1
|
Gong Y, Xu R, Gao G, Li S, Liu Y. The role of fatty acid metabolism on B cells and B cell-related autoimmune diseases. Inflamm Res 2025; 74:75. [PMID: 40299047 DOI: 10.1007/s00011-025-02042-3] [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: 02/08/2025] [Revised: 04/08/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
Fatty acid metabolism plays a critical role in regulating immune cell function, including B cells, which are central to humoral immunity and the pathogenesis of autoimmune diseases. Emerging evidence suggests that fatty acid metabolism influences B cell development, activation, differentiation, and antibody production, thereby impacting B cell-related autoimmune diseases such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and multiple sclerosis (MS). In this review, we discuss the mechanisms by which fatty acid metabolism modulates B cell biology, including energy provision, membrane composition, and signaling pathways. We highlight how alterations in fatty acid synthesis, oxidation, and uptake affect B cell function and contribute to autoimmune pathogenesis. Additionally, we explore the therapeutic potential of targeting fatty acid metabolism in B cells to treat autoimmune diseases. Understanding the interplay between fatty acid metabolism and B cell immunity may provide novel insights into the development of precision therapies for B cell-mediated autoimmune disorders.
Collapse
Affiliation(s)
- Yanmei Gong
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Ruiqi Xu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Guohui Gao
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Simiao Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Ying Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China.
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China, China.
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, Shandong, China.
| |
Collapse
|
2
|
Kotagiri P, Rae WM, Bergamaschi L, Pombal D, Lee JY, Noor NM, Sojwal RS, Rubin SJS, Unger LW, Tolmeijer SH, Manferrari G, Bashford-Rogers RJM, Bingham DB, Stift A, Rogalla S, Gubatan J, Lee JC, Smith KGC, McKinney EF, Boyd SD, Lyons PA. Disease-specific B cell clones are shared between patients with Crohn's disease. Nat Commun 2025; 16:3689. [PMID: 40246842 PMCID: PMC12006383 DOI: 10.1038/s41467-025-58977-y] [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: 01/25/2023] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
B cells have important functions in gut homeostasis, and dysregulated B cell populations are frequently observed in patients with inflammatory bowel diseases, including both ulcerative colitis (UC) and Crohn's disease (CD). How these B cell perturbations contribute to disease remains largely unknown. Here, we perform deep sequencing of the B cell receptor (BCR) repertoire in four cohorts of patients with CD, together with healthy controls and patients with UC. We identify BCR clones that are shared between patients with CD but not found in healthy individuals nor in patients with UC, indicating CD-associated B cell immune responses. Shared clones are present in the inflamed gut mucosa, draining intestinal lymph nodes and blood, suggesting the presence of common CD-associated antigens that drive B cell responses in CD patients.
Collapse
Affiliation(s)
- Prasanti Kotagiri
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK.
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA.
| | - William M Rae
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Discovery Sciences, AstraZeneca, Cambridge Biomedical Campus, Cambridge, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Diana Pombal
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Ji-Yeun Lee
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Nurulamin M Noor
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Raoul S Sojwal
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - Samuel J S Rubin
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - Lukas W Unger
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Sofie H Tolmeijer
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Giulia Manferrari
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Rachael J M Bashford-Rogers
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Department of Biochemistry, South Parks Road, University of Oxford, Oxford, OX1 3QU, UK
| | - David B Bingham
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Anton Stift
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Stephan Rogalla
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - John Gubatan
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - James C Lee
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- The Francis Crick Institute and UCL Institute of Liver and Digestive Health, Division of Medicine, Royal Free Campus, London, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Eoin F McKinney
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Scott D Boyd
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK.
| |
Collapse
|
3
|
Pan C, Yu S, Li C, Li J, Sun P, Guo Y, Li T, Wang D, Wang K, Lyu Y, Liu X, Li X, Wu J, Zhu L, Wang H. Rapid and efficient immune response induced by a designed modular cholera toxin B subunit (CTB)-based self-assembling nanoparticle. Biomaterials 2025; 315:122946. [PMID: 39515192 DOI: 10.1016/j.biomaterials.2024.122946] [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: 08/07/2024] [Revised: 10/21/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Modular self-assembling nanoparticle vaccines, represent a cutting-edge approach in immunology with the potential to revolutionize vaccine design and efficacy. Although many innovative efficient modular self-assembling nanoparticles have been designed for vaccination, the immune activation characteristics underlying such strong protection remain poorly understood, limiting the further expansion of such nanocarrier. Here, we prepared a novel modular nanovaccine, which self-assembled via a pentamer cholera toxin B subunit (CTB) domain and an unnatural trimer domain, presenting S. Paratyphi A O-polysaccharide antigen, and investigated its rapid immune activation mechanism. The nanovaccine efficiently targets draining lymph nodes and antigen-presenting cells, facilitating co-localization with Golgi and endoplasmic reticulum. In addition, dendritic cells, macrophages, B cells, and neutrophils potentially participate in antigen presentation, unveiling a dynamic change of the vaccines in lymph nodes. Single-cell RNA sequencing at early stage and iN vivo/iN vitro experiments reveal its potent humoral immune response capabilities and protection effects. This nanoparticle outperforms traditional CTB carriers in eliciting robust prophylactic effects in various infection models. This work not only provides a promising and efficient candidate vaccine, but also promotes the design and application of the new type of self-assembled nanoparticle, offering a safe and promising vaccination strategy for infection diseases.
Collapse
Affiliation(s)
- Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Shujuan Yu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Caixia Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Juntao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Peng Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China; Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Ting Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Dongshu Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Kangfeng Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yufei Lyu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiankai Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiang Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jun Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Hengliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071, China.
| |
Collapse
|
4
|
Lee YZ, Zhang YN, Newby ML, Ward G, Gomes KB, Auclair S, DesRoberts C, Allen JD, Ward AB, Stanfield RL, He L, Crispin M, Wilson IA, Zhu J. Rational design of next-generation filovirus vaccines with glycoprotein stabilization, nanoparticle display, and glycan modification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.02.641072. [PMID: 40060701 PMCID: PMC11888476 DOI: 10.1101/2025.03.02.641072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Filoviruses pose a significant threat to human health with frequent outbreaks and high mortality. Although two vector-based vaccines are available for Ebola virus, a broadly protective filovirus vaccine remains elusive. In this study, we evaluate a general strategy for stabilizing glycoprotein (GP) structures of Ebola, Sudan, and Bundibugyo ebolaviruses and Ravn marburgvirus. A 3.2 Å-resolution crystal structure provides atomic details for the redesigned Ebola virus GP, and cryo-electron microscopy reveals how a pan-ebolavirus neutralizing antibody targets a conserved site on the Sudan virus GP (3.13 Å-resolution), in addition to a low-resolution model of antibody-bound Ravn virus GP. A self-assembling protein nanoparticle (SApNP), I3-01v9, is redesigned at the N-terminus to allow the optimal surface display of filovirus GP trimers. Following detailed in vitro characterization, the lymph node dynamics of Sudan virus GP and GP-presenting SApNPs are investigated in a mouse model. Compared with soluble GP trimer, SApNPs show ~112 times longer retention in lymph node follicles, up-to-28 times greater presentation on follicular dendritic cell dendrites, and up-to-3 times stronger germinal center reactions. Functional antibody responses induced by filovirus GP trimers and SApNPs bearing wildtype and modified glycans are assessed in mice. Our study provides a foundation for next-generation filovirus vaccine development.
