1
|
Nelson CS, Podestà MA, Gempler MG, Lee JM, Batty CJ, Mathenge PG, Sainju A, Chang MR, Ke H, Chandrakar P, Bechu E, Richardson S, Cavazzoni CB, Tullius SG, Abdi R, Ghebremichael M, Haigis MC, Marasco WA, Sage PT. The inflammaging microenvironment induces dysfunctional rewiring of Tfh cell differentiation. JCI Insight 2025; 10:e187271. [PMID: 40036082 PMCID: PMC12016926 DOI: 10.1172/jci.insight.187271] [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: 09/19/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025] Open
Abstract
Humoral immunity is orchestrated by follicular helper T (Tfh) cells, which promote cognate B cells to produce high-affinity, protective antibodies. In aged individuals, humoral immunity after vaccination is diminished despite the presence of Tfh cells, suggesting defects after initial Tfh cell formation. In this study, we utilized both murine and human systems to investigate how aging alters Tfh cell differentiation after influenza vaccination. We found that young Tfh cells underwent progressive differentiation after influenza vaccination, culminating in clonal expansion of effector-like cells in both draining lymph nodes and blood. In aging, early stages of Tfh cell development occurred normally. However, aging rewired the later stages of development in Tfh cells, resulting in a transcriptional program reflective of cellular senescence, sustained pro-inflammatory cytokine production, and metabolic reprogramming. We investigated the extent to which this rewiring of aged Tfh cells is due to the age-associated inflammatory ("inflammaging") microenvironment and found that this setting was sufficient to both block the transition of Tfh cells to a post-effector resting state and skew Tfh cells toward the age-rewired state. Together, these data suggest that aging dampens humoral immunity by cytokine-mediated rewiring of late effector Tfh cell differentiation into an activated, yet less functional, cellular state.
Collapse
Affiliation(s)
- Cody S. Nelson
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine; and
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Manuel A. Podestà
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine; and
- Unit of Nephrology, Dialysis, and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maya G. Gempler
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine; and
| | - Jeong-Mi Lee
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine; and
| | - Cole J. Batty
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Peterson G. Mathenge
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Asra Sainju
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Matthew R. Chang
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Hanzhong Ke
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Pragya Chandrakar
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine; and
| | - Elsa Bechu
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine; and
| | - Sierra Richardson
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine; and
| | - Cecilia B. Cavazzoni
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine; and
| | - Stefan G. Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Reza Abdi
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine; and
| | - Musie Ghebremichael
- Ragon Institute of Mass General Brigham, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Marcia C. Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Wayne A. Marasco
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Peter T. Sage
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine; and
| |
Collapse
|
2
|
Gutierrez AH, Terry FE, Rosenberg AS, Martin WD, De Groot AS. Regulatory T cell epitope content in human antibodies decreases during maturation. Front Immunol 2025; 16:1535826. [PMID: 40313951 PMCID: PMC12043479 DOI: 10.3389/fimmu.2025.1535826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/24/2025] [Indexed: 05/03/2025] Open
Abstract
Introduction Antibody maturation in the lymphoid follicle produces antibodies with improved binding affinity. This process requires iterative rounds of mutation and B cell expansion, supported by T cells that recognize epitopes presented on the B cell's MHC-II. In this comprehensive antibody repertoire analysis, we find that established regulatory T cell epitopes (Tregitopes) decline in maturing antibody sequences as somatic hypermutation (SHM) increases, but potential T effector epitopes do not decline. A previous analysis of B cell receptor (BCR)-derived HLA-DR epitopes present in memory antibody repertoires from seven healthy human donors revealed a decrease in donor-specific epitope content with SHM. Moreover, T cell epitope depletion was associated with class-switching and long-term secretion of antibody into serum. Significant depletion of high-affinity germline-encoded epitopes in high SHM sequences was also observed, but the predicted phenotype of T cells responding to the BCR-derived epitopes (regulatory vs. effector) was not previously evaluated. Methods In this follow-on study, we screened a different set of four donor repertoires to investigate the dynamics of donor-specific HLA-DR T cell epitopes and three subsets of T cell epitope content: previously validated T cell epitopes recognized by thymus-derived Tregs (Tregitopes), potentially tolerated T cell epitopes, and potential effector T cell epitopes. Results Our results show that Tregitope content reduction is correlated with SHM, suggesting that Tregitopes are removed during maturation. Moreover, T cell epitopes that are likely to be tolerated or tolerogenic were also removed with SHM progression. In contrast, potential T effector epitope content increased with SHM. Tregitope depletion occurred in multiple V-gene pair combinations and was the most frequent T cell epitope change. Furthermore, Tregitope content in IgA and IgG sequences was lower and had greater negative correlation with SHM than IgM, indicating that Tregitope removal is likely associated with class-switching. Tregitope depletion was also associated with maturation to plasmablasts. In vitro, representative Tregitopes inhibited CD4+ T cell proliferation. Mutations introduced by SHM altered Tregitope HLA-DR binding affinities. Discussion The correlation of Tregitope depletion with increasing SHM implies that the activity of thymus-derived Treg cells in immune responses to antibodies is diminished with SHM, maturation, and isotype switching, supporting the generation of anti-idiotype responses.
Collapse
|
3
|
Zhang H, Podestà MA, Cavazzoni CB, Wu Y, Lee JM, Li X, Raeder PL, Chandrakar P, Gempler M, Richardson S, Ghosh D, Sayin I, Blazar BR, Abdi R, Weins A, Chong AS, Sage PT. Follicular regulatory T cells restrain kidney allograft rejection in mice by suppressing alloreactive B cells. Nat Commun 2025; 16:2151. [PMID: 40038336 DOI: 10.1038/s41467-025-57468-4] [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: 03/24/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
Pathogenic antibodies produced by alloreactive B cells mediate antibody-mediated rejection after kidney transplantation, but the mechanisms remain poorly understood. Follicular regulatory T (Tfr) cells modulate follicular helper T cell-mediated B cell responses, but the functions of Tfr in controlling alloreactive antibody are unknown. Here we study the developmental signals and functions of Tfr cells in mouse allogeneic kidney transplantation models, and show that costimulatory blockade alters the development of Tfr cells disproportionately by decreasing germinal center (GC)-like Tfr cells but increasing follicular-like Tfr cells. Functionally, global Tfr cell deletion results in accelerated graft rejection and increases in donor-specific B cells in both draining lymph nodes and kidney allografts. Mechanistically, Tfr cell deletion increases GC B cell expression of pro-inflammatory cytokines such as IL-15, while neutralization of IL-15 compensates for the loss of Tfr cells and prolongs the survival of mice receiving kidney transplants. Together our preclinical mouse data demonstrate how Tfr restrains kidney allograft rejection by limiting alloreactive B cell responses.
Collapse
Affiliation(s)
- Hengcheng Zhang
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Manuel A Podestà
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cecilia B Cavazzoni
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yumeng Wu
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeong-Mi Lee
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofei Li
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paulo Lisboa Raeder
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Pragya Chandrakar
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maya Gempler
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sierra Richardson
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Deepjyoti Ghosh
- Section of Rheumatology and The Knapp Center for Lupus and Immunology Research, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Ismail Sayin
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, IL, USA
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapies, University of Minnesota, Minneapolis, MN, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Astrid Weins
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anita S Chong
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, IL, USA
| | - Peter T Sage
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Chandrakar P, Nelson CS, Podestà MA, Cavazzoni CB, Gempler M, Lee JM, Richardson S, Zhang H, Samarpita S, Ciofani M, Chatila T, Kuchroo VK, Sage PT. Progressively differentiated T FH13 cells are stabilized by JunB to mediate allergen germinal center responses. Nat Immunol 2025; 26:473-483. [PMID: 39891019 DOI: 10.1038/s41590-025-02077-y] [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: 04/29/2024] [Accepted: 01/06/2025] [Indexed: 02/03/2025]
Abstract
Allergic diseases are common and affect a large proportion of the population. Interleukin-13 (IL-13)-expressing follicular helper T (TFH13) cells are a newly identified population of TFH cells that have been associated with high-affinity IgE responses. However, the origins, developmental signals, transcriptional programming and precise functions of TFH13 cells are unknown. Here, we examined the developmental signals for TFH13 cells and found a direct and progressive differentiation pathway marked by the production of IL-21. These two pathways differed in kinetics and extrinsic requirements. However, both pathways converged, forming transcriptionally similar TFH13 cells that express the transcription factor JunB as a critical stabilizing factor. Using an intersectional genetics-based TFH13-diphtheria toxin receptor model to perturb these cells, we found that TFH13 cells were essential to drive broad germinal center responses and allergen-specific IgG and IgE. Moreover, we found that IL-21 is a broad positive regulator of allergen germinal center B cells and synergizes with IL-13 produced by TFH13 cells to amplify allergic responses. Thus, TFH13 cells orchestrate multiple features of allergic inflammation.
