascular Surgery, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Matthias Cavassini
- Service of Infectious Diseases, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Agostino Riva
- Division of Infectious Diseases, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Leonidas Stamatatos
- Department of Global Health, Seattle University of Washington, Seattle, WA, United States
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Adrian B. McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Richard A. Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Craig Fenwick
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Matthieu Perreau
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland,Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland,*Correspondence: Giuseppe Pantaleo,
| |
Collapse
|
24
|
Baiyegunhi OO, Mann J, Khaba T, Nkosi T, Mbatha A, Ogunshola F, Chasara C, Ismail N, Ngubane T, Jajbhay I, Pansegrouw J, Dong KL, Walker BD, Ndung'u T, Ndhlovu ZM. CD8 lymphocytes mitigate HIV-1 persistence in lymph node follicular helper T cells during hyperacute-treated infection. Nat Commun 2022; 13:4041. [PMID: 35831418 PMCID: PMC9279299 DOI: 10.1038/s41467-022-31692-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 06/29/2022] [Indexed: 11/09/2022] Open
Abstract
HIV persistence in tissue sites despite ART is a major barrier to HIV cure. Detailed studies of HIV-infected cells and immune responses in native lymph node tissue environment is critical for gaining insight into immune mechanisms impacting HIV persistence and clearance in tissue sanctuary sites. We compared HIV persistence and HIV-specific T cell responses in lymph node biopsies obtained from 14 individuals who initiated therapy in Fiebig stages I/II, 5 persons treated in Fiebig stages III-V and 17 late treated individuals who initiated ART in Fiebig VI and beyond. Using multicolor immunofluorescence staining and in situ hybridization, we detect HIV RNA and/or protein in 12 of 14 Fiebig I/II treated persons on suppressive therapy for 1 to 55 months, and in late treated persons with persistent antigens. CXCR3+ T follicular helper cells harbor the greatest amounts of gag mRNA transcripts. Notably, HIV-specific CD8+ T cells responses are associated with lower HIV antigen burden, suggesting that these responses may contribute to HIV suppression in lymph nodes during therapy. These results reveal HIV persistence despite the initiation of ART in hyperacute infection and highlight the contribution of virus-specific responses to HIV suppression in tissue sanctuaries during suppressive ART.
Collapse
Affiliation(s)
- Omolara O Baiyegunhi
- Africa Health Research Institute (AHRI), Durban, South Africa
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Jaclyn Mann
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Trevor Khaba
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Thandeka Nkosi
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Anele Mbatha
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Funsho Ogunshola
- Africa Health Research Institute (AHRI), Durban, South Africa
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, USA
| | | | - Nasreen Ismail
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Thandekile Ngubane
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | | | | | - Krista L Dong
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, USA
| | - Bruce D Walker
- Africa Health Research Institute (AHRI), Durban, South Africa
- HIV Pathogenesis Programme, The 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, USA
- Institute for Medical Sciences and Engineering and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Thumbi Ndung'u
- Africa Health Research Institute (AHRI), Durban, South Africa
- HIV Pathogenesis Programme, The 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, USA
- Max Planck Institute for Infection Biology, Berlin, Germany
- Division of Infection and Immunity, University College London, London, UK
| | - Zaza M Ndhlovu
- Africa Health Research Institute (AHRI), Durban, South Africa.
- HIV Pathogenesis Programme, The 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, USA.
| |
Collapse
|
25
|
Yu D, Walker LSK, Liu Z, Linterman MA, Li Z. Targeting T FH cells in human diseases and vaccination: rationale and practice. Nat Immunol 2022; 23:1157-1168. [PMID: 35817844 DOI: 10.1038/s41590-022-01253-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 05/24/2022] [Indexed: 12/13/2022]
Abstract
The identification of CD4+ T cells localizing to B cell follicles has revolutionized the knowledge of how humoral immunity is generated. Follicular helper T (TFH) cells support germinal center (GC) formation and regulate clonal selection and differentiation of memory and antibody-secreting B cells, thus controlling antibody affinity maturation and memory. TFH cells are essential in sustaining protective antibody responses necessary for pathogen clearance in infection and vaccine-mediated protection. Conversely, aberrant and excessive TFH cell responses mediate and sustain pathogenic antibodies to autoantigens, alloantigens, and allergens, facilitate lymphomagenesis, and even harbor viral reservoirs. TFH cell generation and function are determined by T cell antigen receptor (TCR), costimulation, and cytokine signals, together with specific metabolic and survival mechanisms. Such regulation is crucial to understanding disease pathogenesis and informing the development of emerging therapies for disease or novel approaches to boost vaccine efficacy.
Collapse
Affiliation(s)
- Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia. .,Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, Division of Infection & Immunity, University College London, Royal Free Campus, London, UK
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Zhanguo Li
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
26
|
Active PD-L1 incorporation within HIV virions functionally impairs T follicular helper cells. PLoS Pathog 2022; 18:e1010673. [PMID: 35788752 PMCID: PMC9286290 DOI: 10.1371/journal.ppat.1010673] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/15/2022] [Accepted: 06/14/2022] [Indexed: 11/19/2022] Open
Abstract
The limited development of broadly neutralizing antibodies (BnAbs) during HIV infection is classically attributed to an inadequate B-cell help brought by functionally impaired T follicular helper (Tfh) cells. However, the determinants of Tfh-cell functional impairment and the signals contributing to this condition remain elusive. In the present study, we showed that PD-L1 is incorporated within HIV virions through an active mechanism involving p17 HIV matrix protein. We subsequently showed that in vitro produced PD-L1high but not PD-L1low HIV virions, significantly reduced Tfh-cell proliferation and IL-21 production, ultimately leading to a decreased of IgG1 secretion from GC B cells. Interestingly, Tfh-cell functions were fully restored in presence of anti-PD-L1/2 blocking mAbs treatment, demonstrating that the incorporated PD-L1 proteins were functionally active. Taken together, the present study unveils an immunovirological mechanism by which HIV specifically exploits the regulatory potential of PD-L1 to suppress the immune system during the course of HIV infection. During HIV infection, the development of effective BnAbs remains a rare phenomenon, occurring in only 15–20% of HIV-infected individuals after years of infection. Although multiple mechanisms may be involved, recent studies have suggested that functional impairment of Tfh cells, through immune checkpoint (IC)/IC-Ligand (IC-L) interactions, may lead to a decrease in B-cell help leading to low BnAbs production. Our laboratory recently showed that PD-L1 was predominantly expressed on lymph node (LN) migratory dendritic cells located predominantly in extra-follicular areas, implying that the source of IC-L contributing to Tfh-cell functional impairment may be independent of cellular expression of IC-L. These observations prompted us to investigate the potential contribution of IC-L incorporated within HIV virion envelope to Tfh-cell functional impairment. We subsequently demonstrated that PD-L1 was incorporated into a large fraction of HIV virions in the plasma of viremic HIV-infected individuals. Interestingly, PD-L1 remains active when incorporated into HIV virions envelope and could impaired Tfh-cell proliferation, resulting in decreased IgG1 production by B cells in vitro. These findings demonstrate an unsuspected mechanism contributing to the regulation of Tfh-cell function, which may contribute to the low production of BnAbs by B cells during HIV infection.