Collapse
Affiliation(s)
- Yi-Zong Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Maddy L. Newby
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Garrett Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Sarah Auclair
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Connor DesRoberts
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Joel D. Allen
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Robyn L. Stanfield
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Max Crispin
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Uvax Bio, LLC, Newark, DE 19702, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
5
|
Lu B, Chaudhary O, Banoth B, Nadkarni J, Zong W, Mausser E, Danz H, Motwani M, Ruiz S, Zhang D, Nageshwaran G, Rokbi B, Warren W, DeRosa F, Chivukula S. Impact of Extended Dosing Intervals and Ipsilateral Versus Contralateral Boosting on mRNA Vaccine Immunogenicity in Mice. Vaccines (Basel) 2025; 13:263. [PMID: 40266125 PMCID: PMC11946721 DOI: 10.3390/vaccines13030263] [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: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Although mRNA vaccines have the potential to be developed and deployed rapidly to combat infectious diseases, the ideal method of administration and boosting schedule strategy for generating optimal immunogenicity is an area of active research. We compared the immune responses resulting from different schedules for prime-boost and boosting either ipsilaterally or contralaterally in relation to the initial vaccine dose. METHODS Influenza hemagglutinin (HA) was used as a model antigen for different vaccination regimens in mice using both mRNA lipid nanoparticles (mRNA-LNP) and AF03-adjuvanted recombinant protein (rHA-AF03) vaccines. RESULTS Increasing the prime-boost interval resulted in higher levels of serum anti-HA IgG and functional antibody hemagglutination inhibition (HAI) responses in mRNA-LNP-vaccinated animals, which correlated with an induction of germinal center (GC) B cells and follicular helper T (Tfh) cells in lymph nodes. In addition, longer prime-boost intervals resulted in higher levels of IL-2 and TNF-α producing CD4+ T cells two weeks after boosting. The number of Ig-secreting long-lived plasma cells increased with the length of prime-boost intervals. Contralateral boosting resulted in an increase in HAI titers and GC B cells compared to an ipsilateral boost. However, significantly higher numbers of GC B cells were induced in the draining lymph nodes following ipsilateral boosting than in the non-draining lymph nodes. CONCLUSIONS Overall, our data provides insights into the immune mechanisms of action of mRNA-LNP to develop the optimal vaccine regimen for mRNA vaccine platforms.
Collapse
Affiliation(s)
- Bin Lu
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Omkar Chaudhary
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Balaji Banoth
- Former Employee of Sanofi, 200 West St., Waltham, MA 02451, USA
| | - Janhavi Nadkarni
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Wei Zong
- Translational and Early Development Biostatistics, Sanofi, 200 West St., Waltham, MA 02451, USA (D.Z.)
| | - Emilie Mausser
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Hillary Danz
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Mona Motwani
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Sophie Ruiz
- mRNA Center of Excellence, Sanofi, 1541 Avenue Marcel Mérieux, 69280 MarcyL’Etoile, France;
| | - Donghui Zhang
- Translational and Early Development Biostatistics, Sanofi, 200 West St., Waltham, MA 02451, USA (D.Z.)
| | - Gopinath Nageshwaran
- Global Antigen Design, Sanofi, 200 West St., Waltham, MA 02451, USA; (G.N.); (W.W.)
| | - Bachra Rokbi
- Global Antigen Design, Sanofi, 1541 Avenue Marcel Mérieux, 69280 Marcy L’Etoile, France;
| | - William Warren
- Global Antigen Design, Sanofi, 200 West St., Waltham, MA 02451, USA; (G.N.); (W.W.)
| | - Frank DeRosa
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Sudha Chivukula
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| |
Collapse
|
6
|
Wan D, Zhao S, Zhang C, Xu F, Wang H, Tao S, Qiu Z, Jiang H, Li D, Wang F, Li D, Chen J, Wang Y, Yan Y, Zhao Y, Gao X, Jin B, Liu D, Zhang M, Feng J, Hou S, Wang M, Chen T, Lin M, Han J, Wen X, Jiang W, Liu L, Long Y, Zhao Y, Kira JI, Liu Z, Chai G, Hao J. Novel Meningoencephalomyelitis Associated With Vimentin IgG Autoantibodies. JAMA Neurol 2025; 82:247-257. [PMID: 39836414 PMCID: PMC11894498 DOI: 10.1001/jamaneurol.2024.4763] [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: 08/12/2024] [Accepted: 11/07/2024] [Indexed: 01/22/2025]
Abstract
Importance Autoantibodies targeting astrocytes, such as those against glial fibrillary acidic protein (GFAP) or aquaporin protein 4, are crucial diagnostic markers for autoimmune astrocytopathy among central nervous system (CNS) autoimmune disorders. However, diagnosis remains challenging for patients lacking specific autoantibodies. Objective To characterize a syndrome of unknown meningoencephalomyelitis associated with an astrocytic autoantibody. Design, Setting, and Participants This retrospective case series study included samples collected from April 2021 to May 2024 at a tertiary referral hospital among patients with uncharacterized CNS autoimmune disorders and similar clinical and radiological features. Single-cell RNA sequencing (scRNA-seq) was performed on cerebrospinal fluid (CSF) cells of 2 index patients to identify the putative target antigen of the clonally expanded B cells. A comprehensive screening for additional patients was conducted using blinded cell-based and tissue-based assay. Candidate patients were followed up for a median (range) duration of 23 (5-31) months. Exposures scRNA-seq, autoantibody characterization, and testing. Main Outcomes and Measures Detection of the autoantibody and characterization of the associated autoimmune meningoencephalomyelitis. Results Fourteen candidate patients (10 [71%] female; median [IQR] age, 33 [23-41] years) were identified. Initially, CSF from 2 female patients with unknown encephalomyelitis showed astrocytic reactivity on rat tissue but was negative for GFAP IgG. A total of 17 of 37 clonally expanded B cell clonotypes (46%) in their CSF expressed IgG autoantibodies targeting the astrocytic intermediate filament protein vimentin. Subsequent screening identified 12 additional patients. These 14 patients shared a unique clinical profile characterized by relapsing courses and symptoms prominently involving the cerebellum, brainstem, and corticospinal tract (CST). All patients also exhibited elevated CSF protein and cells, intrathecal immunoglobulin synthesis, and magnetic resonance imaging (MRI) showing bilateral lesions on CST. Notably, 8 of 12 patients (67%) who received first-line immunotherapy at their first episode responded well. At the last follow-up, 11 patients (79%) experienced significant disability (modified Rankin Scale ≥3). Conclusions and Relevance In this case series, autoantibodies targeting the astrocytic intermediate filament protein vimentin were identified in patients with previously undifferentiated meningoencephalomyelitis and common radiographic features.