Collapse
Affiliation(s)
- Pragya Chandrakar
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cody S Nelson
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Manuel A Podestà
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Unit of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cecilia B Cavazzoni
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maya Gempler
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeong-Mi Lee
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sierra Richardson
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hengcheng Zhang
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Snigdha Samarpita
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Ciofani
- Department of Integrative Immunology, Duke University Medical Center, Durham, NC, USA
| | - Talal Chatila
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vijay K Kuchroo
- Gene Lay Institute of Immunology and Inflammatory Diseases, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Peter T Sage
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Jiang Y, Sanyal M, Hussein NA, Baghdasaryan A, Zhang M, Wang F, Ren F, Li J, Zhu G, Meng Y, Adamska JZ, Mellins E, Dai H. A SARS-CoV-2 vaccine on an NIR-II/SWIR emitting nanoparticle platform. SCIENCE ADVANCES 2025; 11:eadp5539. [PMID: 39919189 PMCID: PMC11804919 DOI: 10.1126/sciadv.adp5539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025]
Abstract
The COVID-19 pandemic caused a global health crisis that resulted in millions of deaths. Effective vaccines have played central roles in curtailing the pandemic. Here, we developed a down-converting near-infrared IIb (NIR-IIb; 1500 to 1700 nanometers) luminescent, pure NaErF4@NaYF4 rare-earth nanoparticle (pEr) as vaccine carriers. The pEr nanoparticles were coated with three layers of cross-linked biocompatible polymers (pEr-P3; ~55 nanometers) and conjugated to the receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. Upon subcutaneous injection of the pEr-P3-RBD nanovaccine in mice, in vivo NIR-IIb imaging revealed active vaccine trafficking and migration to lymph nodes through lymphatic vessels. Two doses of the adjuvant-free vaccine elicited long-lasting (>7 months) high titers of serum viral neutralization antibody and anti-RBD immunoglobulin G, along with robust RBD-specific germinal center B cells and T follicular helper cells. We devised in vivo NIR-II molecular imaging of RBD-specific cells in lymph nodes, opening noninvasive assessments of vaccine-elicited immune responses longitudinally.
Collapse
Affiliation(s)
- Yingying Jiang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
| | - Mrinmoy Sanyal
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Noor A. Hussein
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Ani Baghdasaryan
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Mengzhen Zhang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Feifei Wang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Fuqiang Ren
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Jiachen Li
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Guanzhou Zhu
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Yifan Meng
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Julia Zofia Adamska
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Hongjie Dai
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
| |
Collapse
|
6
|
Kim D, Kim J, Yeo H, Chung Y. Immunometabolic regulation of germinal centers and its implications for aging. Curr Opin Immunol 2024; 91:102485. [PMID: 39357081 DOI: 10.1016/j.coi.2024.102485] [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: 07/02/2024] [Revised: 09/02/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
Aging, metabolism, and immunity have long been considered distinct domains. Aging is primarily associated with the gradual decline of physiological functions, metabolism regulates energy production and maintains cellular processes, and the immune system manages innate and adaptive responses against pathogens and vaccines. However, recent studies have revealed that these three systems are intricately interconnected, collectively influencing an individual's response to stress and disease. This review explores the interplay between immunometabolism, T follicular helper cells, B cells, and aging, focusing on how these interactions impact immune function in the elderly.
Collapse
Affiliation(s)
- Daehong Kim
- Institute of Pharmaceutical Sciences, College of Pharmacy, and Seoul National University, Seoul 08826, Seoul, Republic of Korea; BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul 08826, Seoul, Republic of Korea
| | - Jaemin Kim
- Institute of Pharmaceutical Sciences, College of Pharmacy, and Seoul National University, Seoul 08826, Seoul, Republic of Korea; BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul 08826, Seoul, Republic of Korea
| | - Hyeonuk Yeo
- Institute of Pharmaceutical Sciences, College of Pharmacy, and Seoul National University, Seoul 08826, Seoul, Republic of Korea; BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul 08826, Seoul, Republic of Korea
| | - Yeonseok Chung
- Institute of Pharmaceutical Sciences, College of Pharmacy, and Seoul National University, Seoul 08826, Seoul, Republic of Korea; BK21 Plus Program, College of Pharmacy, Seoul National University, Seoul 08826, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Marchese AM, Fries L, Beyhaghi H, Vadivale M, Zhu M, Cloney-Clark S, Plested JS, Chung AW, Dunkle LM, Kalkeri R. Mechanisms and implications of IgG4 responses to SARS-CoV-2 and other repeatedly administered vaccines. J Infect 2024; 89:106317. [PMID: 39419185 DOI: 10.1016/j.jinf.2024.106317] [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: 08/22/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Vaccine-induced immunoglobulin G (IgG) profiles can vary with respect to the predominant subclasses that characterize the response. Among IgG subclasses, IgG4 is reported to have anti-inflammatory properties, but can also exhibit reduced capacity for virus neutralization and activation of Fc-dependent effector functions. Here, we review evidence that IgG4 subclass responses can be disproportionately increased in response to some types of vaccines targeting an array of diseases, including pertussis, HIV, malaria, and COVID-19. The basis for enhanced IgG4 induction by vaccines is poorly understood but may be associated with platform- or dose regimen-specific differences in antigen exposure and/or cytokine stimulation. The clinical implications of vaccine-induced IgG4 responses remain uncertain, though collective evidence suggests that proportional increases in IgG4 might reduce vaccine antigen-specific immunity. Additional work is needed to determine underlying mechanisms and to elucidate what role IgG4 may play in modifications of vaccine-induced immunity to disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Amy W Chung
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3000, Australia
| | | | | |
Collapse
|
8
|
Fahlquist-Hagert C, Wittenborn TR, Pedersen MK, Jensen L, Degn SE. T-follicular regulatory cells expand to control germinal center plasma cell output but fail to curb autoreactivity. iScience 2024; 27:110887. [PMID: 39319261 PMCID: PMC11417334 DOI: 10.1016/j.isci.2024.110887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/17/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024] Open
Abstract
Autoantibodies generated in germinal centers (GCs) contribute to the pathogenesis of autoimmune diseases. GCs are controlled by specialized FoxP3+ T-follicular regulatory cells (Tfr), but their role in established autoimmunity is unclear. We generated autoimmune bone marrow chimeras in which Tfr could be specifically ablated by diphtheria toxin. Furthermore, we tracked the clonal persistence and evolution of Tfr populations using Confetti reporters. Ablation of Tfr caused increased early plasma cell output, but longer-term ablation did not increase plasma cell levels beyond those of Tfr-sufficient controls, suggesting that Tfr fail to contain chronic autoreactive GC responses. In agreement, Tfr were robustly induced in early autoreactive GCs but then waned. Moreover, we observed polyclonal Tfr expansion when ablating part of the Tfr subset. Hence, under homeostatic conditions, a polyclonal population of Tfr operates to control autoreactivity by limiting the output of plasma cells from GCs, but in chronic autoimmunity, this mechanism fails.
Collapse
Affiliation(s)
- Cecilia Fahlquist-Hagert
- Laboratory for Lymphocyte Biology, Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Thomas Rea Wittenborn
- Laboratory for Lymphocyte Biology, Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Mattias Krogh Pedersen
- Laboratory for Lymphocyte Biology, Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Lisbeth Jensen
- Laboratory for Lymphocyte Biology, Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Søren Egedal Degn
- Laboratory for Lymphocyte Biology, Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| |
Collapse
|
9
|
Sircy LM, Ramstead AG, Gibbs LC, Joshi H, Baessler A, Mena I, García-Sastre A, Emerson LL, Fairfax KC, Williams MA, Hale JS. Generation of antigen-specific memory CD4 T cells by heterologous immunization enhances the magnitude of the germinal center response upon influenza infection. PLoS Pathog 2024; 20:e1011639. [PMID: 39283916 PMCID: PMC11404825 DOI: 10.1371/journal.ppat.1011639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 08/05/2024] [Indexed: 09/22/2024] Open
Abstract
Current influenza vaccine strategies have yet to overcome significant obstacles, including rapid antigenic drift of seasonal influenza viruses, in generating efficacious long-term humoral immunity. Due to the necessity of germinal center formation in generating long-lived high affinity antibodies, the germinal center has increasingly become a target for the development of novel or improvement of less-efficacious vaccines. However, there remains a major gap in current influenza research to effectively target T follicular helper cells during vaccination to alter the germinal center reaction. In this study, we used a heterologous infection or immunization priming strategy to seed an antigen-specific memory CD4+ T cell pool prior to influenza infection in mice to evaluate the effect of recalled memory T follicular helper cells in increased help to influenza-specific primary B cells and enhanced generation of neutralizing antibodies. We found that heterologous priming with intranasal infection with acute lymphocytic choriomeningitis virus (LCMV) or intramuscular immunization with adjuvanted recombinant LCMV glycoprotein induced increased antigen-specific effector CD4+ T and B cellular responses following infection with a recombinant influenza strain that expresses LCMV glycoprotein. Heterologously primed mice had increased expansion of secondary Th1 and Tfh cell subsets, including increased CD4+ TRM cells in the lung. However, the early enhancement of the germinal center cellular response following influenza infection did not impact influenza-specific antibody generation or B cell repertoires compared to primary influenza infection. Overall, our study suggests that while heterologous infection or immunization priming of CD4+ T cells is able to enhance the early germinal center reaction, further studies to understand how to target the germinal center and CD4+ T cells specifically to increase long-lived antiviral humoral immunity are needed.