Collapse
|
27
|
Rahman SA, Billingsley JM, Sharma AA, Styles TM, Govindaraj S, Shanmugasundaram U, Babu H, Riberio SP, Ali SA, Tharp GK, Ibegbu C, Waggoner SN, Johnson RP, Sekaly RP, Villinger F, Bosinger SE, Amara RR, Velu V. Lymph node CXCR5+ NK cells associate with control of chronic SHIV infection. JCI Insight 2022; 7:155601. [PMID: 35271506 PMCID: PMC9089783 DOI: 10.1172/jci.insight.155601] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 03/04/2022] [Indexed: 11/28/2022] Open
Abstract
The persistence of virally infected cells as reservoirs despite effective antiretroviral therapy is a major barrier to an HIV/SIV cure. These reservoirs are predominately contained within cells present in the B cell follicles (BCFs) of secondary lymphoid tissues, a site that is characteristically difficult for most cytolytic antiviral effector cells to penetrate. Here, we identified a population of NK cells in macaque lymph nodes that expressed BCF-homing receptor CXCR5 and accumulated within BCFs during chronic SHIV infection. These CXCR5+ follicular NK cells exhibited an activated phenotype coupled with heightened effector functions and a unique transcriptome characterized by elevated expression of cytolytic mediators (e.g., perforin and granzymes, LAMP-1). CXCR5+ NK cells exhibited high expression of FcγRIIa and FcγRIIIa, suggesting a potential for elevated antibody-dependent effector functionality. Consistently, accumulation of CXCR5+ NK cells showed a strong inverse association with plasma viral load and the frequency of germinal center follicular Th cells that comprise a significant fraction of the viral reservoir. Moreover, CXCR5+ NK cells showed increased expression of transcripts associated with IL-12 and IL-15 signaling compared with the CXCR5- subset. Indeed, in vitro treatment with IL-12 and IL-15 enhanced the proliferation of CXCR5+ granzyme B+ NK cells. Our findings suggest that follicular homing NK cells might be important in immune control of chronic SHIV infection, and this may have important implications for HIV cure strategies.
Collapse
Affiliation(s)
- Sheikh Abdul Rahman
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Microbiology and Immunology and
| | - James M Billingsley
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Ashish Arunkumar Sharma
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Tiffany M Styles
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Sakthivel Govindaraj
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Uma Shanmugasundaram
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Hemalatha Babu
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Susan Pereira Riberio
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Syed A Ali
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| | - Gregory K Tharp
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Chris Ibegbu
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Stephen N Waggoner
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - R Paul Johnson
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Microbiology and Immunology and.,Infectious Disease Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Rafick-Pierre Sekaly
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| | - Steve E Bosinger
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Rama Rao Amara
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Microbiology and Immunology and
| | - Vijayakumar Velu
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
28
|
Mayberry CL, Logan NA, Wilson JJ, Chang CH. Providing a Helping Hand: Metabolic Regulation of T Follicular Helper Cells and Their Association With Disease. Front Immunol 2022; 13:864949. [PMID: 35493515 PMCID: PMC9047778 DOI: 10.3389/fimmu.2022.864949] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/25/2022] [Indexed: 01/02/2023] Open
Abstract
T follicular helper (Tfh) cells provide support to B cells upon arrival in the germinal center, and thus are critical for the generation of a robust adaptive immune response. Tfh express specific transcription factors and cellular receptors including Bcl6, CXCR5, PD-1, and ICOS, which are critical for homing and overall function. Generally, the induction of an immune response is tightly regulated. However, deviation during this process can result in harmful autoimmunity or the inability to successfully clear pathogens. Recently, it has been shown that Tfh differentiation, activation, and proliferation may be linked with the cellular metabolic state. In this review we will highlight recent discoveries in Tfh differentiation and explore how these cells contribute to functional immunity in disease, including autoimmune-related disorders, cancer, and of particular emphasis, during infection.
Collapse
Affiliation(s)
| | | | | | - Chih-Hao Chang
- The Jackson Laboratory, Bar Harbor, ME, United States
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- *Correspondence: Chih-Hao Chang,
| |
Collapse
|
29
|
Lu X, Zhang X, Cheung AKL, Moog C, Xia H, Li Z, Wang R, Ji Y, Xia W, Liu Z, Yuan L, Wang X, Wu H, Zhang T, Su B. Abnormal Shift in B Memory Cell Profile Is Associated With the Expansion of Circulating T Follicular Helper Cells via ICOS Signaling During Acute HIV-1 Infection. Front Immunol 2022; 13:837921. [PMID: 35222430 PMCID: PMC8867039 DOI: 10.3389/fimmu.2022.837921] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Interactions between T follicular helper (Tfh) cells and germinal center B cells are essential for the differentiation of B cells and specific antibody responses against HIV-1 infection. However, the extent to which HIV-1 infection affects the dynamic interplay between these two cell populations in the bloodstream remains unclear. In this study, the dynamics of circulating Tfh (cTfh) and B cells and their relationship in individuals with acute and chronic HIV-1 infection were investigated. Twenty-five study subjects were enrolled from the Beijing PRIMO clinical cohort, a prospective cohort of HIV-1-negative men who have sex with men (MSM) for the identification of cases of acute HIV-1 infection (AHI) at Beijing Youan Hospital, Capital Medical University. Individuals with AHI were selected at random. Matched samples were also collected and analyzed from the same patients with chronic HIV-1 infection. None of the study subjects received antiretroviral therapy during acute or chronic infection. Multicolor flow cytometry was used for the immunophenotypic and functional characterization of cTfh cell and B cell subsets. AHI resulted in increased proportions in bulk cTfh, ICOS+cTfh or IL-21+ICOS+cTfh cells. In both acute and chronic infections, activated memory (AM), tissue-like memory (TLM), and plasmablast (PB) B cell levels were increased whilst resting memory (RM) and naïve mature (NM) B cell levels were decreased. Classical memory (CM) B cells were unaffected during infection. Association analyses showed that the levels of ICOS+cTfh and IL-21+ICOS+cTfh cells were negatively correlated with those of AM, CM, RM cells, and positively correlated with those of NM cells in AHI but not chronic HIV-1 infection stage (CHI). Moreover, the frequency of IL-21+ICOS+cTfh cells was also positively correlated with plasma HIV-1 viral load, and had an opposite association trend with CD4+T cell count in AHI. Our data suggests that HIV-1 infection drives the expansion of cTfh cells, which in turn leads to perturbations of B cell differentiation through ICOS signaling during acute infection stage. These findings provide insight on the role of ICOS in the regulation of cTfh/B cell interaction during AHI and may potentially guide the design of effective strategies for restoring anti-HIV-1 immunity in the infected patients.
Collapse
Affiliation(s)
- Xiaofan Lu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xin Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Allen Ka Loon Cheung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Christiane Moog
- Laboratoire d'ImmunoRhumatologie Moléculaire, plateforme GENOMAX, INSERM UMR_S 1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Huan Xia
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhen Li
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Rui Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yunxia Ji
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Wei Xia
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhiying Liu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lin Yuan
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiuwen Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hao Wu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Jeger-Madiot R, Vaineau R, Heredia M, Tchitchek N, Bertrand L, Pereira M, Konza O, Gouritin B, Hoareau-Coudert B, Corneau A, Blanc C, Savier E, Buffet P, Six A, Klatzmann D, Moris A, Graff-Dubois S. Naive and memory CD4 + T cell subsets can contribute to the generation of human Tfh cells. iScience 2022; 25:103566. [PMID: 34984326 PMCID: PMC8693005 DOI: 10.1016/j.isci.2021.103566] [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: 06/18/2021] [Revised: 09/07/2021] [Accepted: 12/01/2021] [Indexed: 11/28/2022] Open
Abstract
CD4+ T follicular helper cells (Tfh) promote B cell maturation and antibody production in secondary lymphoid organs. By using an innovative culture system based on splenocyte stimulation, we studied the dynamics of naive and memory CD4+ T cells during the generation of a Tfh cell response. We found that both naive and memory CD4+ T cells can acquire phenotypic and functional features of Tfh cells. Moreover, we show here that the transition of memory as well as naive CD4+ T cells into the Tfh cell profile is supported by the expression of pro-Tfh genes, including transcription factors known to orchestrate Tfh cell development. Using this culture system, we provide pieces of evidence that HIV infection differentially alters these newly identified pathways of Tfh cell generation. Such diversity in pathways of Tfh cell generation offers a new framework for the understanding of Tfh cell responses in physiological and pathological contexts.