Collapse
Affiliation(s)
- Dongshan Wan
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Shufang Zhao
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Chen Zhang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
- Department of Neurology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Fang Xu
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Huizi Wang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Shaoxin Tao
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Zhandong Qiu
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Hao Jiang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Dawei Li
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Fei Wang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Dong Li
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Jiahao Chen
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Yan Wang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Yao Yan
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Yan Zhao
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Xiaohan Gao
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Bingxue Jin
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Di Liu
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Mengyao Zhang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Jingjing Feng
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Shiyue Hou
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Mingyang Wang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Teng Chen
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Ming Lin
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Jinming Han
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Xinmei Wen
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Wei Jiang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Liang Liu
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Youming Long
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yinan Zhao
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Jun-Ichi Kira
- Translational Neuroscience Center, Graduate School of Medicine, International University of Health and Welfare, Okawa, Japan
| | - Zheng Liu
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Guoliang Chai
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Junwei Hao
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, China
| |
Collapse
|
7
|
Wu J, Zhou J, Li G, Sun X, Xiang C, Chen H, Jiang P. Metabolic determinants of germinal center B cell formation and responses. Nat Chem Biol 2025; 21:371-382. [PMID: 39060389 DOI: 10.1038/s41589-024-01690-6] [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: 07/24/2023] [Accepted: 07/03/2024] [Indexed: 07/28/2024]
Abstract
Germinal center (GC) B cells are crucial for the generation of GCs and long-lived humoral immunity. Here we report that one-carbon metabolism determines the formation and responses of GC B cells. Upon CD40 stimulation, GC B cells selectively upregulate methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) expression to generate purines and the antioxidant glutathione. MTHFD2 depletion reduces GC B cell frequency and antigen-specific antibody production. Moreover, supplementation with nucleotides and antioxidants suffices to promote GC B cell formation and function in vitro and in vivo through activation of the mammalian target of rapamycin complex 1 signaling pathway. Moreover, we found that antigen stimulation enhances YY1 binding to the Mthfd2 promoter and promotes MTHFD2 transcription. Interestingly, these findings can be generalized to the pentose phosphate pathway, which is another major source of reducing power and nucleotides. Therefore, these results suggest that an increased capacity for nucleotide synthesis and redox balance is required for GC B cell formation and responses, revealing a key aspect of GC B cell fate determination.
Collapse
Affiliation(s)
- Jun Wu
- State Key Laboratory of Molecular Oncology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Jiawen Zhou
- State Key Laboratory of Molecular Oncology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Gen Li
- State Key Laboratory of Molecular Oncology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xuan Sun
- State Key Laboratory of Molecular Oncology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Chen Xiang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Haiyan Chen
- State Key Laboratory of Molecular Oncology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Peng Jiang
- State Key Laboratory of Molecular Oncology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
8
|
Kumar S, Hazlett K, Bai G. Mucosal immunity elicited by a human-Fcγ receptor-I targeted intranasal vaccine platform enhances resistance against nasopharyngeal colonization of Streptococcus pneumoniae and induces broadly protective immunity against respiratory pathogens. Vaccine 2025; 48:126729. [PMID: 39823848 DOI: 10.1016/j.vaccine.2025.126729] [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: 11/14/2024] [Revised: 12/20/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025]
Abstract
The development of safe and effective mucosal vaccines are hampered by safety concerns associated with adjuvants or live attenuated microbes. We previously demonstrated that targeting antigens to the human-Fc-gamma-receptor-I (hFcγRI) eliminates the need for adjuvants, thereby mitigating safety concerns associated with the mucosal delivery of adjuvant formulated vaccines. Here we evaluated the role of the route of immunization in the mucosal immunity elicited by the hFcγRI-targeted vaccine approach. To enable Ag targeting, PspA from Streptococcus pneumoniae (Sp) was genetically fused with the hFcγRI-targeting antibody (α-hFcγRI) to generate PspA-FP. Intranasal (IN) immunization with the PspA-FP induced significantly higher IgA, IgG, and memory T cell response in the lung mucosa compared to that of the intramuscular (IM) route, while both routes exhibited similar increase in the systemic IgG response. The IN immunization elicited better resistance against nasal colonization (NC) of Sp compared to the IM immunization. Additionally, the resistance to NC with the IN administered PspA-FP was higher than the PspA-Alum formulation administered by the IM route. While the protection form lethal pulmonary Sp infection correlated with the systemic Ab response, the resistance from NC (of Sp) correlated with the mucosal immune response. Similar to the pneumococcal pneumoniae model, the hFcγRI-targeted vaccine (based on HA as Ag) was equally protective against pulmonary Influenza virus infection via both routes. However, the IN route promoted better protection compared to the IM route against a lethal pulmonary infection with Francisella tularensis (Ft). The enhanced protection against Ft correlated with the superior mucosal immune response elicited by the IN route compared to the IM route. These observations showed a differential requirement for mucosal delivery for protection depending on the type of pathogen. Moreover, this study revealed that the hFcγRI-targeted vaccine platform is broadly-effective as an adjuvant-free mucosal vaccine platform against respiratory pathogens.
Collapse
Affiliation(s)
- Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| | - Karsten Hazlett
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States
| | - Guangchun Bai
- Department of Immunology and Microbial Disease, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States
| |
Collapse
|
9
|
Claireaux M, Elias G, Kerster G, Kuijper LH, Duurland MC, Paul AGA, Burger JA, Poniman M, Olijhoek W, de Jong N, de Jongh R, Wynberg E, van Willigen HDG, Prins M, De Bree GJ, de Jong MD, Kuijpers TW, Eftimov F, van der Schoot CE, Rispens T, Garcia-Vallejo JJ, ten Brinke A, van Gils MJ, van Ham SM. Deep profiling of B cells responding to various pathogens uncovers compartments in IgG memory B cell and antibody-secreting lineages. SCIENCE ADVANCES 2025; 11:eado1331. [PMID: 39970201 PMCID: PMC11837990 DOI: 10.1126/sciadv.ado1331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025]
Abstract
Improving our understanding of B cell transition to memory B cells (MBCs) and antibody-secreting cells (ASCs) is crucial for clinical monitoring and vaccine strategies. To explore these dynamics, we compared prepandemic antigen responses (influenza hemagglutinin, respiratory syncytial virus fusion glycoprotein, and tetanus toxoid) with recently encountered severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antigen responses in convalescent COVID-19 patients using spectral flow cytometry. Our analysis revealed the CD43+CD71+IgG+ activated B cell subset, highly enriched for SARS-CoV-2 specificities, as a juncture for ASC and MBC differentiation, with CD86+ phenotypically similar to ASCs and CD86- to IgG+ MBCs. Moreover, subpopulations within IgG+ MBCs were further identified based on CD73 and CD24 expression. Activated MBCs (CD73-/CD24lo) were predominantly SARS-CoV-2-specific, while resting MBCs (CD73+/CD24hi) recognized prepandemic antigens. A CD95- subcluster within resting MBCs accounted for over 40% of prepandemic-specific cells, indicating long-lasting memory. These findings advance our understanding of IgG+ MBC and ASC development stages, shedding light on the decision-making process guiding their differentiation.