Collapse
Affiliation(s)
- Linda M. Sircy
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Andrew G. Ramstead
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Lisa C. Gibbs
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Hemant Joshi
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Andrew Baessler
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lyska L. Emerson
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Keke C. Fairfax
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Matthew A. Williams
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - J. Scott Hale
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
10
|
Song Y, Wang J, Yang Z, He Q, Bao C, Xie Y, Sun Y, Li S, Quan Y, Yang H, Li C. Heterologous booster vaccination enhances antibody responses to SARS-CoV-2 by improving Tfh function and increasing B-cell clonotype SHM frequency. Front Immunol 2024; 15:1406138. [PMID: 38975334 PMCID: PMC11224535 DOI: 10.3389/fimmu.2024.1406138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
Heterologous prime-boost has broken the protective immune response bottleneck of the COVID-19 vaccines. however, the underlying mechanisms have not been fully elucidated. Here, we investigated antibody responses and explored the response of germinal center (GC) to priming with inactivated vaccines and boosting with heterologous adenoviral-vectored vaccines or homologous inactivated vaccines in mice. Antibody responses were dramatically enhanced by both boosting regimens. Heterologous immunization induced more robust GC activation, characterized by increased Tfh cell populations and enhanced helper function. Additionally, increased B-cell activation and antibody production were observed in a heterologous regimen. Libra-seq was used to compare the differences of S1-, S2- and NTD-specific B cells between homologous and heterologous vaccination, respectively. S2-specific CD19+ B cells presented increased somatic hypermutations (SHMs), which were mainly enriched in plasma cells. Moreover, a heterologous booster dose promoted the clonal expansion of B cells specific to S2 and NTD regions. In conclusion, the functional role of Tfh and B cells following SARS-CoV-2 heterologous vaccination may be important for modulating antibody responses. These findings provide new insights for the development of SARS-CoV-2 vaccines that induce more robust antibody response.
Collapse
Affiliation(s)
- Yanli Song
- Division of the Second Vaccines, Wuhan Institute of Biological Products Co. Ltd., Wuhan, China
| | - Jiaolei Wang
- Divsion of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Zhihui Yang
- Division of the Second Vaccines, Wuhan Institute of Biological Products Co. Ltd., Wuhan, China
| | - Qian He
- Divsion of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Chunting Bao
- Divsion of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Ying Xie
- Divsion of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Yufang Sun
- Divsion of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Shuyan Li
- Divsion of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Yaru Quan
- Divsion of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Huijie Yang
- Divsion of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Changgui Li
- Divsion of Respiratory Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
11
|
Roe K. Immunoregulatory natural killer cells. Clin Chim Acta 2024; 558:117896. [PMID: 38583553 DOI: 10.1016/j.cca.2024.117896] [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: 02/29/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/09/2024]
Abstract
This review discusses a broader scope of functional roles for NK cells. Despite the well-known cytolytic and inflammatory roles of NK cells against tumors and pathogenic diseases, extensive evidence demonstrates certain subsets of NK cells have defacto immunoregulatory effects and have a role in inducing anergy or lysis of antigen-activated T cells and regulating several autoimmune diseases. Furthermore, recent evidence suggests certain subsets of immunoregulatory NK cells can cause anergy or lysis of antigen-activated T cells to regulate hyperinflammatory diseases, including multisystem inflammatory syndrome. Several pathogens induce T cell and NK cell exhaustion and/or suppression, which impair the immune system's control of the replication speed of virulent pathogens and tumors and result in extensive antigens and antigen-antibody immune complexes, potentially inducing to some extent a Type III hypersensitivity immune reaction. The Type III hypersensitivity immune reaction induces immune cell secretion of proteinases, which can cleave specific proteins to create autoantigens which activate T cells to initiate autoimmune and/or hyperinflammatory diseases. Furthermore, pathogen induced NK cell exhaustion and/or suppression will inhibit NK cells which would have induced the anergy or lysis of activated T cells to regulate autoimmune and hyperinflammatory diseases. Autoimmune and hyperinflammatory diseases can be consequences of the dual lymphocyte exhaustion and/or suppression effects during infections, by creating autoimmune and/or hyperinflammatory diseases, while also impairing immunoregulatory lymphocytes which otherwise would have regulated these diseases.
Collapse
Affiliation(s)
- Kevin Roe
- Retired USPTO, San Jose, CA, United States of America.
| |
Collapse
|
12
|
Akama-Garren EH, Yin X, Prestwood TR, Ma M, Utz PJ, Carroll MC. T cell help shapes B cell tolerance. Sci Immunol 2024; 9:eadj7029. [PMID: 38363829 PMCID: PMC11095409 DOI: 10.1126/sciimmunol.adj7029] [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: 07/11/2023] [Accepted: 12/29/2023] [Indexed: 02/18/2024]
Abstract
T cell help is a crucial component of the normal humoral immune response, yet whether it promotes or restrains autoreactive B cell responses remains unclear. Here, we observe that autoreactive germinal centers require T cell help for their formation and persistence. Using retrogenic chimeras transduced with candidate TCRs, we demonstrate that a follicular T cell repertoire restricted to a single autoreactive TCR, but not a foreign antigen-specific TCR, is sufficient to initiate autoreactive germinal centers. Follicular T cell specificity influences the breadth of epitope spreading by regulating wild-type B cell entry into autoreactive germinal centers. These results demonstrate that TCR-dependent T cell help can promote loss of B cell tolerance and that epitope spreading is determined by TCR specificity.
Collapse
Affiliation(s)
- Elliot H. Akama-Garren
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, MA 02115, USA
| | - Xihui Yin
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tyler R. Prestwood
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Minghe Ma
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Paul J. Utz
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael C. Carroll
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
13
|
Ke F, Benet ZL, Shelyakin P, Britanova OV, Gupta N, Dent AL, Moore BB, Grigorova IL. Targeted checkpoint control of B cells undergoing positive selection in germinal centers by follicular regulatory T cells. Proc Natl Acad Sci U S A 2024; 121:e2304020121. [PMID: 38261619 PMCID: PMC10835130 DOI: 10.1073/pnas.2304020121] [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: 03/10/2023] [Accepted: 11/20/2023] [Indexed: 01/25/2024] Open
Abstract
Follicular regulatory T cells (Tfr) can play opposite roles in the regulation of germinal center (GC) responses. Depending on the studies, Tfr suppress or support GC and B cell affinity maturation. However, which factors determine positive vs. negative effects of Tfr on the GC B cell is unclear. In this study, we show that GC centrocytes that express MYC up-regulate expression of CCL3 chemokine that is needed for both the positive and negative regulation of GC B cells by Tfr. B cell-intrinsic expression of CCL3 contributes to Tfr-dependent positive selection of foreign Ag-specific GC B cells. At the same time, expression of CCL3 is critical for direct Tfr-mediated suppression of GC B cells that acquire cognate to Tfr nuclear proteins. Our study suggests that CCR5 and CCR1 receptors promote Tfr migration to CCL3 and highlights Ccr5 expression on the Tfr subset that expresses Il10. Based on our findings and previous studies, we suggest a model of chemotactically targeted checkpoint control of B cells undergoing positive selection in GCs by Tfr, where Tfr directly probe and license foreign antigen-specific B cells to complete their positive selection in GCs but, at the same time, suppress GC B cells that present self-antigens cognate to Tfr.
Collapse
Affiliation(s)
- Fang Ke
- Department of Microbiology and Immunology, Michigan Medicine University of Michigan, Ann Arbor, MI48109
| | - Zachary L. Benet
- Department of Microbiology and Immunology, Michigan Medicine University of Michigan, Ann Arbor, MI48109
| | - Pavel Shelyakin
- Abu Dhabi Stem Cells Center, Abu Dhabi4600, United Arab Emirates
- Molecular Technologies Division, Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow117997, Russian Federation
| | - Olga V. Britanova
- Molecular Technologies Division, Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow117997, Russian Federation
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow117997, Russian Federation
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel24105, Germany
| | - Neetu Gupta
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Alexander L. Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN46123
| | - Bethany B. Moore
- Department of Microbiology and Immunology, Michigan Medicine University of Michigan, Ann Arbor, MI48109
- Department of Internal Medicine, Michigan Medicine University of Michigan, Ann Arbor, MI48109
| | - Irina L. Grigorova
- Department of Microbiology and Immunology, Michigan Medicine University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
14
|
Aleman A, van Kesteren M, Zajdman AK, Srivastava K, Cognigni C, Mischka J, Chen LY, Upadhyaya B, Serebryakova K, Nardulli JR, Lyttle N, Kappes K, Jackson H, Gleason CR, Oostenink A, Cai GY, Van Oekelen O, van Bakel H, Sordillo EM, Cordon-Cardo C, Merad M, Jagannath S, Wajnberg A, Simon V, Parekh S. Cellular mechanisms associated with sub-optimal immune responses to SARS-CoV-2 bivalent booster vaccination in patients with Multiple Myeloma. EBioMedicine 2023; 98:104886. [PMID: 37995467 PMCID: PMC10708991 DOI: 10.1016/j.ebiom.2023.104886] [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: 08/23/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND The real-world impact of bivalent vaccines for wild type (WA.1) and Omicron variant (BA.5) is largely unknown in immunocompromised patients with Multiple Myeloma (MM). We characterize the humoral and cellular immune responses in patients with MM before and after receiving the bivalent booster, including neutralizing assays to identify patterns associated with continuing vulnerability to current variants (XBB1.16, EG5) in the current post-pandemic era. METHODS We studied the humoral and cellular immune responses before and after bivalent booster immunization in 48 MM patients. Spike binding IgG antibody levels were measured by SARS-CoV-2 spike binding ELISA and neutralization capacity was assessed by a SARS-CoV-2 multi-cycle microneutralization assays to assess inhibition of live virus. We measured spike specific T-cell function using the QuantiFERON SARS-CoV-2 (Qiagen) assay as well as flow-cytometry based T-cell. In a subset of 38 patients, high-dimensional flow cytometry was performed to identify immune cell subsets associated with lack of humoral antibodies. FINDINGS We find that bivalent vaccination provides significant boost in protection to the omicron variant in our MM patients, in a treatment specific manner. MM patients remain vulnerable to newer variants with mutations in the spike portion. Anti-CD38 and anti-BCMA therapies affect the immune machinery needed to produce antibodies. INTERPRETATION Our study highlights varying immune responses observed in MM patients after receiving bivalent COVID-19 vaccination. Specifically, a subgroup of MM patients undergoing anti-CD38 and anti-BCMA therapy experience impairment in immune cells such DCs, B cells, NK cells and TFH cells, leading to an inability to generate adequate humoral and cellular responses to vaccination. FUNDING National Cancer Institute (National Institutes of Health), National Institute of Allergy and Infectious Diseases (National Institutes of Health), NCI Serological Sciences Network for COVID-19 (SeroNet) and The Icahn School of Medicine at Mount Sinai.