Collapse
Affiliation(s)
- Raphaël Jeger-Madiot
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France.,Sorbonne Université, INSERM, CNRS, Center for Immunology and Microbial Infections, Paris, France
| | - Romain Vaineau
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France
| | - Maud Heredia
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France.,Sorbonne Université, INSERM, CNRS, Center for Immunology and Microbial Infections, Paris, France
| | - Nicolas Tchitchek
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France
| | - Lisa Bertrand
- Sorbonne Université, INSERM, CNRS, Center for Immunology and Microbial Infections, Paris, France.,Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell, Gif-sur-Yvette, France
| | - Mathias Pereira
- Sorbonne Université, INSERM, CNRS, Center for Immunology and Microbial Infections, Paris, France.,Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell, Gif-sur-Yvette, France
| | - Océane Konza
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France
| | - Bruno Gouritin
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France
| | | | - Aurélien Corneau
- Sorbonne Université, INSERM UMS037 PASS, Cytometry facility (CyPS), Paris, France
| | - Catherine Blanc
- Sorbonne Université, INSERM UMS037 PASS, Cytometry facility (CyPS), Paris, France
| | - Eric Savier
- Assistance Publique-Hôpitaux de Paris (AP-HP), Pitie-Salpetriere Hospital, Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, Paris, France.,Sorbonne Université, INSERM, St Antoine Research Center CRSA, Paris, France
| | - Pierre Buffet
- Université de Paris, INSERM, UMRS 1134, Biologie Intégrée du Globule Rouge, Paris, France
| | - Adrien Six
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Biotherapy and Département Hospitalo-Universitaire Inflammation-Immunopathology-Biotherapy (i2B), Paris, France
| | - David Klatzmann
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Biotherapy and Département Hospitalo-Universitaire Inflammation-Immunopathology-Biotherapy (i2B), Paris, France
| | - Arnaud Moris
- Sorbonne Université, INSERM, CNRS, Center for Immunology and Microbial Infections, Paris, France.,Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell, Gif-sur-Yvette, France
| | - Stéphanie Graff-Dubois
- Sorbonne Université, INSERM, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), Paris, France.,Sorbonne Université, INSERM, CNRS, Center for Immunology and Microbial Infections, Paris, France.,Sorbonne Université, INSERM UMS037 PASS, Cytometry facility (CyPS), Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP), Pitie-Salpetriere Hospital, Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, Paris, France
| |
Collapse
|
31
|
Immunogenicity of a Two-Dose Human Papillomavirus Vaccine Schedule in HIV-Infected Adolescents with Immune Reconstitution. Vaccines (Basel) 2022; 10:vaccines10010118. [PMID: 35062779 PMCID: PMC8779595 DOI: 10.3390/vaccines10010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/10/2022] Open
Abstract
HIV-infected patients are at increased risk of human papillomavirus (HPV) acquisition and HPV-associated diseases. This study set out to determine whether a two-dose (2D) HPV vaccination schedule was sufficient in HIV-infected adolescents with immune reconstitution (IR) following antiretroviral treatment. Participants aged 9–15 years who had CD4 cell counts > 500 cells/mm3 and HIV-1 RNA < 40 copies/mL for at least one year were assigned to the 2D schedule, while older participants or those without IR received a three-dose (3D) schedule. Antibodies to HPV-16 and -18 were measured using a pseudovirion-based neutralization assay. A total of 96 subjects were enrolled; 31.3% and 68.7% received the 2D and 3D schedule, respectively. Of these, 66.7% and 57.6% of the 2D and 3D participants, respectively, were male. The seroconversion rates for HPV-16 and HPV-18 were 100% in all cases, except for HPV-18 in males who received the 3D schedule (97.4%). In males, the anti-HPV-16 geometric mean titers (GMTs) were 6859.3 (95% confidence interval, 4394.3–10,707.1) and 7011.1 (4648.8–10,573.9) in the 2D and 3D groups (p = 0.946), respectively, and the anti-HPV-18 GMTs were 2039.3 (1432.2–2903.8) and 2859.8 (1810.0–4518.4) in the 2D and 3D (p = 0.313) groups, respectively. In females, the anti-HPV-16 GMTs were 15,758.7 (8868.0–28,003.4) and 26,241.6 (16,972.7–40,572.3) in the 2D and 3D groups (p = 0.197), respectively, and the anti-HPV-18 GMTs were 5971.4 (3026.8–11,780.6) and 9993.1 (5950.8–16,781.1) in the 2D and 3D groups (p = 0.271), respectively. In summary, a 2D schedule is as immunogenic in young adolescents with IR as a 3D schedule in older subjects and those without IR.
Collapse
|
32
|
Immunopathogenesis in HIV-associated pediatric tuberculosis. Pediatr Res 2022; 91:21-26. [PMID: 33731810 PMCID: PMC8446109 DOI: 10.1038/s41390-021-01393-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/25/2020] [Accepted: 01/18/2021] [Indexed: 11/09/2022]
Abstract
Tuberculosis (TB) is an increasing global emergency in human immunodeficiency virus/acquired immune deficiency syndrome (HIV/AIDS) patients, in which host immunity is dysregulated and compromised. However, the pathogenesis and efficacy of therapeutic strategies in HIV-associated TB in developing infants are essentially lacking. Bacillus Calmette-Guerin vaccine, an attenuated live strain of Mycobacterium bovis, is not adequately effective, which confers partial protection against Mycobacterium tuberculosis (Mtb) in infants when administered at birth. However, pediatric HIV infection is most devastating in the disease progression of TB. It remains challenging whether early antiretroviral therapy (ART) could maintain immune development and function, and restore Mtb-specific immune function in HIV-associated TB in children. A better understanding of the immunopathogenesis in HIV-associated pediatric Mtb infection is essential to provide more effective interventions, reducing the risk of morbidity and mortality in HIV-associated Mtb infection in infants. IMPACT: Children living with HIV are more likely prone to opportunistic infection, predisposing high risk of TB diseases. HIV and Mtb coinfection in infants may synergistically accelerate disease progression. Early ART may probably induce immune reconstitution inflammatory syndrome and TB pathology in HIV/Mtb coinfected infants.