Collapse
Affiliation(s)
- Mathieu Claireaux
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - George Elias
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Gius Kerster
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Lisan H. Kuijper
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Mariël C. Duurland
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | | | - Judith A. Burger
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Meliawati Poniman
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Wouter Olijhoek
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Nina de Jong
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Rivka de Jongh
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Elke Wynberg
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, Netherlands
| | - Hugo D. G. van Willigen
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Maria Prins
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Godelieve J. De Bree
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Menno D. de Jong
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - Taco W. Kuijpers
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Filip Eftimov
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - C. Ellen van der Schoot
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Theo Rispens
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Juan J. Garcia-Vallejo
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Molecular Cell Biology & Immunology, Amsterdam University Medical Center (VUmc location), Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Anja ten Brinke
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
| | - S. Marieke van Ham
- Amsterdam Institute for Immunology and Infectious diseases, Amsterdam, Netherlands
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Amsterdam, Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
10
|
Domeier PP, Ziegler SF. Thymic stromal lymphopoietin signaling in B cells from progenitors to plasma cells. J Leukoc Biol 2025; 117:qiae216. [PMID: 39373526 PMCID: PMC11878999 DOI: 10.1093/jleuko/qiae216] [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: 06/15/2024] [Revised: 08/28/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024] Open
Abstract
Thymic stromal lymphopoietin is an established pleotropic alarmin cytokine that is generated at barrier tissues to induce type 2 immune responses, but its role in regulating the diversity of B cells is poorly understood. Here, we will highlight the key findings that underpin our limited understanding of the role thymic stromal lymphopoietin in modulating different stages of B cell development. We will also provide an overview of how thymic stromal lymphopoietin drives B cell-mediated immune disease and how novel thymic stromal lymphopoietin-blocking biologics could be used to modulate B cell responses. Thymic stromal lymphopoietin is critical for the regulation, diversity, and longevity of humoral immunity.
Collapse
Affiliation(s)
- Phillip P Domeier
- Center for Fundamental Immunology, Benaroya Research Institute, 1201 9th Avenue, Seattle, WA 98101, United States
| | - Steven F Ziegler
- Center for Fundamental Immunology, Benaroya Research Institute, 1201 9th Avenue, Seattle, WA 98101, United States
| |
Collapse
|
11
|
Lin G, Tang YL, Fu Z, Chen R, Liu Y, Liu Z, Kuang X, Sun J, Zhao J, Zhang Y. Enhancing protective immunity against SARS-CoV-2 with a self-amplifying RNA lipid nanoparticle vaccine. J Control Release 2025; 378:250-265. [PMID: 39645087 DOI: 10.1016/j.jconrel.2024.12.003] [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: 07/24/2024] [Revised: 11/12/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
RNA-based vaccines against SARS-CoV-2 have demonstrated promising protective immunity against the global COVID-19 epidemic. Enhancing the intensity and duration of mRNA antigen expression is anticipated to markedly boost antiviral immune responses. Self-amplifying RNA (saRNA) represents a next-generation platform for RNA-based vaccines, amplifying transcripts in situ to augment the expression of encoded immunogens. Here, we develop a saRNA nanovaccine, formulated with a mutated saRNA encoding the receptor-binding domain (RBD) of the SARS-CoV-2 spike glycoprotein, encapsulated within a lipid nanoparticle (LNP-saRNA-RBD). This LNP-saRNA vaccine platform enables efficient delivery of saRNA-RBD, inducing enhanced and prolonged expression of the RBD antigen. LNP-saRNA-RBD vaccination stimulated the generation of antigen-specific T cells, promoting their differentiation into a long-lived effector memory phenotype. Immunization with LNP-saRNA-RBD induced a germinal center response in draining lymph nodes, leading to the production of anti-RBD IgG antibodies with the ability to neutralize SARS-CoV-2 pseudovirus. Furthermore, prime-boost immunizations with LNP-saRNA-RBD conferred protection to mice against SARS-CoV-2 challenge by suppressing viral infection and replication, as well as pulmonary inflammatory responses and associated damage. Taken together, these findings provide strong support for advancing the development of LNP-saRNA-RBD as a safe and efficacious vaccine candidate against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Guibin Lin
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Yan-Lai Tang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhengqiang Fu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Runjun Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Yan Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Zhanyan Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Xueli Kuang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, China; Institute of Infectious Disease, Guangzhou Eighth People's Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Yuan Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
12
|
Al-Aubodah TA, Piccirillo CA, Trachtman H, Takano T. The autoimmune architecture of childhood idiopathic nephrotic syndrome. Kidney Int 2025; 107:271-279. [PMID: 39571906 DOI: 10.1016/j.kint.2024.10.027] [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: 07/30/2024] [Revised: 09/26/2024] [Accepted: 10/17/2024] [Indexed: 12/10/2024]
Abstract
Idiopathic nephrotic syndrome, the most common glomerular disorder in children, has long been considered an immune-mediated disease based on the efficacy of glucocorticoids at inducing remission. Nevertheless, the immune processes leading to podocytopathy have largely remained elusive. The success of B-cell depletion with rituximab, descriptions of B-cell dysregulation during active disease, and the most recent discovery of autoantibodies targeting the major podocyte antigen nephrin point to an autoimmune humoral etiology for idiopathic nephrotic syndrome. Investigations of the immune factors involved in idiopathic nephrotic syndrome pathogenesis have uncovered common features with other autoimmune disorders that will aid in prognostication and in guiding the expansion of our glucocorticoid-sparing therapeutic arsenal. In this review, we discuss the emerging autoimmune architecture of idiopathic nephrotic syndrome, with a specific focus on pediatric steroid-sensitive disease, including the podocyte-reactive B-cell response that causes anti-podocyte antibodies, the predisposing genetic factors that shape the podocyte-reactive immune landscape, and the immune triggers driving active disease.