Collapse
Affiliation(s)
- Adolfo Aleman
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, 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
| | - Morgan van Kesteren
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ariel Kogan Zajdman
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Komal Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christian Cognigni
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jacob Mischka
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lucia Y Chen
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Bhaskar Upadhyaya
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kseniya Serebryakova
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica R Nardulli
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Neko Lyttle
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katerina Kappes
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hayley Jackson
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charles R Gleason
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Annika Oostenink
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gianna Y Cai
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oliver Van Oekelen
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Harm van Bakel
- Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Emilia Mia Sordillo
- Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, 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; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sundar Jagannath
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ania Wajnberg
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Infectious Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogen Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Samir Parekh
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, 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.
| |
Collapse
|
15
|
Podestà MA, Cavazzoni CB, Hanson BL, Bechu ED, Ralli G, Clement RL, Zhang H, Chandrakar P, Lee JM, Reyes-Robles T, Abdi R, Diallo A, Sen DR, Sage PT. Stepwise differentiation of follicular helper T cells reveals distinct developmental and functional states. Nat Commun 2023; 14:7712. [PMID: 38001088 PMCID: PMC10674016 DOI: 10.1038/s41467-023-43427-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Follicular helper T (Tfh) cells are essential for the formation of high affinity antibodies after vaccination or infection. Although the signals responsible for initiating Tfh differentiation from naïve T cells have been studied, the signals controlling sequential developmental stages culminating in optimal effector function are not well understood. Here we use fate mapping strategies for the cytokine IL-21 to uncover sequential developmental stages of Tfh differentiation including a progenitor-like stage, a fully developed effector stage and a post-effector Tfh stage that maintains transcriptional and epigenetic features without IL-21 production. We find that progression through these stages are controlled intrinsically by the transcription factor FoxP1 and extrinsically by follicular regulatory T cells. Through selective deletion of Tfh stages, we show that these cells control antibody dynamics during distinct stages of the germinal center reaction in response to a SARS-CoV-2 vaccine. Together, these studies demonstrate the sequential phases of Tfh development and how they promote humoral immunity.
Collapse
Affiliation(s)
- Manuel A Podestà
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Renal Division, Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| | - Cecilia B Cavazzoni
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin L Hanson
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elsa D Bechu
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Garyfallia Ralli
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachel L Clement
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hengcheng Zhang
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pragya Chandrakar
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeong-Mi Lee
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alos Diallo
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Debattama R Sen
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Peter T Sage
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Roe K. Pathogen regulatory RNA usage enables chronic infections, T-cell exhaustion and accelerated T-cell exhaustion. Mol Cell Biochem 2023; 478:2505-2516. [PMID: 36941498 PMCID: PMC10027582 DOI: 10.1007/s11010-023-04680-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 02/15/2023] [Indexed: 03/23/2023]
Abstract
Pathogens evade or disable cellular immune defenses using regulatory ribonucleic acids (RNAs), including microRNAs and long non-coding RNAs. Pathogenic usage of regulatory RNA enables chronic infections. Chronic infections, using host regulatory RNAs and/or creating pathogenic regulatory RNAs against cellular defenses, can cause T-cell exhaustion and latent pathogen reactivations. Concurrent pathogen infections of cells enable several possibilities. A first pathogen can cause an accelerated T-cell exhaustion for a second pathogen cellular infection. Accelerated T-cell exhaustion for the second pathogen weakens T-cell targeting of the second pathogen and enables a first-time infection by the second pathogen to replicate quickly and extensively. This can induce a large antibody population, which may be inadequately targeted against the second pathogen. Accelerated T-cell exhaustion can explain the relatively short median and average times from diagnosis to mortality in some viral epidemics, e.g., COVID-19, where the second pathogen can lethally overwhelm individuals' immune defenses. Alternatively, if an individual survives, the second pathogen could induce a very high titer of antigen-antibody immune complexes. If the antigen-antibody immune complex titer quickly becomes very high, it can exceed the immune system's phagocytic capability in immuno-deficient individuals, resulting in a Type III hypersensitivity immune reaction. Accelerated T-cell exhaustion in immuno-deficient individuals can be a fundamental cause of several hyperinflammatory diseases and autoimmune diseases. This would be possible when impaired follicular helper CD4+ T-cell assistance to germinal center B-cell somatic hypermutation, affinity maturation and isotype switching of antibodies results in high titers of inadequate antibodies, and this initiates a Type III hypersensitivity immune reaction with proteinase releases which express or expose autoantigens.
Collapse
|
17
|
Zhang H, Cavazzoni CB, Podestà MA, Bechu ED, Ralli G, Chandrakar P, Lee JM, Sayin I, Tullius SG, Abdi R, Chong AS, Blazar BR, Sage PT. IL-21-producing effector Tfh cells promote B cell alloimmunity in lymph nodes and kidney allografts. JCI Insight 2023; 8:e169793. [PMID: 37870962 PMCID: PMC10619486 DOI: 10.1172/jci.insight.169793] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/12/2023] [Indexed: 10/25/2023] Open
Abstract
Follicular helper T (Tfh) cells have been implicated in controlling rejection after allogeneic kidney transplantation, but the precise subsets, origins, and functions of Tfh cells in this process have not been fully characterized. Here we show that a subset of effector Tfh cells marked by previous IL-21 production is potently induced during allogeneic kidney transplantation and is inhibited by immunosuppressive agents. Single-cell RNA-Seq revealed that these lymph node (LN) effector Tfh cells have transcriptional and clonal overlap with IL-21-producing kidney-infiltrating Tfh cells, implicating common origins and developmental trajectories. To investigate the precise functions of IL-21-producing effector Tfh cells in LNs and allografts, we used a mouse model to selectively eliminate these cells and assessed allogeneic B cell clonal dynamics using a single B cell culture system. We found that IL-21-producing effector Tfh cells were essential for transplant rejection by regulating donor-specific germinal center B cell clonal dynamics both systemically in the draining LN and locally within kidney grafts. Thus, IL-21-producing effector Tfh cells have multifaceted roles in Ab-mediated rejection after kidney transplantation by promoting B cell alloimmunity.
Collapse
Affiliation(s)
- Hengcheng Zhang
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Cecilia B. Cavazzoni
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Manuel A. Podestà
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Elsa D. Bechu
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Garyfallia Ralli
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pragya Chandrakar
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeong-Mi Lee
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ismail Sayin
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, Illinois, USA
| | - Stefan G. Tullius
- Division of Transplant Surgery & Transplant Surgery Research Laboratory, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anita S. Chong
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, Illinois, USA
| | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapies, University of Minnesota, Minneapolis, Minnesota, USA
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Yang S, Duan L, Wang C, Zhang C, Hou S, Wang H, Song J, Zhang T, Li Z, Wang M, Tang J, Zheng Q, Wang H, Wang Q, Zhao W. Activation and induction of antigen-specific T follicular helper cells play a critical role in recombinant SARS-CoV-2 RBD vaccine-induced humoral responses. MOLECULAR BIOMEDICINE 2023; 4:34. [PMID: 37853288 PMCID: PMC10584785 DOI: 10.1186/s43556-023-00145-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023] Open
Abstract
The role of follicular T helper (Tfh) cells in humoral response has been considered essential in recent years. Understanding how Tfh cells control complex humoral immunity is critical to developing strategies to improve the efficacy of vaccines against SARS-CoV-2 and other emerging pathogens. However, the immunologic mechanism of Tfh cells in SARS-CoV-2 receptor binding domain (RBD) vaccine strategy is limited. In this study, we expressed and purified recombinant SARS-CoV-2 RBD protein in Drosophila S2 cells for the first time and explored the mechanism of Tfh cells induced by RBD vaccine in humoral immune response. We mapped the dynamic of Tfh cell in lymph node and spleen following RBD vaccination and revealed the relationship between Tfh cells and humoral immune response induced by SARS-CoV-2 RBD vaccine through correlation analysis, blocking of IL-21 signaling pathway, and co-culture of Tfh with memory B cells. Recombinant RBD protein elicited a predominant Tfh1 and Tfh1-17 subset response and strong GC responses in spleen and lymph nodes, especially to enhanced vaccination. IL-21 secreted by Tfh cells affected the development and differentiation of B cells and played a key role in the humoral immune response. These observations will help us further understand the mechanism of protective immune response induced by COVID-19 vaccine and has guiding significance for the development of vaccines against newly emerging mutants.
Collapse
Affiliation(s)
- Songhao Yang
- School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
- Key Laboratory of Hydatid Disease, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
- Center of Scientific Technology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
| | - Liangwei Duan
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Chan Wang
- School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
- Key Laboratory of Hydatid Disease, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
| | - Cuiying Zhang
- School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
- Key Laboratory of Hydatid Disease, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
| | - Siyu Hou
- School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
- Key Laboratory of Hydatid Disease, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
| | - Hao Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Jiahui Song
- Key Laboratory of Hydatid Disease, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
- Center of Scientific Technology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
| | - Tingting Zhang
- School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
- Key Laboratory of Hydatid Disease, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
| | - Zihua Li
- School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
- Key Laboratory of Hydatid Disease, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
| | - Mingxia Wang
- School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
- Key Laboratory of Hydatid Disease, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
| | - Jing Tang
- School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
- Key Laboratory of Hydatid Disease, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China
| | - Qianqian Zheng
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Qi Wang
- School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China.
| | - Wei Zhao
- School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China.