Collapse
|
33
|
Wong CS, Buckner CM, Lage SL, Pei L, Assis FL, Dahlstrom EW, Anzick SL, Virtaneva K, Rupert A, Davis JL, Zhou T, Laidlaw E, Manion M, Galindo F, Anderson M, Seamon CA, Sneller MC, Lisco A, Deleage C, Pittaluga S, Moir S, Sereti I. Rapid Emergence of T Follicular Helper and Germinal Center B Cells Following Antiretroviral Therapy in Advanced HIV Disease. Front Immunol 2021; 12:752782. [PMID: 34938286 PMCID: PMC8686113 DOI: 10.3389/fimmu.2021.752782] [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: 08/03/2021] [Accepted: 11/09/2021] [Indexed: 02/01/2023] Open
Abstract
Low nadir CD4 T-cell counts in HIV+ patients are associated with high morbidity and mortality and lasting immune dysfunction, even after antiretroviral therapy (ART). The early events of immune recovery of T cells and B cells in severely lymphopenic HIV+ patients have not been fully characterized. In a cohort of lymphopenic (CD4 T-cell count < 100/µL) HIV+ patients, we studied mononuclear cells isolated from peripheral blood (PB) and lymph nodes (LN) pre-ART (n = 40) and 6-8 weeks post-ART (n = 30) with evaluation of cellular immunophenotypes; histology on LN sections; functionality of circulating T follicular helper (cTfh) cells; transcriptional and B-cell receptor profile on unfractionated LN and PB samples; and plasma biomarker measurements. A group of 19 healthy controls (HC, n = 19) was used as a comparator. T-cell and B-cell lymphopenia was present in PB pre-ART in HIV+ patients. CD4:CD8 and CD4 T- and B-cell PB subsets partly normalized compared to HC post-ART as viral load decreased. Strikingly in LN, ART led to a rapid decrease in interferon signaling pathways and an increase in Tfh, germinal center and IgD-CD27- B cells, consistent with histological findings of post-ART follicular hyperplasia. However, there was evidence of cTfh cells with decreased helper capacity and of limited B-cell receptor diversification post-ART. In conclusion, we found early signs of immune reconstitution, evidenced by a surge in LN germinal center cells, albeit limited in functionality, in HIV+ patients who initiate ART late in disease.
Collapse
Affiliation(s)
- Chun-Shu Wong
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Clarisa M. Buckner
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Silvia Lucena Lage
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Luxin Pei
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Felipe L. Assis
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Eric W. Dahlstrom
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, MT, United States
| | - Sarah L. Anzick
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, MT, United States
| | - Kimmo Virtaneva
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, MT, United States
| | - Adam Rupert
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Jeremy L. Davis
- Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Ting Zhou
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Elizabeth Laidlaw
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Maura Manion
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Frances Galindo
- Intramural Clinical Management and Operations Branch, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Megan Anderson
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Catherine A. Seamon
- Critical Care Medicine, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Michael C. Sneller
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Andrea Lisco
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Stefania Pittaluga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Irini Sereti
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
34
|
Mi Y, Han J, Zhu J, Jin T. Role of the PD-1/PD-L1 Signaling in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis: Recent Insights and Future Directions. Mol Neurobiol 2021; 58:6249-6271. [PMID: 34480337 PMCID: PMC8639577 DOI: 10.1007/s12035-021-02495-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is an autoimmunity-related chronic demyelination disease of the central nervous system (CNS), causing young disability. Currently, highly specific immunotherapies for MS are still lacking. Programmed cell death 1 (PD-1) is an immunosuppressive co-stimulatory molecule, which is expressed on activated T lymphocytes, B lymphocytes, natural killer cells, and other immune cells. PD-L1, the ligand of PD-1, is expressed on T lymphocytes, B lymphocytes, dendritic cells, and macrophages. PD-1/PD-L1 delivers negative regulatory signals to immune cells, maintaining immune tolerance and inhibiting autoimmunity. This review comprehensively summarizes current insights into the role of PD-1/PD-L1 signaling in MS and its animal model experimental autoimmune encephalomyelitis (EAE). The potentiality of PD-1/PD-L1 as biomarkers or therapeutic targets for MS will also be discussed.
Collapse
Affiliation(s)
- Yan Mi
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
| | - Jinming Han
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
- Present Address: Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
| |
Collapse
|
35
|
Bajrai LH, Sohrab SS, Alandijany TA, Mobashir M, Reyaz M, Kamal MA, Firoz A, Parveen S, Azhar EI. Gene Expression Profiling of Early Acute Febrile Stage of Dengue Infection and Its Comparative Analysis With Streptococcus pneumoniae Infection. Front Cell Infect Microbiol 2021; 11:707905. [PMID: 34778101 PMCID: PMC8581568 DOI: 10.3389/fcimb.2021.707905] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/30/2021] [Indexed: 02/05/2023] Open
Abstract
Infectious diseases are the disorders caused by organisms such as bacteria, viruses, fungi, or parasites. Although many of them are permentantly hazardous, a number of them live in and on our bodies and they are normally harmless or even helpful. Under certain circumstances, some organisms may cause diseases and these infectious diseases may be passed directly from person to person or via intermediate vectors including insects and other animals. Dengue virus and Streptococcus pneumoniae are the critical and common sources of infectious diseases. So, it is critical to understand the gene expression profiling and their inferred functions in comparison to the normal and virus infected conditions. Here, we have analyzed the gene expression profiling for dengue hemorrhagic fever, dengue fever, and normal human dataset. Similar to it, streptococcus pneumoniae infectious data were analyzed and both the outcomes were compared. Our study leads to the conclusion that the dengue hemorrhagic fever arises in result to potential change in the gene expression pattern, and the inferred functions obviously belong to the immune system, but also there are some additional potential pathways which are critical signaling pathways. In the case of pneumoniae infection, 19 pathways were enriched, almost all these pathways are associated with the immune system and 17 of the enriched pathways were common with dengue infection except platelet activation and antigen processing and presentation. In terms of the comparative study between dengue virus and Streptococcus pneumoniae infection, we conclude that cell adhesion molecules (CAMs), MAPK signaling pathway, natural killer cell mediated cytotoxicity, regulation of actin cytoskeleton, and cytokine-cytokine receptor interaction are commonly enriched in all the three cases of dengue infection and Streptococcus pneumoniae infection, focal adhesion was enriched between classical dengue fever — dengue hemorrhagic fever, dengue hemorrhagic fever—normal samples, and SP, and antigen processing and presentation and Leukocyte transendothelial migration were enriched in classical dengue fever —normal samples, dengue hemorrhagic fever—normal samples, and Streptococcus pneumoniae infection.
Collapse
Affiliation(s)
- Leena H Bajrai
- Special Infectious Agents Unit - BSL-3, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia.,Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sayed S Sohrab
- Special Infectious Agents Unit - BSL-3, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Thamir A Alandijany
- Special Infectious Agents Unit - BSL-3, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Mobashir
- SciLifeLab, Department of Oncology and Pathology Karolinska Institutet, Stockholm, Sweden
| | - Muddassir Reyaz
- Department of Healthcare Management, Jamia Hamdard Hamdard Nagar, New Delhi, India
| | - Mohammad A Kamal
- West China School of Nursing/Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.,King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Enzymoics, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Ahmad Firoz
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Shabana Parveen
- Department of Bioscience, Jamia Millia Islamia, New Delhi, India
| | - Esam I Azhar
- Special Infectious Agents Unit - BSL-3, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
36
|
Mitchell JL, Pollara J, Dietze K, Edwards RW, Nohara J, N'guessan KF, Zemil M, Buranapraditkun S, Takata H, Li Y, Muir R, Kroon E, Pinyakorn S, Jha S, Manasnayakorn S, Chottanapund S, Thantiworasit P, Prueksakaew P, Ratnaratorn N, Nuntapinit B, Fox L, Tovanabutra S, Paquin-Proulx D, Wieczorek L, Polonis VR, Maldarelli F, Haddad EK, Phanuphak P, Sacdalan CP, Rolland M, Phanuphak N, Ananworanich J, Vasan S, Ferrari G, Trautmann L. Anti-HIV antibody development up to one year after antiretroviral therapy initiation in acute HIV infection. J Clin Invest 2021; 132:150937. [PMID: 34762600 PMCID: PMC8718150 DOI: 10.1172/jci150937] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/10/2021] [Indexed: 11/17/2022] Open
Abstract
Early initiation of antiretroviral therapy (ART) in acute HIV infection (AHI) is effective at limiting seeding of the HIV viral reservoir, but little is known about how the resultant decreased antigen load affects long-term Ab development after ART. We report here that Env-specific plasma antibody (Ab) levels and Ab-dependent cellular cytotoxicity (ADCC) increased during the first 24 weeks of ART and correlated with Ab levels persisting after 48 weeks of ART. Participants treated in AHI stage 1 had lower Env-specific Ab levels and ADCC activity on ART than did those treated later. Importantly, participants who initiated ART after peak viremia in AHI developed elevated cross-clade ADCC responses that were detectable 1 year after ART initiation, even though clinically undetectable viremia was reached by 24 weeks. These data suggest that there is more germinal center (GC) activity in the later stages of AHI and that Ab development continues in the absence of detectable viremia during the first year of suppressive ART. The development of therapeutic interventions that can enhance earlier development of GCs in AHI and Abs after ART initiation could provide important protection against the viral reservoir that is seeded in individuals treated early in the disease.