Collapse
Affiliation(s)
- Tho-Alfakar Al-Aubodah
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada; Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada; Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada; Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Howard Trachtman
- Division of Nephrology, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Tomoko Takano
- Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada; Division of Nephrology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
13
|
Park J, Bartzoka F, von Beck T, Li ZN, Mishina M, Hebert LS, Kain J, Liu F, Sharma S, Cao W, Eddins DJ, Kumar A, Kim JE, Lee JS, Wang Y, Schwartz EA, Brilot AF, Satterwhite E, Towers DM, McKnight E, Pohl J, Thompson MG, Gaglani M, Dawood FS, Naleway AL, Stevens J, Kennedy RB, Jacob J, Lavinder JJ, Levine MZ, Gangappa S, Ippolito GC, Sambhara S, Georgiou G. Molecular features of the serological IgG repertoire elicited by egg-based, cell-based, or recombinant haemagglutinin-based seasonal influenza vaccines: a comparative, prospective, observational cohort study. THE LANCET. MICROBE 2025; 6:100935. [PMID: 39667375 PMCID: PMC11807745 DOI: 10.1016/j.lanmic.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/15/2024] [Accepted: 06/27/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Egg-based inactivated quadrivalent seasonal influenza vaccine (eIIV4), cell culture-based inactivated quadrivalent seasonal influenza vaccine (ccIIV4), and recombinant haemagglutinin (HA)-based quadrivalent seasonal influenza vaccine (RIV4) have been licensed for use in the USA. In this study, we used antigen-specific serum proteomics analysis to assess how the molecular composition and qualities of the serological antibody repertoires differ after seasonal influenza immunisation by each of the three vaccines and how different vaccination platforms affect the HA binding affinity and breadth of the serum antibodies that comprise the polyclonal response. METHODS In this comparative, prospective, observational cohort study, we included female US health-care personnel (mean age 47·6 years [SD 8]) who received a single dose of RIV4, eIIV4, or ccIIV4 during the 2018-19 influenza season at Baylor Scott & White Health (Temple, TX, USA). Eligible individuals were selected based on comparable day 28 serum microneutralisation titres and similar vaccination history. Laboratory investigators were blinded to assignment until testing was completed. The preplanned exploratory endpoints were assessed by deconvoluting the serological repertoire specific to A/Singapore/INFIMH-16-0019/2016 (H3N2) HA before (day 0) and after (day 28) immunisation using bottom-up liquid chromatography-mass spectrometry proteomics (referred to as Ig-Seq) and natively paired variable heavy chain-variable light chain high-throughput B-cell receptor sequencing (referred to as BCR-Seq). Features of the antigen-specific serological repertoire at day 0 and day 28 for the three vaccine groups were compared. Antibodies identified with high confidence in sera were recombinantly expressed and characterised in depth to determine the binding affinity and breadth to time-ordered H3 HA proteins. FINDINGS During September and October of the 2018-19 influenza season, 15 individuals were recruited and assigned to receive RIV4 (n=5), eIIV4 (n=5), or ccIIV4 (n=5). For all three cohorts, the serum antibody repertoire was dominated by back-boosted antibody lineages (median 98% [95% CI 88-99]) that were present in the serum before vaccination. Although vaccine platform-dependent differences were not evident in the repertoire diversity, somatic hypermutation, or heavy chain complementarity determining region 3 biochemical features, antibodies boosted by RIV4 showed substantially higher binding affinity to the vaccine H3/HA (median half-maximal effective concentration [EC50] to A/Singapore/INFIMH-16-0019/2016 HA: 0·037 μg/mL [95% CI 0·012-0·12] for RIV4; 4·43 μg/mL [0·030-100·0] for eIIV4; and 18·50 μg/mL [0·99-100·0] μg/mL for ccIIV4) and also the HAs from contemporary H3N2 strains than did those elicited by eIIV4 or ccIIV4 (median EC50 to A/Texas/50/2012 HA: 0·037 μg/mL [0·017-0·32] for RIV4; 1·10 μg/mL [0·045-100] for eIIV4; and 12·6 μg/mL [1·8-100] for ccIIV4). Comparison of B-cell receptor sequencing repertoires on day 7 showed that eIIV4 increased the median frequency of canonical egg glycan-targeting B cells (0·20% [95% CI 0·067-0·37] for eIIV4; 0·058% [0·050-0·11] for RIV4; and 0·035% [0-0·062] for ccIIV4), whereas RIV4 vaccination decreased the median frequency of B-cell receptors displaying stereotypical features associated with membrane proximal anchor-targeting antibodies (0·062% [95% CI 0-0·084] for RIV4; 0·12% [0·066-0·16] for eIIV4; and 0·18% [0·016-0·20] for ccIIV4). In exploratory analysis, we characterised the structure of a highly abundant monoclonal antibody that binds to both group 1 and 2 HAs and recognises the HA trimer interface, despite its sequence resembling the stereotypical sequence motif found in membrane-proximal anchor binding antibodies. INTERPRETATION Although all three licensed seasonal influenza vaccines elicit serological antibody repertoires with indistinguishable features shaped by heavy imprinting, the RIV4 vaccine selectively boosts higher affinity monoclonal antibodies to contemporary strains and elicits greater serum binding potency and breadth, possibly as a consequence of the multivalent structural features of the HA immunogen in this vaccine formulation. Collectively, our findings show advantages of RIV4 vaccines and more generally highlight the benefits of multivalent HA immunogens in promoting higher affinity serum antibody responses. FUNDING Centers for Disease Control and Prevention, National Institutes of Health, and Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Juyeon Park
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA; Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Foteini Bartzoka
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Troy von Beck
- Department of Microbiology and Immunology, Emory Vaccine Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - Zhu-Nan Li
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Luke S Hebert
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Jessica Kain
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Feng Liu
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Suresh Sharma
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Weiping Cao
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Devon J Eddins
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Amrita Kumar
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jin Eyun Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Justin S Lee
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Yuanyuan Wang
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Evan A Schwartz
- Sauer Structural Biology Laboratory, Center for Biomedical Research Support, The University of Texas at Austin, Austin, TX, USA
| | - Axel F Brilot
- Sauer Structural Biology Laboratory, Center for Biomedical Research Support, The University of Texas at Austin, Austin, TX, USA
| | - Ed Satterwhite
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Dalton M Towers
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Eric McKnight
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Jan Pohl
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Mark G Thompson
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Manjusha Gaglani
- Baylor Scott & White Health, Baylor College of Medicine and Texas A&M University College of Medicine, Temple, TX, USA
| | | | | | - James Stevens
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Joshy Jacob
- Department of Microbiology and Immunology, Emory Vaccine Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - Jason J Lavinder
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA; Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Min Z Levine
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Gregory C Ippolito
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | | | - George Georgiou
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA; Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA; Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
14
|
Song S, Wang C, Chen Y, Zhou X, Han Y, Zhang H. Dynamic roles of tumor-infiltrating B lymphocytes in cancer immunotherapy. Cancer Immunol Immunother 2025; 74:92. [PMID: 39891668 PMCID: PMC11787113 DOI: 10.1007/s00262-024-03936-7] [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: 12/04/2024] [Accepted: 12/27/2024] [Indexed: 02/03/2025]
Abstract
The amazing diversity of B cells within the tumor microenvironment is the basis for the diverse development of B cell-based immunotherapies. Here, we focus on elucidating the mechanisms of tumor intervention mediated by four tumor-infiltrating B lymphocytes. Naive B cells present the initial antigen, germinal center B cell subsets enhance antibody affinity, and immunoglobulin subtypes exert multiple immune effects, while regulatory B cells establish immune tolerance. Together they reflect the complexity of the changing dynamics of cancer immunity. Additionally, we have investigated the dynamic effects of tumor-infiltrating B lymphocytes in immunotherapy and their relationship to prognosis, providing new insights into potential treatment strategies for patients.