- Key Laboratory of Hydatid Disease, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China.
- Center of Scientific Technology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, People's Republic of China.
| |
Collapse
|
19
|
Francis ME, Jansen EB, Yourkowski A, Selim A, Swan CL, MacPhee BK, Thivierge B, Buchanan R, Lavender KJ, Darbellay J, Rogers MB, Lew J, Gerdts V, Falzarano D, Skowronski DM, Sjaarda C, Kelvin AA. Previous infection with seasonal coronaviruses does not protect male Syrian hamsters from challenge with SARS-CoV-2. Nat Commun 2023; 14:5990. [PMID: 37752151 PMCID: PMC10522707 DOI: 10.1038/s41467-023-41761-1] [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: 02/22/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023] Open
Abstract
SARS-CoV-2 variants and seasonal coronaviruses continue to cause disease and coronaviruses in the animal reservoir pose a constant spillover threat. Importantly, understanding of how previous infection may influence future exposures, especially in the context of seasonal coronaviruses and SARS-CoV-2 variants, is still limited. Here we adopted a step-wise experimental approach to examine the primary immune response and subsequent immune recall toward antigenically distinct coronaviruses using male Syrian hamsters. Hamsters were initially inoculated with seasonal coronaviruses (HCoV-NL63, HCoV-229E, or HCoV-OC43), or SARS-CoV-2 pango B lineage virus, then challenged with SARS-CoV-2 pango B lineage virus, or SARS-CoV-2 variants Beta or Omicron. Although infection with seasonal coronaviruses offered little protection against SARS-CoV-2 challenge, HCoV-NL63-infected animals had an increase of the previously elicited HCoV-NL63-specific neutralizing antibodies during challenge with SARS-CoV-2. On the other hand, primary infection with HCoV-OC43 induced distinct T cell gene signatures. Gene expression profiling indicated interferon responses and germinal center reactions to be induced during more similar primary infection-challenge combinations while signatures of increased inflammation as well as suppression of the antiviral response were observed following antigenically distant viral challenges. This work characterizes and analyzes seasonal coronaviruses effect on SARS-CoV-2 secondary infection and the findings are important for pan-coronavirus vaccine design.
Collapse
Affiliation(s)
- Magen E Francis
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ethan B Jansen
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Anthony Yourkowski
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Alaa Selim
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Cynthia L Swan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Brian K MacPhee
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Brittany Thivierge
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Rachelle Buchanan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kerry J Lavender
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Joseph Darbellay
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Matthew B Rogers
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jocelyne Lew
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Volker Gerdts
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Darryl Falzarano
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada
| | - Danuta M Skowronski
- BC Centre for Disease Control, Immunization Programs and Vaccine Preventable Diseases Service, Vancouver, BC, Canada
- University of British Columbia, School of Population and Public Health, Vancouver, BC, Canada
| | - Calvin Sjaarda
- Department of Psychiatry, Queen's University, Kingston, ON, Canada
- Queen's Genomics Lab at Ongwanada (Q-GLO), Ongwanada Resource Centre, Kingston, ON, Canada
| | - Alyson A Kelvin
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK, Canada.
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
20
|
Schips M, Mitra T, Bandyopadhyay A, Meyer-Hermann M. Suppressive might of a few: T follicular regulatory cells impede auto-reactivity despite being outnumbered in the germinal centres. Front Immunol 2023; 14:1253704. [PMID: 37818361 PMCID: PMC10561256 DOI: 10.3389/fimmu.2023.1253704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/29/2023] [Indexed: 10/12/2023] Open
Abstract
The selection of high-affinity B cells and the production of high-affinity antibodies are mediated by T follicular helper cells (Tfhs) within germinal centres (GCs). Therein, somatic hypermutation and selection enhance B cell affinity but risk the emergence of self-reactive B cell clones. Despite being outnumbered compared to their helper counterpart, the ablation of T follicular regulatory cells (Tfrs) results in enhanced dissemination of self-reactive antibody-secreting cells (ASCs). The specific mechanisms by which Tfrs exert their regulatory action on self-reactive B cells are largely unknown. We developed computer simulations to investigate how Tfrs regulate either selection or differentiation of B cells to prevent auto-reactivity. We observed that Tfr-induced apoptosis of self-reactive B cells during the selection phase impedes self-reactivity with physiological Tfr numbers, especially when Tfrs can access centrocyte-enriched GC areas. While this aided in selecting non-self-reactive B cells by restraining competition, higher Tfr numbers distracted non-self-reactive B cells from receiving survival signals from Tfhs. Thus, the location and number of Tfrs must be regulated to circumvent such Tfr distraction and avoid disrupting GC evolution. In contrast, when Tfrs regulate differentiation of selected centrocytes by promoting recycling to the dark zone phenotype of self-reactive GC resident pre-plasma cells (GCPCs), higher Tfr numbers were required to impede the circulation of self-reactive ASCs (s-ASCs). On the other hand, Tfr-engagement with GCPCs and subsequent apoptosis of s-ASCs can control self-reactivity with low Tfr numbers, but does not confer selection advantage to non-self-reactive B cells. The simulations predict that to restrict auto-reactivity, natural redemption of self-reactive B cells is insufficient and that Tfrs should increase the mutation probability of self-reactive B cells.
Collapse
Affiliation(s)
- Marta Schips
- Department of Systems Immunology, Helmholtz Center for Infection Research, Helmholtz Association of German Research Centers (HZI), Braunschweig, Germany
| | - Tanmay Mitra
- Department of Systems Immunology, Helmholtz Center for Infection Research, Helmholtz Association of German Research Centers (HZI), Braunschweig, Germany
| | - Arnab Bandyopadhyay
- Department of Systems Immunology, Helmholtz Center for Infection Research, Helmholtz Association of German Research Centers (HZI), Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology, Helmholtz Center for Infection Research, Helmholtz Association of German Research Centers (HZI), Braunschweig, Germany
- Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universitat Braunschweig, Braunschweig, Germany
| |
Collapse
|
21
|
Yang J, Li B, Yang D, Wu J, Yang A, Wang W, Lin F, Wan X, Li Y, Chen Z, Lv S, Pang D, Liao W, Meng S, Lu J, Guo J, Wang Z, Shen S. The immunogenicity of Alum+CpG adjuvant SARS-CoV-2 inactivated vaccine in mice. Vaccine 2023; 41:6064-6071. [PMID: 37640568 DOI: 10.1016/j.vaccine.2023.08.061] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023]
Abstract
The ongoing evolution and emergence of SARS-CoV-2 variants have raised concerns regarding the efficacy of existing vaccines and therapeutic agents. This study aimed to investigate the immunogenicity of an aluminum hydroxide (Alum) and CpG adjuvanted inactivated vaccine (IAV) candidate against SARS-CoV-2 in mice. A comparison was made between the immune response of mice vaccinated with the Alum+CpG adjuvant IAV and those vaccinated with the Alum adjuvant IAV. Mice immunized with Alum+CpG adjuvant IAV demonstrated high antibody titers and a durable humoral immune response, as well as a Th1-type cellular immune response. Notably, compared to Alum alone vaccine, the Alum+CpG adjuvant IAV induced significantly higher proportions of GC B cells in the splenocytes of immunized mice. Importantly, the changes in inflammatory cytokine levels in the sera of mice vaccinated with the Alum+CpG adjuvant IAV followed a similar trend to that of the Alum adjuvant IAV, which had been proven safe in clinical trials. Overall, our results demonstrate that Alum+CpG adjuvant has the potential to serve as a novel adjuvant, thereby providing valuable insights into the development of vaccine formulations.
Collapse
Affiliation(s)
- Jie Yang
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Boran Li
- Hubei Province Medical Products Administration Center for Drug Evaluation, No. 19 Gongzheng Road, Wuchang District, Wuhan 430071, China
| | - Dongsheng Yang
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Jie Wu
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Anna Yang
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Wenhui Wang
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Fengjie Lin
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Xin Wan
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - YuWei Li
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Zhuo Chen
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Shiyun Lv
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Deqin Pang
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Wenbo Liao
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Shengli Meng
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Jia Lu
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Jing Guo
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Zejun Wang
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China
| | - Shuo Shen
- Wuhan Institute of Biological Products Co. Ltd., No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China; National Engineering Technology Research Center of Combined Vaccines, No. 1 Huangjin Industrial Park Road, Jiangxia District, Wuhan 430200, China.
| |
Collapse
|
22
|
Linterman MA. Age-dependent changes in T follicular helper cells shape the humoral immune response to vaccination. Semin Immunol 2023; 69:101801. [PMID: 37379670 DOI: 10.1016/j.smim.2023.101801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
Vaccination is an excellent strategy to limit the morbidity and mortality associated with infectious disease. Vaccination creates protective, long-lived antibody-mediated immunity by inducing the germinal centre response, an intricate immune reaction that produces memory B cells and long-lived antibody-secreting plasma cells that provide protection against (re)infection. The magnitude and quality of the germinal centre response declines with age, contributing to poor vaccine-induced immunity in older individuals. T follicular helper cells are essential for the formation and function of the germinal centre response. This review will discuss how age-dependent changes in T follicular helper cells influence the germinal centre response, and the evidence that age-dependent changes need not be a barrier to successful vaccination in the later years of life.