Collapse
Affiliation(s)
- Julie L Mitchell
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, United States of America
| | - Justin Pollara
- Department of Surgery, Duke University Medical Center, Durham, United States of America
| | - Kenneth Dietze
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, United States of America
| | - R Whitney Edwards
- Department of Surgery, Duke University Medical Center, Durham, United States of America
| | - Junsuke Nohara
- Department of Surgery, Duke University Medical Center, Durham, United States of America
| | - Kombo F N'guessan
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, United States of America
| | - Michelle Zemil
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, United States of America
| | - Supranee Buranapraditkun
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, United States of America
| | - Hiroshi Takata
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, United States of America
| | - Yifan Li
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, United States of America
| | - Roshell Muir
- Demartment of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University, Philadelphia, United States of America
| | - Eugene Kroon
- Institute of HIV Research and Innovation, Bangkok, Thailand
| | - Suteeraporn Pinyakorn
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, United States of America
| | - Shalini Jha
- Department of Surgery, Duke University Madical Center, Durham, United States of America
| | - Sopark Manasnayakorn
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Suthat Chottanapund
- Department of Surgery, Bamrasnaradura Infectious Disease Institute, Nonthaburi, Thailand
| | - Pattarawat Thantiworasit
- Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, Thailand
| | | | | | - Bessara Nuntapinit
- Armed Forces Research Institute of Medical Sciences in Bangkok, Bangkok, Thailand
| | - Lawrence Fox
- Division of AIDS, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, United States of America
| | - Sodsai Tovanabutra
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, United States of America
| | - Dominic Paquin-Proulx
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, United States of America
| | - Lindsay Wieczorek
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, United States of America
| | - Victoria R Polonis
- Department of Vaccine Research, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, United States of America
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, NCI/NIH, Frederick, United States of America
| | - Elias K Haddad
- Demartment of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, United States of America
| | | | | | - Morgane Rolland
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, United States of America
| | | | | | - Sandhya Vasan
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, United States of America
| | - Guido Ferrari
- Department of Surgery, Duke University Medical Center, Durham, United States of America
| | - Lydie Trautmann
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, United States of America
| |
Collapse
|
37
|
Silveira ELV, Hong JJ, Amancha PK, Rogers KA, Ansari AA, Byrareddy SN, Villinger F. Viremia controls Env-specific antibody-secreting cell responses in simian immunodeficiency virus infected macaques pre and post-antiretroviral therapy. AIDS 2021; 35:2085-2094. [PMID: 34148985 PMCID: PMC8490307 DOI: 10.1097/qad.0000000000002998] [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] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the kinetics of Env (gp140)-specific antibody-secreting cells (ASCs) during acute and early chronic simian immunodeficiency virus (SIV) infection, and prior to and postantiretroviral therapy (ART) in rhesus macaques. DESIGN AND METHODS At week 0, rhesus macaques were inoculated intravenously with SIVmac239 and the viral loads were allowed to develop. Daily ART was initiated at week 5 post infection until week 18, though the animals were monitored until week 28 for the following parameters: enumeration of SIV gp140-specific ASCs by ELISPOT; quantification of viremia and SIV gp140-specific IgG titres through qRT-PCR and ELISA, respectively; estimation of monocytes, follicular helper T cells (Tfh) and memory B cell frequencies using polychromatic flow cytometry. RESULTS Direct correlations were consistently found between blood SIV gp140-specific ASC responses and viremia or SIV Env-specific IgG titres. In contrast, SIV gp140-specific ASC responses showed inverse correlations with the percentage of total memory B cells in the blood. In lymph nodes, the magnitude of the SIV gp140-specific ASC responses also followed the viral load kinetics. In contrast, the number of SIV gp140-specific ASCs presented did not correlate with frequencies of circulating activated monocyte (CD14+CD16+) or Tfh cells. CONCLUSION Blood and/or lymph node viral loads may regulate the onset and magnitude of SIV gp140-specific ASCs during SIV infection and following ART in rhesus macaques.
Collapse
Affiliation(s)
- Eduardo L. V. Silveira
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329 - USA
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329 - USA
| | - Jung Joo Hong
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329 - USA
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329 - USA
| | - Praveen K. Amancha
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329 - USA
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329 - USA
| | - Kenneth A Rogers
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329 - USA
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329 - USA
| | - Aftab A. Ansari
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322 – USA
| | - Siddappa N. Byrareddy
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322 – USA
| | - Francois Villinger
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329 - USA
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329 - USA
| |
Collapse
|
38
|
Kvistad D, Pallikkuth S, Sirupangi T, Pahwa R, Kizhner A, Petrovas C, Villinger F, Pahwa S. IL-21 enhances influenza vaccine responses in aged macaques with suppressed SIV infection. JCI Insight 2021; 6:e150888. [PMID: 34491910 PMCID: PMC8564910 DOI: 10.1172/jci.insight.150888] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/01/2021] [Indexed: 11/28/2022] Open
Abstract
Natural aging and HIV infection are associated with chronic low-grade systemic inflammation, immune senescence, and impaired antibody responses to vaccines such as the influenza (flu) vaccine. We investigated the role of IL-21, a CD4+ T follicular helper cell (Tfh) regulator, on flu vaccine antibody response in nonhuman primates (NHPs) in the context of age and controlled SIV mac239 infection. Three doses of the flu vaccine with or without IL-21–IgFc were administered at 3-month intervals in aged SIV+ NHPs following virus suppression with antiretroviral therapy. IL-21–treated animals demonstrated higher day 14–postboost antibody responses, which associated with expanded CD4+ T central memory cells and peripheral Tfh–expressing (pTfh–expressing) T cell immunoreceptor with Ig and ITIM domains (TIGIT), expanded activated memory B cells, and contracted CD11b+ monocytes. Draining lymph node (LN) tissue from IL-21–treated animals revealed direct association between LN follicle Tfh density and frequency of circulating TIGIT+ pTfh cells. We conclude that IL-21 enhances flu vaccine–induced antibody responses in SIV+ aged rhesus macaques (RMs), acting as an adjuvant modulating LN germinal center activity. A strategy to supplement IL-21 in aging could be a valuable addition in the toolbox for improving vaccine responses in an aging HIV+ population.