Collapse
Affiliation(s)
- Shishengnan Song
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Chong Wang
- Department of Thoracic Surgery, Beijing Chest Hospital Affiliated to Capital Medical University (Beijing Tuberculosis and Thoracic Tumor Research Institute), 9 Beiguan Street, Tongzhou, 101149, Beijing, China
| | - Yangchao Chen
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, 999077, NT, China
| | - Xiaorong Zhou
- Department of Immunology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Yi Han
- Department of Thoracic Surgery, Beijing Chest Hospital Affiliated to Capital Medical University (Beijing Tuberculosis and Thoracic Tumor Research Institute), 9 Beiguan Street, Tongzhou, 101149, Beijing, China.
| | - Haijian Zhang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
15
|
Bucheli OTM, Rodrigues D, Ulbricht C, Hauser AE, Eyer K. Dynamic Activation of NADPH Oxidases in Immune Responses Modulates Differentiation, Function, and Lifespan of Plasma Cells. Eur J Immunol 2025; 55:e202350975. [PMID: 39931760 PMCID: PMC11811814 DOI: 10.1002/eji.202350975] [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: 12/30/2023] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
NADPH-oxidase (NOX)-derived reactive oxygen species (ROS) have been described to play essential roles in B-cell activation processes. However, several key questions concerning NOX activity and subsequent ROS production remain unaddressed, including fundamental processes such as differentiation, functional competence, cellular metabolism, and viability. This study investigated these questions in a murine B-cell response after secondary immunization. We combined single-cell transcriptomics and single-cell detection of NOX activity and observed that various subsets of B cells dynamically express NOX1 and NOX2. The NOX+ cellular phenotype correlated with increased activity of metabolic pathways, augmented lactate production, lower IgG secretion rates, and markers for longevity. The NOX+ cellular phenotype was also associated with increased cellular stress and apoptosis, underscoring the intricate relationship between ROS and cellular survival. Consequently, these insights advance our understanding of how long-lived humoral immunity is formed.
Collapse
Affiliation(s)
- Olivia T. M. Bucheli
- ETH Laboratory for Functional Immune Repertoire AnalysisInstitute of Pharmaceutical Sciences, D‐CHAB, ETH ZürichZürichSwitzerland
| | - Daniela Rodrigues
- ETH Laboratory for Functional Immune Repertoire AnalysisInstitute of Pharmaceutical Sciences, D‐CHAB, ETH ZürichZürichSwitzerland
| | - Carolin Ulbricht
- Department of Rheumatology and Clinical ImmunologyCharité ‐ Universitätsmedizin Berlincorporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Immune DynamicsDeutsches Rheuma‐Forschungszentrum (DRFZ)a Leibniz Institute, Charitéplatz 1BerlinGermany
| | - Anja E. Hauser
- Department of Rheumatology and Clinical ImmunologyCharité ‐ Universitätsmedizin Berlincorporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Immune DynamicsDeutsches Rheuma‐Forschungszentrum (DRFZ)a Leibniz Institute, Charitéplatz 1BerlinGermany
| | - Klaus Eyer
- ETH Laboratory for Functional Immune Repertoire AnalysisInstitute of Pharmaceutical Sciences, D‐CHAB, ETH ZürichZürichSwitzerland
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| |
Collapse
|
16
|
Zhu Y, Lu Z, Wang Z, Liu J, Ning K. Based on the immune system: the role of the IL-2 family in pancreatic disease. Front Immunol 2025; 16:1480496. [PMID: 39958351 PMCID: PMC11825815 DOI: 10.3389/fimmu.2025.1480496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/09/2025] [Indexed: 02/18/2025] Open
Abstract
The IL-2 family, consisting of IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21, is a key regulator of the immune response. As an important endocrine and digestive organ, the function of the pancreas is regulated by the immune system. Studies have shown that each cytokine of the IL-2 family influences the occurrence and development of pancreatic diseases by participating in the regulation of the immune system. In this paper, we review the structural and functional characteristics of IL-2 family members, focus on their molecular mechanisms in pancreatic diseases including acute pancreatitis, chronic pancreatitis and pancreatic cancer, and highlight the importance of the related proteins in the regulation of immune response and disease progression, which will provide valuable insights for new biomarkers in pancreatic diseases, early diagnosis of the diseases, assessment of the disease severity, and development of new therapeutic regimens. The insights of the study are summarized in the following sections.
Collapse
Affiliation(s)
| | | | | | | | - Ke Ning
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
17
|
Wang M, Zhang Q, Ju R, Xia J, Xu C, Chen W, Zhang X. Characterization of TCRβ and IGH Repertoires in the Spleen of Two Chicken Lines with Differential ALV-J Susceptibility Under Normal and Infection Conditions. Animals (Basel) 2025; 15:334. [PMID: 39943104 PMCID: PMC11816060 DOI: 10.3390/ani15030334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
This study investigates the immunological factors underlying the differential susceptibility of two chicken strains, E- and M-lines, to avian leukosis virus subgroup J (ALV-J). During the eradication of avian leukosis at a chicken breeder farm in Guangdong, we observed strain-specific differences in susceptibility to ALV-J. Moreover, E-line chickens exhibited a slower antibody response to ALV-J compared to M-line chickens. As the T cell receptor (TCR) and B cell receptor (BCR) are critical for antigen recognition, their activation triggers specific immune responses, including antibody production. Using high-throughput sequencing, we characterized the T cell receptor beta (TCRβ) and immunoglobulin heavy chain (IGH) repertoires in spleen tissues from both chicken strains. The M-line demonstrated higher clonal diversity in both TCRβ and IGH repertoires under normal conditions compared to the E-line, suggesting a broader baseline antigen recognition capacity. Following ALV-J infection, the TCRβ repertoire diversity remained unchanged, while the IGH repertoire displayed distinct clonal expansion patterns and complementarity-determining region 3 (CDR3) length distributions between the two lines, potentially affecting their ability to recognize ALV-J antigens. Our study provides the first comprehensive comparison of TCRβ and IGH repertoire dynamics in chickens with different ALV-J susceptibilities, offering new insights into the molecular and immunological mechanisms underlying resistance to ALV-J.
Collapse
Affiliation(s)
- Meihuizi Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.W.); (Q.Z.); (R.J.); (J.X.); (C.X.); (W.C.)
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Qihong Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.W.); (Q.Z.); (R.J.); (J.X.); (C.X.); (W.C.)
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Rongyang Ju
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.W.); (Q.Z.); (R.J.); (J.X.); (C.X.); (W.C.)
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Junliang Xia
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.W.); (Q.Z.); (R.J.); (J.X.); (C.X.); (W.C.)
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Chengxun Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.W.); (Q.Z.); (R.J.); (J.X.); (C.X.); (W.C.)
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Weiding Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.W.); (Q.Z.); (R.J.); (J.X.); (C.X.); (W.C.)
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Xiquan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.W.); (Q.Z.); (R.J.); (J.X.); (C.X.); (W.C.)
- Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
18
|
Antoniolli M, Solovey M, Hildebrand JA, Freyholdt T, Strobl CD, Bararia D, Keay WD, Adolph L, Heide M, Passerini V, Winter L, Wange L, Enard W, Thieme S, Blum H, Rudelius M, Mergner J, Ludwig C, Bultmann S, Schmidt-Supprian M, Leonhardt H, Subklewe M, von Bergwelt-Baildon M, Colomé-Tatché M, Weigert O. ARID1A mutations protect follicular lymphoma from FAS-dependent immune surveillance by reducing RUNX3/ETS1-driven FAS-expression. Cell Death Differ 2025:10.1038/s41418-025-01445-3. [PMID: 39843653 DOI: 10.1038/s41418-025-01445-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/29/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
The cell death receptor FAS and its ligand (FASLG) play crucial roles in the selection of B cells during the germinal center (GC) reaction. Failure to eliminate potentially harmful B cells via FAS can lead to lymphoproliferation and the development of B cell malignancies. The classic form of follicular lymphoma (FL) is a prototypic GC-derived B cell malignancy, characterized by the t(14;18)(q32;q21)IGH::BCL2 translocation and overexpression of antiapoptotic BCL2. Additional alterations were shown to be clinically relevant, including mutations in ARID1A. ARID1A is part of the SWI/SNF nucleosome remodeling complex that regulates DNA accessibility ("openness"). However, the mechanism how ARID1A mutations contribute to FL pathogenesis remains unclear. We analyzed 151 FL biopsies of patients with advanced-stage disease at initial diagnosis and found that ARID1A mutations were recurrent and mainly disruptive, with an overall frequency of 18%. Additionally, we observed that ARID1A mutant FL showed significantly lower FAS protein expression in the FL tumor cell population. Functional experiments in BCL2-translocated lymphoma cells demonstrated that ARID1A is directly involved in the regulation of FAS, and ARID1A loss leads to decreased FAS protein and gene expression. However, ARID1A loss did not affect FAS promotor openness. Instead, we identified and experimentally validated a previously unknown co-transcriptional complex consisting of RUNX3 and ETS1 that regulates FAS expression, and ARID1A loss leads to reduced RUNX3 promotor openness and gene expression. The reduced FAS levels induced by ARID1A loss rendered lymphoma cells resistant to both soluble and T cell membrane-anchored FASLG-induced apoptosis, and significantly diminished CAR T cell killing in functional experiments. In summary, we have identified a functionally and clinically relevant mechanism how FL cells can escape FAS-dependent immune surveillance, which may also impact the efficacy of T cell-based therapies, including CAR T cells.
Collapse
Affiliation(s)
- Martina Antoniolli
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), LMU University Hospital, Munich, Germany
- Department of Medicine III, LMU University Hospital, Munich, Germany
| | - Maria Solovey
- Biomedical Center (BMC), Department of Physiological Chemistry, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Munich, Germany
| | - Johannes Adrian Hildebrand
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), LMU University Hospital, Munich, Germany
- Department of Medicine III, LMU University Hospital, Munich, Germany
- German Cancer Consortium (DKTK), Munich, Germany; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tabea Freyholdt
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), LMU University Hospital, Munich, Germany
- Department of Medicine III, LMU University Hospital, Munich, Germany
| | - Carolin Dorothea Strobl
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), LMU University Hospital, Munich, Germany
- Department of Medicine III, LMU University Hospital, Munich, Germany
| | - Deepak Bararia
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), LMU University Hospital, Munich, Germany
- Department of Medicine III, LMU University Hospital, Munich, Germany
| | - William David Keay
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), LMU University Hospital, Munich, Germany
- Department of Medicine III, LMU University Hospital, Munich, Germany
- German Cancer Consortium (DKTK), Munich, Germany; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Louisa Adolph
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), LMU University Hospital, Munich, Germany
- Department of Medicine III, LMU University Hospital, Munich, Germany
| | - Michael Heide
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), LMU University Hospital, Munich, Germany
- Department of Medicine III, LMU University Hospital, Munich, Germany
- German Cancer Consortium (DKTK), Munich, Germany; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena Passerini
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), LMU University Hospital, Munich, Germany
- Department of Medicine III, LMU University Hospital, Munich, Germany
| | - Lis Winter
- Department of Medicine III, LMU University Hospital, Munich, Germany
- Laboratory for Translational Cancer Immunology, Gene Center, LMU Munich, Munich, Germany
| | - Lucas Wange
- Anthropology and Human Genomics, Faculty of Biology, LMU Munich, Planegg, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, LMU Munich, Planegg, Germany
| | - Susanne Thieme
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Martina Rudelius
- Department of Medicine III, LMU University Hospital, Munich, Germany
- Institute of Pathology, LMU University Hospital, Munich, Germany
| | - Julia Mergner
- Bavarian Center for Biomolecular Mass Spectrometry at Klinikum Rechts der Isar (BayBioMS@MRI), Technical University Munich, Munich, Germany
| | - Christina Ludwig
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioM), TUM School of Life Science, Technical University Munich, Munich, Germany
| | - Sebastian Bultmann
- Faculty of Biology and Center for Molecular Biosystems (BioSysM), Human Biology and BioImaging, LMU Munich, Planegg, Germany
| | - Marc Schmidt-Supprian
- German Cancer Consortium (DKTK), Munich, Germany; and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Experimental Hematology, TranslaTUM, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Heinrich Leonhardt
- Faculty of Biology and Center for Molecular Biosystems (BioSysM), Human Biology and BioImaging, LMU Munich, Planegg, Germany
| | - Marion Subklewe
- Department of Medicine III, LMU University Hospital, Munich, Germany
- German Cancer Consortium (DKTK), Munich, Germany; and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Laboratory for Translational Cancer Immunology, Gene Center, LMU Munich, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), LMU University Hospital, Munich, Germany
- Department of Medicine III, LMU University Hospital, Munich, Germany
- German Cancer Consortium (DKTK), Munich, Germany; and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Comprehensive Cancer Center Munich (CCCM), University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Centre (BZKF), Munich, Germany
| | - Maria Colomé-Tatché
- Biomedical Center (BMC), Department of Physiological Chemistry, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Munich, Germany.
- Institute of Computational Biology, Helmholtz Zentrum Munich, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Oliver Weigert
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), LMU University Hospital, Munich, Germany.
- Department of Medicine III, LMU University Hospital, Munich, Germany.
- German Cancer Consortium (DKTK), Munich, Germany; and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Cancer Research Centre (BZKF), Munich, Germany.
| |
Collapse
|
19
|
Schulz S, Tan TJC, Wu NC, Wang S. Epistatic hotspots organize antibody fitness landscape and boost evolvability. Proc Natl Acad Sci U S A 2025; 122:e2413884122. [PMID: 39773024 PMCID: PMC11745389 DOI: 10.1073/pnas.2413884122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
The course of evolution is strongly shaped by interaction between mutations. Such epistasis can yield rugged sequence-function maps and constrain the availability of adaptive paths. While theoretical intuition is often built on global statistics of large, homogeneous model landscapes, mutagenesis measurements necessarily probe a limited neighborhood of a reference genotype. It is unclear to what extent local topography of a real epistatic landscape represents its global shape. Here, we demonstrate that epistatic landscapes can be heterogeneously rugged and this heterogeneity may render biomolecules more evolvable. By characterizing a multipeaked fitness landscape of a SARS-CoV-2 antibody mutant library, we show that heterogeneous ruggedness arises from sparse epistatic hotspots, whose mutation impacts the fitness effect of numerous sequence sites. Surprisingly, mutating an epistatic hotspot may enhance, rather than reduce, the accessibility of the fittest genotype, while increasing the overall ruggedness. Further, migratory constraints in real space alleviate mutational constraints in sequence space, which not only diversify direct paths taken but may also turn a road-blocking fitness peak into a stepping stone leading toward the global optimum. Our results suggest that a hierarchy of epistatic hotspots may organize the fitness landscape in such a way that path-orienting ruggedness confers global smoothness.