Collapse
Affiliation(s)
- Michelle A Linterman
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom.
| |
Collapse
|
23
|
Sircy LM, Ramstead AG, Joshi H, Baessler A, Mena I, García-Sastre A, Williams MA, Scott Hale J. Generation of antigen-specific memory CD4 T cells by heterologous immunization enhances the magnitude of the germinal center response upon influenza infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.29.555253. [PMID: 37693425 PMCID: PMC10491174 DOI: 10.1101/2023.08.29.555253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Current influenza vaccine strategies have yet to overcome significant obstacles, including rapid antigenic drift of seasonal influenza viruses, in generating efficacious long-term humoral immunity. Due to the necessity of germinal center formation in generating long-lived high affinity antibodies, the germinal center has increasingly become a target for the development of novel or improvement of less-efficacious vaccines. However, there remains a major gap in current influenza research to effectively target T follicular helper cells during vaccination to alter the germinal center reaction. In this study, we used a heterologous infection or immunization priming strategy to seed an antigen-specific memory CD4+ T cell pool prior to influenza infection in mice to evaluate the effect of recalled memory T follicular helper cells in increased help to influenza-specific primary B cells and enhanced generation of neutralizing antibodies. We found that heterologous priming with intranasal infection with acute lymphocytic choriomeningitis virus (LCMV) or intramuscular immunization with adjuvanted recombinant LCMV glycoprotein induced increased antigen-specific effector CD4+ T and B cellular responses following infection with a recombinant influenza strain that expresses LCMV glycoprotein. Heterologously primed mice had increased expansion of secondary Th1 and Tfh cell subsets, including increased CD4+ TRM cells in the lung. However, the early enhancement of the germinal center cellular response following influenza infection did not impact influenza-specific antibody generation or B cell repertoires compared to primary influenza infection. Overall, our study suggests that while heterologous infection/immunization priming of CD4+ T cells is able to enhance the early germinal center reaction, further studies to understand how to target the germinal center and CD4+ T cells specifically to increase long-lived antiviral humoral immunity are needed.
Collapse
Affiliation(s)
- Linda M. Sircy
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Andrew G. Ramstead
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Hemant Joshi
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Andrew Baessler
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Matthew A. Williams
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - J. Scott Hale
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
24
|
Roe K. Treatment alternatives for multidrug-resistant fungal pathogens. Drug Discov Today 2023; 28:103596. [PMID: 37086779 DOI: 10.1016/j.drudis.2023.103596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 04/24/2023]
Abstract
Several fungal pathogens are becoming resistant to conventional fungal infection treatments, and some fungal pathogens have become multidrug resistant. Alternative treatments include fungal vaccines, natural or synthetic monoclonal antibody (mAb) injections, or potentially natural or synthetic mAbs produced in vivo by packaged mRNA. Specifically synthesized proteins can mask distinctive pathogenic fungal surface proteins and target pathogenic fungal proteins to stop fungal infections. Treatments could use direct injections or injections of packaged mRNA with instructions for patient synthesis of either the natural or synthetic mAbs. These alternative treatments offer potentially significant advantages compared with existing treatments for fungal pathogens. Teaser: New fungal pathogen treatment approaches can use natural or synthetic monoclonal antibodies to activate immune cells and treat specific fungal infections that are now multidrug resistant to conventional antifungal drugs.
Collapse
|
25
|
Tavukcuoglu E, Yanik H, Parveen M, Uluturk S, Durusu-Tanriover M, Inkaya AC, Akova M, Unal S, Esendagli G. Human memory T cell dynamics after aluminum-adjuvanted inactivated whole-virion SARS-CoV-2 vaccination. Sci Rep 2023; 13:4610. [PMID: 36944716 PMCID: PMC10028771 DOI: 10.1038/s41598-023-31347-8] [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: 11/22/2022] [Accepted: 03/10/2023] [Indexed: 03/23/2023] Open
Abstract
This study evaluates the functional capacity of CD4+ and CD8+ terminally-differentiated effector (TEMRA), central memory (TCM), and effector memory (TEM) cells obtained from the volunteers vaccinated with an aluminum-adjuvanted inactivated whole-virion SARS-CoV-2 vaccine (CoronaVac). The volunteers were followed for T cell immune responses following the termination of a randomized phase III clinical trial. Seven days and four months after the second dose of the vaccine, the memory T cell subsets were collected and stimulated by autologous monocyte-derived dendritic cells (mDCs) loaded with SARS-CoV-2 spike glycoprotein S1. Compared to the placebo group, memory T cells from the vaccinated individuals significantly proliferated in response to S1-loaded mDCs. CD4+ and CD8+ memory T cell proliferation was detected in 86% and 78% of the vaccinated individuals, respectively. More than 73% (after a short-term) and 62% (after an intermediate-term) of the vaccinated individuals harbored TCM and/or TEM cells that responded to S1-loaded mDCs by secreting IFN-γ. The expression of CD25, CD38, 4-1BB, PD-1, and CD107a indicated a modulation in the memory T cell subsets. Especially on day 120, PD-1 was upregulated on CD4+ TEMRA and TCM, and on CD8+ TEM and TCM cells; accordingly, proliferation and IFN-γ secretion capacities tended to decline after 4 months. In conclusion, the combination of inactivated whole-virion particles with aluminum adjuvants possesses capacities to induce functional T cell responses.
Collapse
Affiliation(s)
- Ece Tavukcuoglu
- Department of Basic Oncology, Hacettepe University Cancer Institute, 06100, Sihhiye, Ankara, Turkey
| | - Hamdullah Yanik
- Department of Basic Oncology, Hacettepe University Cancer Institute, 06100, Sihhiye, Ankara, Turkey
| | - Mubaida Parveen
- Department of Basic Oncology, Hacettepe University Cancer Institute, 06100, Sihhiye, Ankara, Turkey
| | - Sila Uluturk
- Department of Basic Oncology, Hacettepe University Cancer Institute, 06100, Sihhiye, Ankara, Turkey
| | - Mine Durusu-Tanriover
- Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ahmet Cagkan Inkaya
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Murat Akova
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Serhat Unal
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Gunes Esendagli
- Department of Basic Oncology, Hacettepe University Cancer Institute, 06100, Sihhiye, Ankara, Turkey.
| |
Collapse
|
26
|
Garg AK, Mitra T, Schips M, Bandyopadhyay A, Meyer-Hermann M. Amount of antigen, T follicular helper cells and affinity of founder cells shape the diversity of germinal center B cells: A computational study. Front Immunol 2023; 14:1080853. [PMID: 36993964 PMCID: PMC10042134 DOI: 10.3389/fimmu.2023.1080853] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/16/2023] [Indexed: 03/14/2023] Open
Abstract
A variety of B cell clones seed the germinal centers, where a selection stringency expands the fitter clones to generate higher affinity antibodies. However, recent experiments suggest that germinal centers often retain a diverse set of B cell clones with a range of affinities and concurrently carry out affinity maturation. Amid a tendency to flourish germinal centers with fitter clones, how several B cell clones with differing affinities can be concurrently selected remains poorly understood. Such a permissive selection may allow non-immunodominant clones, which are often rare and of low-affinity, to somatically hypermutate and result in a broad and diverse B cell response. How the constituent elements of germinal centers, their quantity and kinetics may modulate diversity of B cells, has not been addressed well. By implementing a state-of-the-art agent-based model of germinal center, here, we study how these factors impact temporal evolution of B cell clonal diversity and its underlying balance with affinity maturation. While we find that the extent of selection stringency dictates clonal dominance, limited antigen availability on follicular dendritic cells is shown to expedite the loss of diversity of B cells as germinal centers mature. Intriguingly, the emergence of a diverse set of germinal center B cells depends on high affinity founder cells. Our analysis also reveals a substantial number of T follicular helper cells to be essential in balancing affinity maturation with clonal diversity, as a low number of T follicular helper cells impedes affinity maturation and also contracts the scope for a diverse B cell response. Our results have implications for eliciting antibody responses to non-immunodominant specificities of the pathogens by controlling the regulators of the germinal center reaction, thereby pivoting a way for vaccine development to generate broadly protective antibodies.
Collapse
Affiliation(s)
- Amar K. Garg
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Tanmay Mitra
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- *Correspondence: Tanmay Mitra, ; Michael Meyer-Hermann,
| | - Marta Schips
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Arnab Bandyopadhyay
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
- *Correspondence: Tanmay Mitra, ; Michael Meyer-Hermann,
| |
Collapse
|
27
|
He C, Chen L, Yang J, Chen Z, Lei H, Hong W, Song X, Yang L, Li J, Wang W, Shen G, Lu G, Wei X. Trimeric protein vaccine based on Beta variant elicits robust immune response against BA.4/5-included SARS-CoV-2 Omicron variants. MOLECULAR BIOMEDICINE 2023; 4:9. [PMID: 36894743 PMCID: PMC9998262 DOI: 10.1186/s43556-023-00121-7] [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: 10/08/2022] [Accepted: 02/07/2023] [Indexed: 03/11/2023] Open
Abstract
The current Coronavirus Disease 2019 (COVID-19) pandemic, induced by newly emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variants, posed great threats to global public health security. There is an urgent need to design effective next‑generation vaccines against Omicron lineages. Here, we investigated the immunogenic capacity of the vaccine candidate based on the receptor binding domain (RBD). An RBDβ-HR self-assembled trimer vaccine including RBD of Beta variant (containing K417, E484 and N501) and heptad repeat (HR) subunits was developed using an insect cell expression platform. Sera obtained from immunized mice effectively blocked RBD-human angiotensin-converting enzyme 2 (hACE2) binding for different viral variants, showing robust inhibitory activity. In addition, RBDβ-HR/trimer vaccine durably exhibited high titers of specific binding antibodies and high levels of cross-protective neutralizing antibodies against newly emerging Omicron lineages, as well as other major variants including Alpha, Beta, and Delta. Consistently, the vaccine also promoted a broad and potent cellular immune response involving the participation of T follicular helper (Tfh) cells, germinal center (GC) B cells, activated T cells, effector memory T cells, and central memory T cells, which are critical facets of protective immunity. These results demonstrated that RBDβ-HR/trimer vaccine candidates provided an attractive next-generation vaccine strategy against Omicron variants in the global effort to halt the spread of SARS-CoV-2.