Collapse
Affiliation(s)
- Daniel Kvistad
- Department of Microbiology and Immunology, University of Miami School of Medicine, Miami, Florida, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami School of Medicine, Miami, Florida, USA
| | - Tirupataiah Sirupangi
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| | - Rajendra Pahwa
- Department of Microbiology and Immunology, University of Miami School of Medicine, Miami, Florida, USA
| | - Alexander Kizhner
- Department of Microbiology and Immunology, University of Miami School of Medicine, Miami, Florida, USA
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, Maryland, USA.,Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Francois Villinger
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami School of Medicine, Miami, Florida, USA
| |
Collapse
|
39
|
Devanathan AS, Kashuba AD. Human Immunodeficiency Virus Persistence in the Spleen: Opportunities for Pharmacologic Intervention. AIDS Res Hum Retroviruses 2021; 37:725-735. [PMID: 33499746 DOI: 10.1089/aid.2020.0266] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The persistence of HIV in the spleen, despite combination antiretroviral therapy, is not well understood. Sustained immune dysregulation and delayed immune recovery, in addition to immune cell exhaustion, may contribute to persistence of infection in the spleen. Eliminating HIV from this secondary lymphoid organ will require a thorough understanding of antiretroviral (ARV) pharmacology in the spleen, which has been minimally investigated. Low ARV exposure within the spleen may hinder the achievement of a functional or sterilizing cure if cells are not protected from HIV infection. In this study, we provide an overview of the anatomy and physiology of the spleen, review the evidence of the spleen as a site for persistence of HIV, discuss the consequences of persistence of HIV in the spleen, address challenges to eradicating HIV in the spleen, and examine opportunities for future curative efforts.
Collapse
Affiliation(s)
| | - Angela D.M. Kashuba
- UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
40
|
T-Cell Responses in Merkel Cell Carcinoma: Implications for Improved Immune Checkpoint Blockade and Other Therapeutic Options. Int J Mol Sci 2021; 22:ijms22168679. [PMID: 34445385 PMCID: PMC8395396 DOI: 10.3390/ijms22168679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023] Open
Abstract
Merkel cell carcinoma (MCC) is a rare and aggressive skin cancer with rising incidence and high mortality. Approximately 80% of the cases are caused by the human Merkel cell polyomavirus, while the remaining 20% are induced by UV light leading to mutations. The standard treatment of metastatic MCC is the use of anti-PD-1/-PD-L1-immune checkpoint inhibitors (ICI) such as Pembrolizumab or Avelumab, which in comparison with conventional chemotherapy show better overall response rates and longer duration of responses in patients. Nevertheless, 50% of the patients do not respond or develop ICI-induced, immune-related adverse events (irAEs), due to diverse mechanisms, such as down-regulation of MHC complexes or the induction of anti-inflammatory cytokines. Other immunotherapeutic options such as cytokines and pro-inflammatory agents or the use of therapeutic vaccination offer great ameliorations to ICI. Cytotoxic T-cells play a major role in the effectiveness of ICI, and tumour-infiltrating CD8+ T-cells and their phenotype contribute to the clinical outcome. This literature review presents a summary of current and future checkpoint inhibitor therapies in MCC and demonstrates alternative therapeutic options. Moreover, the importance of T-cell responses and their beneficial role in MCC treatment is discussed.
Collapse
|
41
|
The Role of Coinfections in the EBV-Host Broken Equilibrium. Viruses 2021; 13:v13071399. [PMID: 34372605 PMCID: PMC8310153 DOI: 10.3390/v13071399] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/29/2021] [Accepted: 07/12/2021] [Indexed: 12/17/2022] Open
Abstract
The Epstein–Barr virus (EBV) is a well-adapted human virus, and its infection is exclusive to our species, generally beginning in the childhood and then persisting throughout the life of most of the affected adults. Although this infection generally remains asymptomatic, EBV can trigger life-threatening conditions under unclear circumstances. The EBV lifecycle is characterized by interactions with other viruses or bacteria, which increases the probability of awakening its pathobiont capacity. For instance, EBV infects B cells with the potential to alter the germinal center reaction (GCR)—an adaptive immune structure wherein mutagenic-driven processes take place. HIV- and Plasmodium falciparum-induced B cell hyperactivation also feeds the GCR. These agents, along with the B cell tropic KSHV, converge in the ontogeny of germinal center (GC) or post-GC lymphomas. EBV oral transmission facilitates interactions with local bacteria and HPV, thereby increasing the risk of periodontal diseases and head and neck carcinomas. It is less clear as to how EBV is localized in the stomach, but together with Helicobacter pylori, they are known to be responsible for gastric cancer. Perhaps this mechanism is reminiscent of the local inflammation that attracts different herpesviruses and enhances graft damage and chances of rejection in transplanted patients. In this review, we discussed the existing evidence suggestive of EBV possessing the potential to synergize or cooperate with these agents to trigger or worsen the disease.
Collapse
|
42
|
Chakhtoura M, Fang M, Cubas R, O’Connor MH, Nichols CN, Richardson B, Talla A, Moir S, Cameron MJ, Tardif V, Haddad EK. Germinal Center T follicular helper (GC-Tfh) cell impairment in chronic HIV infection involves c-Maf signaling. PLoS Pathog 2021; 17:e1009732. [PMID: 34280251 PMCID: PMC8289045 DOI: 10.1371/journal.ppat.1009732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
We have recently demonstrated that the function of T follicular helper (Tfh) cells from lymph nodes (LN) of HIV-infected individuals is impaired. We found that these cells were unable to provide proper help to germinal center (GC)-B cells, as observed by altered and inefficient anti-HIV antibody response and premature death of memory B cells. The underlying molecular mechanisms of this dysfunction remain poorly defined. Herein, we have used a unique transcriptional approach to identify these molecular defects. We consequently determined the transcriptional profiles of LN GC-Tfh cells following their interactions with LN GC-B cells from HIV-infected and HIV-uninfected individuals, rather than analyzing resting ex-vivo GC-Tfh cells. We observed that proliferating GC-Tfh cells from HIV-infected subjects were transcriptionally different than their HIV-uninfected counterparts, and displayed a significant downregulation of immune- and GC-Tfh-associated pathways and genes. Our results strongly demonstrated that MAF (coding for the transcription factor c-Maf) and its upstream signaling pathway mediators (IL6R and STAT3) were significantly downregulated in HIV-infected subjects, which could contribute to the impaired GC-Tfh and GC-B cell functions reported during infection. We further showed that c-Maf function was associated with the adenosine pathway and that the signaling upstream c-Maf could be partially restored by adenosine deaminase -1 (ADA-1) supplementation. Overall, we identified a novel mechanism that contributes to GC-Tfh cell impairment during HIV infection. Understanding how GC-Tfh cell function is altered in HIV is crucial and could provide critical information about the mechanisms leading to the development and maintenance of effective anti-HIV antibodies. Human immunodeficiency virus (HIV) remains a worldwide burden despite available treatments. The virus induces dysregulations in major immune cells and organs including lymph nodes. Germinal center T follicular helper (GC-Tfh) cells are immune cells which induce specific anti-HIV antibodies by helping GC-B cells. In chronic HIV, the interaction between these two cell types is defective, leading to modified and inefficient anti-HIV antibody responses. In this study, we examined the underlying mechanisms of this dysfunction. We observed that proliferating GC-Tfh cells from HIV-infected individuals, displayed distinctive gene expression than those from -uninfected subjects, following GC-B cell interaction. Furthermore, GC-Tfh cells from HIV patients showed a reduction in important immune-related pathway and gene expression. A number of essential GC-Tfh cell genes, such as MAF and its associated genes (IL6R and STAT3), were particularly attenuated in HIV, contributing to the impaired cells function. Moreover, we found an association between MAF function and the key enzyme adenosine deaminase-1 (ADA-1), where supplementation with ADA-1 partially restored the dysfunctional signaling in GC-Tfh cells during chronic infection. Understanding how GC-Tfh cells are altered in HIV is critical to elucidate the mechanisms leading to effective anti-HIV antibodies.
Collapse
Affiliation(s)
- Marita Chakhtoura
- Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Mike Fang
- Department of Population and Quantitative Health Services, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Rafael Cubas
- Iovance Biotherapeutics, San Carlos, California, United States of America
| | - Margaret H. O’Connor
- Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Molecular and Cellular Biology and Genetics, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Carmen N. Nichols
- Department of Population and Quantitative Health Services, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Brian Richardson
- Department of Population and Quantitative Health Services, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Aarthi Talla
- Allen Institute for Immunology, Seattle, Washington, United States of America
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mark J. Cameron
- Department of Population and Quantitative Health Services, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Virginie Tardif
- Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- Sorbonne University, INSERM, Center of Reasearch in Myology (Association Institut de Myologie) UMRS 974, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, Paris, France
- * E-mail: (VT); (EKH)
| | - Elias K. Haddad
- Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (VT); (EKH)
| |
Collapse
|
43
|
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: 0.5] [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
|
44
|
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: 12] [Impact Index Per Article: 3.0] [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
|
45
|
Hatic H, Sampat D, Goyal G. Immune checkpoint inhibitors in lymphoma: challenges and opportunities. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1037. [PMID: 34277837 PMCID: PMC8267255 DOI: 10.21037/atm-20-6833] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/25/2021] [Indexed: 12/21/2022]
Abstract
Immune checkpoint inhibitors (ICIs) are immunomodulatory antibodies that intensify the host immune response, thereby leading to cytotoxicity. The primary targets for checkpoint inhibition have included cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell death receptor-1 (PD-1) or programmed cell death ligand-1 (PD-L1). ICIs have resulted in a change in treatment landscape of various neoplasms. Among hematologic malignancies, ICIs have been most successful in certain subtypes of lymphomas such as classic Hodgkin lymphoma (cHL) and primary mediastinal B-cell lymphoma (PMBCL). However, there have been several challenges in harnessing the host immune system through ICI use in other lymphomas. The underlying reasons for the low efficacy of ICI monotherapy in most lymphomas may include defects in antigen presentation, non-inflamed tumor microenvironment (TME), immunosuppressive metabolites, genetic factors, and an overall lack of predictive biomarkers of response. In this review, we outline the existing and ongoing studies utilizing ICI therapy in various lymphomas. We also describe the challenges leading to the lack of efficacy with ICI use and discuss potential strategies to overcome those challenges including: chimeric antigen receptor T-cell therapy (CAR-T therapy), bispecific T-cell therapy (BiTE), lymphocyte activation gene-3 (LAG-3) inhibitors, T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) inhibitors, vaccines, promotion of inflammatory macrophages, indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors, DNA methyltransferase inhibitors (DNMTi) and histone deacetylase inhibitors (HDACi). Tumor mutational burden and interferon-gamma release assays are potential biomarkers of ICI treatment response beyond PD-L1 expression. Further collaborations between clinicians and scientists are vital to understand the immunopathology in ICI therapy in order to improve clinical outcomes.
Collapse
|
46
|
Elsner RA, Shlomchik MJ. Germinal Center and Extrafollicular B Cell Responses in Vaccination, Immunity, and Autoimmunity. Immunity 2021; 53:1136-1150. [PMID: 33326765 DOI: 10.1016/j.immuni.2020.11.006] [Citation(s) in RCA: 306] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/19/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Activated B cells participate in either extrafollicular (EF) or germinal center (GC) responses. Canonical responses are composed of a short wave of plasmablasts (PBs) arising from EF sites, followed by GC producing somatically mutated memory B cells (MBC) and long-lived plasma cells. However, somatic hypermutation (SHM) and affinity maturation can take place at both sites, and a substantial fraction of MBC are produced prior to GC formation. Infection responses range from GC responses that persist for months to persistent EF responses with dominant suppression of GCs. Here, we review the current understanding of the functional output of EF and GC responses and the molecular switches promoting them. We discuss the signals that regulate the magnitude and duration of these responses, and outline gaps in knowledge and important areas of inquiry. Understanding such molecular switches will be critical for vaccine development, interpretation of vaccine efficacy and the treatment for autoimmune diseases.
Collapse
Affiliation(s)
- Rebecca A Elsner
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Mark J Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15216, USA.
| |
Collapse
|
47
|
A follicular regulatory Innate Lymphoid Cell population impairs interactions between germinal center Tfh and B cells. Commun Biol 2021; 4:563. [PMID: 33980982 PMCID: PMC8115650 DOI: 10.1038/s42003-021-02079-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 03/26/2021] [Indexed: 12/16/2022] Open
Abstract
Innate Lymphoid Cells (ILCs) are immune cells typically found on mucosal surfaces and in secondary lymphoid organs where they regulate the immune response to pathogens. Despite their key role in the immune response, there are still fundamental gaps in our understanding of ILCs. Here we report a human ILC population present in the follicles of tonsils and lymph nodes termed follicular regulatory ILCs (ILCFR) that to our knowledge has not been previously identified. ILCFR have a distinct phenotype and transcriptional program when compared to other defined ILCs. Surprisingly, ILCFR inhibit the ability of follicular helper T (Tfh) cells to provide B cell help. The localization of ILCFR to the germinal centers suggests these cells may interfere with germinal center B cell (GC-B) and germinal center Tfh cell (GC-Tfh) interactions through the production of transforming growth factor beta (TGF-β. Intriguingly, under conditions of impaired GC-Tfh-GC-B cell interactions, such as human immunodeficiency virus (HIV) infection, the frequency of these cells is increased. Overall, we predict a role for ILCFR in regulating GC-Tfh-GC-B cell interactions and propose they expand in chronic inflammatory conditions. Margaret O’Connor et al. report a new Innate Lymphoid Cell population in human tonsils and lymph nodes that inhibit the functional interaction of follicular helper T cells and germinal center B cells. They show that this cell population is expanded under chronic HIV infection and results in decreased antibody production, suggesting a potential role for these cells in diseases with dysregulated immune responses.