Collapse
Affiliation(s)
- Cai He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zimin Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Lei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiangrong Song
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiong Li
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guobo Shen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guangwen Lu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
28
|
Ke F, Benet ZL, Maz MP, Liu J, Dent AL, Kahlenberg JM, Grigorova IL. Germinal center B cells that acquire nuclear proteins are specifically suppressed by follicular regulatory T cells. eLife 2023; 12:e83908. [PMID: 36862132 PMCID: PMC9981149 DOI: 10.7554/elife.83908] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/10/2023] [Indexed: 03/03/2023] Open
Abstract
Follicular regulatory T cells (Tfr) restrict development of autoantibodies and autoimmunity while supporting high-affinity foreign antigen-specific humoral response. However, whether Tfr can directly repress germinal center (GC) B cells that acquire autoantigens is unclear. Moreover, TCR specificity of Tfr to self-antigens is not known. Our study suggests that nuclear proteins contain antigens specific to Tfr. Targeting of these proteins to antigen-specific B cells in mice triggers rapid accumulation of Tfr with immunosuppressive characteristics. Tfr then exert negative regulation of GC B cells with predominant inhibition of the nuclear protein-acquiring GC B cells, suggesting an important role of direct cognate Tfr-GC B cells interactions for the control of effector B cell response.
Collapse
Affiliation(s)
- Fang Ke
- Department of Microbiology and Immunology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Zachary L Benet
- Department of Microbiology and Immunology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Mitra P Maz
- Department of Internal Medicine, Division of Rheumatology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Jianhua Liu
- Department of Internal Medicine, Division of Rheumatology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of MedicineIndianapolisUnited States
| | - Joanne Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Irina L Grigorova
- Department of Microbiology and Immunology, University of Michigan–Ann ArborAnn ArborUnited States
| |
Collapse
|
29
|
Zhang H, Cavazzoni CB, Hanson BL, Bechu ED, Podestà MA, Azzi J, Blazar BR, Chong AS, Kreisel D, Alessandrini A, Sage PT. Transcriptionally Distinct B Cells Infiltrate Allografts After Kidney Transplantation. Transplantation 2023; 107:e47-e57. [PMID: 36398326 PMCID: PMC9877106 DOI: 10.1097/tp.0000000000004398] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Following allogeneic kidney transplantation, a substantial proportion of graft loss is attributed to the formation of donor-specific antibodies and antibody-mediated rejection. B cells infiltrate kidney grafts during antibody-mediated rejection; however, the origins, repertoires, and functions of these intrarenal B cells remain elusive. METHODS Here, we use murine allogeneic kidney transplant models to study the origins, transcriptional programming and B cell receptor repertoire of intragraft B cells, and in vitro stimulation assays to evaluate the ability of intragraft B cells to promote CD4+ T cell expansion. RESULTS B cells infiltrate kidney grafts in settings of allogeneic, but not syngeneic, transplantation. Intragraft B cells have characteristics of activation but are transcriptionally distinct from germinal center B cells and resemble innate-like B cells. B cell receptor sequencing demonstrates that the majority of intragraft B cells do not originate from lymph node germinal center B cells and are largely germline. Class-switched intragraft B cells are rare but can be donor-specific and produce IgG capable of binding to the kidney allograft. Lastly, intrarenal B cells are capable of stimulating naive T cells but have an altered ability to promote T follicular helper cell expansion. CONCLUSIONS Together, these data demonstrate that intrarenal B cells during transplant rejection are transcriptionally distinct from lymph node B cells.
Collapse
Affiliation(s)
- Hengcheng Zhang
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Cecilia B. Cavazzoni
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Benjamin L. Hanson
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Elsa D. Bechu
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Manuel A. Podestà
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
- Renal Division, Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| | - Jamil Azzi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, 55455
| | - Anita S. Chong
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, IL, USA
| | - Daniel Kreisel
- Departments of Surgery, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Alessandro Alessandrini
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| |
Collapse
|
30
|
Roe K. Accelerated T-cell exhaustion: its potential role in autoimmune disease and hyperinflammatory disease pathogenesis. Hum Cell 2023; 36:866-869. [PMID: 36478089 PMCID: PMC9735067 DOI: 10.1007/s13577-022-00839-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
|
31
|
Hyperinflammatory disease and vasculitis-associated autoimmune disease pathogenesis by novel virulent pathogens inducing lymphocyte exhaustion and/or suppression. Hum Cell 2023; 36:872-874. [PMID: 36692672 PMCID: PMC9872057 DOI: 10.1007/s13577-023-00862-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
|
32
|
Roe K. Accelerated T-cell exhaustion: its pathogenesis and potentially severe outcomes. Hum Cell 2023; 36:488-490. [PMID: 36279031 PMCID: PMC9589834 DOI: 10.1007/s13577-022-00814-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 02/02/2023]
|
33
|
Roe K. Concurrent infections of cells by two pathogens can enable a reactivation of the first pathogen and the second pathogen's accelerated T-cell exhaustion. Heliyon 2022; 8:e11371. [PMCID: PMC9718926 DOI: 10.1016/j.heliyon.2022.e11371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/20/2022] [Accepted: 10/26/2022] [Indexed: 12/04/2022] Open
Abstract
When multiple intracellular pathogens, such as viruses, bacteria, fungi and protozoan parasites, infect the same host cell, they can help each other. A pathogen can substantially help another pathogen by disabling cellular immune defenses, using non-coding ribonucleic acids and/or pathogen proteins that target interferon-stimulated genes and other genes that express immune defense proteins. This can enable reactivation of a latent first pathogen and accelerate T-cell exhaustion and/or T-cell suppression regarding a second pathogen. In a worst-case scenario, accelerated T-cell exhaustion and/or T-cell suppression regarding the second pathogen can impair T-cell functionality and allow a first-time, immunologically novel second pathogen infection to escape all adaptive immune system defenses, including antibodies. The interactions of herpesviruses with concurrent intracellular pathogens in epithelial cells and B-cells, the interactions of the human immunodeficiency virus with Mycobacterium tuberculosis in macrophages and the interactions of Toxoplasma gondii with other pathogens in almost any type of animal cell are considered. The reactivation of latent pathogens and the acceleration of T-cell exhaustion for the second pathogen can explain several puzzling aspects of viral epidemics, such as COVID-19 and their unusual comorbidity mortality rates and post-infection symptoms.
Collapse
|
34
|
Zhu Q, Xu Y, Wang T, Xie F. Innate and adaptive immune response in SARS-CoV-2 infection-Current perspectives. Front Immunol 2022; 13:1053437. [PMID: 36505489 PMCID: PMC9727711 DOI: 10.3389/fimmu.2022.1053437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been a global pandemic, caused by a novel coronavirus strain with strong infectivity, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). With the in-depth research, the close relationship between COVID-19 and immune system has been dug out. During the infection, macrophages, dendritic cells, natural killer cells, CD8+ T cells, Th1, Th17, Tfh cells and effector B cells are all involved in the anti-SARS-CoV-2 responses, however, the dysfunctional immune responses will ultimately lead to the excessive inflammation, acute lung injury, even other organ failure. Thus, a detailed understanding of pertinent immune response during COVID-19 will provide insights in predicting disease outcomes and developing appropriate therapeutic approaches. In this review, we mainly clarify the role of immune cells in COVID-19 and the target-vaccine development and treatment.