Collapse
|
48
|
Wood MP, Jones CI, Lippy A, Oliver BG, Walund B, Fancher KA, Fisher BS, Wright PJ, Fuller JT, Murapa P, Habib J, Mavigner M, Chahroudi A, Sather DN, Fuller DH, Sodora DL. Rapid progression is associated with lymphoid follicle dysfunction in SIV-infected infant rhesus macaques. PLoS Pathog 2021; 17:e1009575. [PMID: 33961680 PMCID: PMC8133453 DOI: 10.1371/journal.ppat.1009575] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/19/2021] [Accepted: 04/20/2021] [Indexed: 12/27/2022] Open
Abstract
HIV-infected infants are at an increased risk of progressing rapidly to AIDS in the first weeks of life. Here, we evaluated immunological and virological parameters in 25 SIV-infected infant rhesus macaques to understand the factors influencing a rapid disease outcome. Infant macaques were infected with SIVmac251 and monitored for 10 to 17 weeks post-infection. SIV-infected infants were divided into either typical (TypP) or rapid (RP) progressor groups based on levels of plasma anti-SIV antibody and viral load, with RP infants having low SIV-specific antibodies and high viral loads. Following SIV infection, 11 out of 25 infant macaques exhibited an RP phenotype. Interestingly, TypP had lower levels of total CD4 T cells, similar reductions in CD4/CD8 ratios and elevated activation of CD8 T cells, as measured by the levels of HLA-DR, compared to RP. Differences between the two groups were identified in other immune cell populations, including a failure to expand activated memory (CD21-CD27+) B cells in peripheral blood in RP infant macaques, as well as reduced levels of germinal center (GC) B cells and T follicular helper (Tfh) cells in spleens (4- and 10-weeks post-SIV). Reduced B cell proliferation in splenic germinal GCs was associated with increased SIV+ cell density and follicular type 1 interferon (IFN)-induced immune activation. Further analyses determined that at 2-weeks post SIV infection TypP infants exhibited elevated levels of the GC-inducing chemokine CXCL13 in plasma, as well as significantly lower levels of viral envelope diversity compared to RP infants. Our findings provide evidence that early viral and immunologic events following SIV infection contributes to impairment of B cells, Tfh cells and germinal center formation, ultimately impeding the development of SIV-specific antibody responses in rapidly progressing infant macaques. Despite significant reductions in vertical HIV transmission, nearly 100,000 children succumb to AIDS-related illnesses each year. Indeed, infants face a disproportionately higher risk of progressing to AIDS, with roughly half of HIV+ infants exhibiting a rapid progression to AIDS-associated morbidity and mortality. Here, we evaluated immunological and virological parameters in 25 simian immunodeficiency virus (SIV)-infected infant rhesus macaques to assess the factors that influence a rapid disease outcome. Infant macaques were infected with simian immunodeficiency virus (SIV) and divided into either typical (TypP) or rapid (RP) progressor groups. RP infants exhibited low levels of plasma anti-SIV antibody and high viral loads. Following SIV infection, 11 out of 25 infant macaques exhibited an RP phenotype with some exhibiting AIDS-related symptoms. This study provides evidence that the low levels of anti-SIV antibodies are associated with impairments to both B and T cells in both blood and lymphoid tissues. These changes are associated with the prolonged expression of type 1 interferons which may be impeding development of a healthy humoral immune response in these rapidly progressing SIV-infected infant macaques. These findings have implications regarding potential therapeutic approaches to prevent rapid progression in HIV infected infants.
Collapse
Affiliation(s)
- Matthew P. Wood
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Chloe I. Jones
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Adriana Lippy
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Brian G. Oliver
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Brynn Walund
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Katherine A. Fancher
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Bridget S. Fisher
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Piper J. Wright
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - James T. Fuller
- University of Washington Department of Microbiology, Seattle, Washington, United States of America
| | - Patience Murapa
- University of Washington Department of Microbiology, Seattle, Washington, United States of America
- Washington National Primate Research Center, Seattle Washington, United States of America
| | - Jakob Habib
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Maud Mavigner
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, Georgia United States of America
| | - Ann Chahroudi
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, Georgia United States of America
| | - D. Noah Sather
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Deborah H. Fuller
- University of Washington Department of Microbiology, Seattle, Washington, United States of America
- Washington National Primate Research Center, Seattle Washington, United States of America
| | - Donald L. Sodora
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
49
|
Jha V, Nicholson LK, Gardner EM, Rahkola JT, Pratap H, Scott J, Borgeson M, Jacobelli J, Janoff EN. Impact of HIV-1 Infection and Antigen Class on T Follicular Helper Cell Responses to Pneumococcal Polysaccharide-Protein Conjugate Vaccine-13. THE JOURNAL OF IMMUNOLOGY 2021; 206:2402-2411. [PMID: 33931485 DOI: 10.4049/jimmunol.2001133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/04/2021] [Indexed: 11/19/2022]
Abstract
Pneumococcal infections are common and serious complications of HIV-1 disease. Prevention has been compromised by the limited magnitude and quality of Ab responses to T cell-independent type 2 pneumococcal capsular polysaccharides (PPS). The pneumococcal polysaccharide-protein conjugate vaccine-13 (PCV-13) contains PPS conjugated to the T cell-dependent protein (diphtheria toxoid [DT] [CRM197]). We investigated the differential response to PPS and DT by human Ab-secreting B cells (ASC) after immunization with PCV-13 in newly diagnosed healthy HIV+ and control adults. The numbers of PPS-specific IgG ASC increased significantly and similarly in HIV+ and controls. However, DT-specific IgG ASC increased in controls but not HIV+ subjects. To determine the cellular basis of these disparate responses to DT and PPS, we characterized the frequency and activation of T follicular helper (Tfh) cells, the predominant T cell subset providing B cell help. Expression of inducible T cell costimulator (ICOS), which sustains Tfh function and phenotype, increased significantly among controls, when compared with the HIV+ group. Increases in ICOS+ Tfh correlated with changes in T-dependent, DT-specific IgG ASC in controls but not in HIV+ In contrast, ICOS expression did not correlate with T cell-independent type 2 PPS-specific ASC in either group. Of note, upon optimized ex vivo stimulation, CD4 T cells from HIV+ subjects differentiated into Tfh cells and formed synapses with Raji B cells at frequencies similar to that of controls. In summary, PCV-13-induced increase in ICOS expression on Tfh was associated with responses to DT, which was compromised in recently diagnosed healthy HIV+ adults and can be restored ex vivo by providing effective Tfh-differentiating signals.
Collapse
Affiliation(s)
- Vibha Jha
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO.,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Lindsay K Nicholson
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO.,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | | | - Jeremy T Rahkola
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO.,Barbara Davis Center, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Harsh Pratap
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO.,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | | | - Mandy Borgeson
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO.,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Jordan Jacobelli
- Barbara Davis Center, University of Colorado Anschutz Medical Campus, Aurora, CO.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Edward N Janoff
- Mucosal and Vaccine Research Program Colorado, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO .,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
50
|
Catalán D, Mansilla MA, Ferrier A, Soto L, Oleinika K, Aguillón JC, Aravena O. Immunosuppressive Mechanisms of Regulatory B Cells. Front Immunol 2021; 12:611795. [PMID: 33995344 PMCID: PMC8118522 DOI: 10.3389/fimmu.2021.611795] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Regulatory B cells (Bregs) is a term that encompasses all B cells that act to suppress immune responses. Bregs contribute to the maintenance of tolerance, limiting ongoing immune responses and reestablishing immune homeostasis. The important role of Bregs in restraining the pathology associated with exacerbated inflammatory responses in autoimmunity and graft rejection has been consistently demonstrated, while more recent studies have suggested a role for this population in other immune-related conditions, such as infections, allergy, cancer, and chronic metabolic diseases. Initial studies identified IL-10 as the hallmark of Breg function; nevertheless, the past decade has seen the discovery of other molecules utilized by human and murine B cells to regulate immune responses. This new arsenal includes other anti-inflammatory cytokines such IL-35 and TGF-β, as well as cell surface proteins like CD1d and PD-L1. In this review, we examine the main suppressive mechanisms employed by these novel Breg populations. We also discuss recent evidence that helps to unravel previously unknown aspects of the phenotype, development, activation, and function of IL-10-producing Bregs, incorporating an overview on those questions that remain obscure.
Collapse
Affiliation(s)
- Diego Catalán
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Instituto Milenio en Inmunología e Inmunoterapia, Santiago, Chile
| | - Miguel Andrés Mansilla
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Ashley Ferrier
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Instituto Milenio en Inmunología e Inmunoterapia, Santiago, Chile
| | - Lilian Soto
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Unidad de Dolor, Hospital Clínico, Universidad de Chile (HCUCH), Santiago, Chile
| | | | - Juan Carlos Aguillón
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Octavio Aravena
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| |
Collapse
|