Collapse
Affiliation(s)
- Qiugang Zhu
- Department of Laboratory Medicine, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Yan Xu
- Department of Respiratory Medicine, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Ting Wang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Feiting Xie
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,*Correspondence: Feiting Xie,
| |
Collapse
|
35
|
Costa Silva RCM, Bandeira-Melo C, Paula Neto HA, Vale AM, Travassos LH. COVID-19 diverse outcomes: Aggravated reinfection, type I interferons and antibodies. Med Hypotheses 2022; 167:110943. [PMID: 36105250 PMCID: PMC9461281 DOI: 10.1016/j.mehy.2022.110943] [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: 02/17/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 12/04/2022]
Abstract
SARS-CoV-2 infection intrigued medicine with diverse outcomes ranging from asymptomatic to severe acute respiratory syndrome (SARS) and death. After more than two years of pandemic, reports of reinfection concern researchers and physicists. Here, we will discuss potential mechanisms that can explain reinfections, including the aggravated ones. The major topics of this hypothesis paper are the disbalance between interferon and antibodies responses, HLA heterogeneity among the affected population, and increased proportion of cytotoxic CD4+ T cells polarization in relation to T follicular cells (Tfh) subtypes. These features affect antibody levels and hamper the humoral immunity necessary to prevent or minimize the viral burden in the case of reinfections.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratório de Imunoreceptores e Sinalização, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christianne Bandeira-Melo
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heitor Afonso Paula Neto
- Laboratório de Alvos Moleculares, Faculdade de Farmácia, Departamento de Biotecnologia Farmacêutica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André Macedo Vale
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Holanda Travassos
- Laboratório de Imunoreceptores e Sinalização, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
36
|
Zhang H, Sage PT. Role of T follicular helper and T follicular regulatory cells in antibody-mediated rejection: new therapeutic targets? Curr Opin Organ Transplant 2022; 27:371-375. [PMID: 35959918 PMCID: PMC9474598 DOI: 10.1097/mot.0000000000001018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW Antibody-mediated rejection (AbMR) after solid organ transplantation is tightly controlled by multiple cells of the immune system. Tfh and Tfr cells are essential controllers of antibody responses making them putative targets for therapeutics. However, the mechanisms of how Tfh and Tfr cells regulate B cell and antibody responses are not completely understood. Here, we summarize recent studies elucidating the functions of T follicular helper (Tfh) and T follicular regulatory (Tfr) cells as well as their possible roles in regulating AbMR in solid organ transplantation. RECENT FINDINGS New tools have been developed to study the roles of Tfh and Tfr cells in specific disease states, including AbMR after solid organ transplantation. These tools suggest complex roles for Tfh and Tfr cells in controlling antibody responses. Nevertheless, studies in solid organ transplant rejection suggest that Tfh and Tfr cells may be high value targets for therapeutics. However, specific strategies to target these cells are still being investigated. SUMMARY AbMR is still a substantial clinical problem that restricts long-term survival after solid organ transplantation. Growing evidence has demonstrated a pivotal role for Tfh and Tfr cells in controlling AbMR. In addition to providing an early indication of rejection as a biomarker, targeting Tfh and Tfr cells as a therapeutic strategy offers new hope for alleviating AbMR.
Collapse
Affiliation(s)
- Hengcheng Zhang
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
37
|
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
|
38
|
Duhen R, Beymer M, Jensen SM, Abbina S, Abraham S, Jain N, Thomas A, Geall AJ, Hu HM, Fox BA, Weinberg AD. OX40 agonist stimulation increases and sustains humoral and cell-mediated responses to SARS-CoV-2 protein and saRNA vaccines. Front Immunol 2022; 13:896310. [PMID: 36238275 PMCID: PMC9551348 DOI: 10.3389/fimmu.2022.896310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 09/08/2022] [Indexed: 12/01/2022] Open
Abstract
To prevent SARS-CoV-2 infections and generate long-lasting immunity, vaccines need to generate strong viral-specific B and T cell responses. Previous results from our lab and others have shown that immunizations in the presence of an OX40 agonist antibody lead to higher antibody titers and increased numbers of long-lived antigen-specific CD4 and CD8 T cells. Using a similar strategy, we explored the effect of OX40 co-stimulation in a prime and boost vaccination scheme using an adjuvanted SARS-CoV-2 spike protein vaccine in C57BL/6 mice. Our results show that OX40 engagement during vaccination significantly increases long-lived antibody responses to the spike protein. In addition, after immunization spike protein-specific proliferation was greatly increased for both CD4 and CD8 T cells, with enhanced, spike-specific secretion of IFN-γ and IL-2. Booster (3rd injection) immunizations combined with an OX40 agonist (7 months post-prime) further increased vaccine-specific antibody and T cell responses. Initial experiments assessing a self-amplifying mRNA (saRNA) vaccine encoding the spike protein antigen show a robust antigen-specific CD8 T cell response. The saRNA spike-specific CD8 T cells express high levels of GrzmB, IFN-γ and TNF-α which was not observed with protein immunization and this response was further increased by the OX40 agonist. Similar to protein immunizations the OX40 agonist also increased vaccine-specific CD4 T cell responses. In summary, this study compares and contrasts the effects and benefits of both protein and saRNA vaccination and the extent to which an OX40 agonist enhances and sustains the immune response against the SARS-CoV-2 spike protein.
Collapse
Affiliation(s)
- Rebekka Duhen
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
- *Correspondence: Rebekka Duhen,
| | - Michael Beymer
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
| | - Shawn M. Jensen
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
| | | | | | - Nikita Jain
- Precision NanoSystems Inc, Vancouver, BC, Canada
| | | | | | - Hong-Ming Hu
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
| | - Bernard A. Fox
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
| | - Andrew D. Weinberg
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
| |
Collapse
|
39
|
Young A. T cells in SARS-CoV-2 infection and vaccination. Ther Adv Vaccines Immunother 2022; 10:25151355221115011. [PMID: 36051003 PMCID: PMC9425900 DOI: 10.1177/25151355221115011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/04/2022] [Indexed: 12/24/2022] Open
Abstract
While antibodies garner the lion’s share of attention in SARS-CoV-2 immunity, cellular immunity (T cells) may be equally, if not more important, in controlling infection. Both CD8+ and CD4+ T cells are elicited earlier and are associated with milder disease, than antibodies, and T-cell activation appears to be necessary for control of infection. Variants of concern (VOCs) such as Omicron have escaped the neutralizing antibody responses after two mRNA vaccine doses, but T-cell immunity is largely intact. The breadth and patient-specific nature of the latter offers a formidable line of defense that can limit the severity of illness, and are likely to be responsible for most of the protection from natural infection or vaccination against VOCs which have evaded the antibody response. Comprehensive searches for T-cell epitopes, T-cell activation from infection and vaccination of specific patient groups, and elicitation of cellular immunity by various alternative vaccine modalities are here reviewed. Development of vaccines that specifically target T cells is called for, to meet the needs of patient groups for whom cellular immunity is weaker, such as the elderly and the immunosuppressed. While VOCs have not yet fully escaped T-cell immunity elicited by natural infection and vaccines, some early reports of partial escape suggest that future VOCs may achieve the dreaded result, dislodging a substantial proportion of cellular immunity, enough to cause a grave public health burden. A proactive, rather than reactive, solution which identifies and targets immutable sequences in SARS-CoV-2, not just those which are conserved, may be the only recourse humankind has to disarm these future VOCs before they disarm us.
Collapse
Affiliation(s)
- Arthur Young
- InvVax, 2265 E. Foohill Blvd., Pasadena, CA 91107, USA
| |
Collapse
|
40
|
Wang Y, Tian Q, Ye L. The Differentiation and Maintenance of SARS-CoV-2-Specific Follicular Helper T Cells. Front Cell Infect Microbiol 2022; 12:953022. [PMID: 35909969 PMCID: PMC9329515 DOI: 10.3389/fcimb.2022.953022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/20/2022] [Indexed: 12/24/2022] Open
Abstract
Upon acute viral infection, virus-specific CD4+ T cells differentiate into either TH1 cells or follicular helper T (TFH) cells. The molecular pathways governing such bimodal cell fate commitment remain elusive. Additionally, effector virus-specific TFH cells further differentiate into corresponding memory population, which confer long-term protection against re-infection of same viruses by providing immediate help to virus-specific memory B cells. Currently, the molecular mechanisms underlying the long-term maintenance of memory TFH cells are largely unknown. In this review, we discuss current understanding of early differentiation of virus-specific effector TFH cells and long-term maintenance of virus-specific memory TFH cells in mouse models of viral infection and patients of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection.
Collapse
Affiliation(s)
- Yifei Wang
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Qin Tian
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Institute of Immunology, The People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| | - Lilin Ye
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Institute of Immunology, The People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
- *Correspondence: Lilin Ye,
| |
Collapse
|
41
|
Akama-Garren EH, Carroll MC. T Cell Help in the Autoreactive Germinal Center. Scand J Immunol 2022; 95:e13192. [PMID: 35587582 DOI: 10.1111/sji.13192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/29/2022]
Abstract
The germinal center serves as a site of B cell selection and affinity maturation, critical processes for productive adaptive immunity. In autoimmune disease tolerance is broken in the germinal center reaction, leading to production of autoreactive B cells that may propagate disease. Follicular T cells are crucial regulators of this process, providing signals necessary for B cell survival in the germinal center. Here we review the emerging roles of follicular T cells in the autoreactive germinal center. Recent advances in immunological techniques have allowed study of the gene expression profiles and repertoire of follicular T cells at unprecedented resolution. These studies provide insight into the potential role follicular T cells play in preventing or facilitating germinal center loss of tolerance. Improved understanding of the mechanisms of T cell help in autoreactive germinal centers provides novel therapeutic targets for diseases of germinal center dysfunction.
Collapse
Affiliation(s)
- Elliot H Akama-Garren
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Ghosh S, Leavenworth JW. Current Advances in Follicular Regulatory T-Cell Biology. Crit Rev Immunol 2022; 42:35-47. [PMID: 37017287 PMCID: PMC11034780 DOI: 10.1615/critrevimmunol.2022045746] [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: 11/13/2022]
Abstract
Follicular regulatory T (TFR) cells are a population of CD4+ T-cells that concomitantly express markers for regulatory T-cells and follicular helper T (TFH) cells, and have been predominantly implicated in the regulation of humoral immunity via their suppressive functions. Rapid and robust progress has been made in the field of TFR cell research since the discovery of this subset over a decade ago. However, there is still a significant gap in our understanding of the mechanisms underlying the phenotypic and functional heterogeneity of TFR cells under various physiologic and pathologic settings. In this review article, we aim to highlight the most up-to-date concepts and investigations in both experimental animal models and human studies to provide a perspective on our understanding of TFR biology with particular emphasis on these cells in the context of disease settings.
Collapse
Affiliation(s)
- Sadashib Ghosh
- Department of Neurosurgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Jianmei W. Leavenworth
- Department of Neurosurgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233 USA
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
- The O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| |
Collapse
|