Collapse
Affiliation(s)
- Steven Schulz
- Department of Physics and Astronomy, University of California, Los Angeles, CA90095
| | - Timothy J. C. Tan
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Nicholas C. Wu
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Shenshen Wang
- Department of Physics and Astronomy, University of California, Los Angeles, CA90095
| |
Collapse
|
20
|
Li S, Zhu Q, Huang A, Lan Y, Wei X, He H, Meng X, Li W, Lin Y, Yang S. A machine learning model and identification of immune infiltration for chronic obstructive pulmonary disease based on disulfidptosis-related genes. BMC Med Genomics 2025; 18:7. [PMID: 39780155 PMCID: PMC11715737 DOI: 10.1186/s12920-024-02076-2] [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: 02/06/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a chronic and progressive lung disease. Disulfidptosis-related genes (DRGs) may be involved in the pathogenesis of COPD. From the perspective of predictive, preventive, and personalized medicine (PPPM), clarifying the role of disulfidptosis in the development of COPD could provide a opportunity for primary prediction, targeted prevention, and personalized treatment of the disease. METHODS We analyzed the expression profiles of DRGs and immune cell infiltration in COPD patients by using the GSE38974 dataset. According to the DRGs, molecular clusters and related immune cell infiltration levels were explored in individuals with COPD. Next, co-expression modules and cluster-specific differentially expressed genes were identified by the Weighted Gene Co-expression Network Analysis (WGCNA). Comparing the performance of the random forest (RF), support vector machine (SVM), generalized linear model (GLM), and eXtreme Gradient Boosting (XGB), we constructed the ptimal machine learning model. RESULTS DE-DRGs, differential immune cells and two clusters were identified. Notable difference in DRGs, immune cell populations, biological processes, and pathway behaviors were noted among the two clusters. Besides, significant differences in DRGs, immune cells, biological functions, and pathway activities were observed between the two clusters.A nomogram was created to aid in the practical application of clinical procedures. The SVM model achieved the best results in differentiating COPD patients across various clusters. Following that, we identified the top five genes as predictor genes via SVM model. These five genes related to the model were strongly linked to traits of the individuals with COPD. CONCLUSION Our study demonstrated the relationship between disulfidptosis and COPD and established an optimal machine-learning model to evaluate the subtypes and traits of COPD. DRGs serve as a target for future predictive diagnostics, targeted prevention, and individualized therapy in COPD, facilitating the transition from reactive medical services to PPPM in the management of the disease.
Collapse
Affiliation(s)
- Sijun Li
- Infectious Disease Laboratory, The Fourth People's Hospital of Nanning, Nanning, China
| | - Qingdong Zhu
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Aichun Huang
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Yanqun Lan
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Xiaoying Wei
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Huawei He
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Xiayan Meng
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Weiwen Li
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China
| | - Yanrong Lin
- Department of Tuberculosis, The Fourth People's Hospital of Nanning, Nanning, China.
| | - Shixiong Yang
- Administrative Office, The Fourth People's Hospital of Nanning, Nanning, China.
| |
Collapse
|
21
|
Xing Y, Guo W, Wu M, Xie J, Huang D, Hu P, Zhou M, Zhang L, Zhong Y, Liu M, Chen Y, Yi Z. A Small-Molecule BCL6 Inhibitor as an Anti-Proliferative Agent for Diffuse Large B-Cell Lymphoma. Mol Cancer Ther 2025; 24:81-92. [PMID: 39387112 DOI: 10.1158/1535-7163.mct-23-0830] [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: 11/25/2023] [Revised: 05/09/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
The B-cell lymphoma 6 (BCL6) transcription factor plays a key role in the establishment of germinal center (GC) formation. Diffuse large B-cell lymphoma (DLBCL) originates from the GC reaction due to dysregulation of BCL6. Disrupting BCL6 and its corepressors' interaction has become the foundation for rationally designing lymphoma therapies. However, BCL6 inhibitors with good activities in vitro and in vivo are rare, and there are no clinically approved BCL6 inhibitors. In this study, we discovered and developed a novel range of [1,2,4] triazolo[1,5-a] pyrimidine derivatives targeting BCL6/SMRT interaction. The lead compound WK692 directly bound BCL6BTB, disrupted BCL6BTB/SMRT interaction and activated the expression of BCL6 downstream genes inside cells, inhibited DLBCL growth and induced apoptosis in vitro, inhibited GC formation, decreased the proportion of follicular helper T cells, and impaired Ig affinity maturation. Further studies showed that WK692 inhibits DLBCL growth without toxic effects in vivo and synergizes with the EZH2 and PRMT5 inhibitors. Our results demonstrated that WK692 as a BCL6 inhibitor may be developed as a novel potential anticancer agent against DLBCL.
Collapse
Affiliation(s)
- Yajing Xing
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Weikai Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Min Wu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiuqing Xie
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Dongxia Huang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Pan Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Miaoran Zhou
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lin Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yadong Zhong
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products and Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, China
| | - Zhengfang Yi
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
| |
Collapse
|
22
|
Dent AL. Regulation of the IgE response by T follicular regulatory cells. Allergol Int 2025; 74:20-24. [PMID: 39232918 DOI: 10.1016/j.alit.2024.08.004] [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: 07/18/2024] [Accepted: 07/20/2024] [Indexed: 09/06/2024] Open
Abstract
Allergen-specific IgE is a major mediator of allergic responses and contributes greatly to allergic disease in the human population. Therapies that inhibit the production of IgE would be useful for lessening the burden of allergic disease. A great deal of research has focused on how IgE responses are regulated, and several factors that promote the production of allergic IgE have been characterized. T follicular helper (TFH) cells expressing IL-4 are required for the development of IgE expressing B cells in the germinal center (GC). Ig somatic hypermutation and B cell selection in the GC leads to the development of high affinity allergen-specific IgE that promotes anaphylaxis, a severe form of allergic response. T follicular regulatory (TFR) cells are also found in the GC response and act with TFH cells in the selection of high affinity IgE + B cells. This review examines the current literature on IgE responses and TFR cells. In mouse studies, TFR cells have a suppressive role on IgE responses in allergic airway disease, however TFR cells also play a helper role in the IgE response in food allergy. In human studies, TFR cells correlate with a decreased allergic response but evidence for a direct suppressive role of TFR cells on IgE in vivo is lacking. TFR cells may represent a new target for allergy therapies, but caution must be exercised to promote the suppressor activity of TFR cells and not the helper activity of TFR cells on IgE responses.
Collapse
Affiliation(s)
- Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, 950 W. Walnut St., R2 302 Indianapolis, IN 46202, USA.
| |
Collapse
|
23
|
Nielsen OH, Hammerhøj A, Ainsworth MA, Gubatan J, D'Haens G. Immunogenicity of Therapeutic Antibodies Used for Inflammatory Bowel Disease: Treatment and Clinical Considerations. Drugs 2025; 85:67-85. [PMID: 39532820 DOI: 10.1007/s40265-024-02115-3] [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] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
The introduction of tumor necrosis factor inhibitors has led to a paradigm shift in the management of inflammatory bowel disease (IBD). The subsequent introduction of both anti-integrins and cytokine blockers has since expanded the biologic armamentarium. However, immunogenicity, defined as the production of anti-drug antibodies (ADAs) to the prescribed biopharmaceutical, means a significant fraction of patients exposed to biologic agents will experience a secondary loss of response to one or more of the drugs. In clinical settings, immunogenicity may be caused by several factors, both patient related (e.g., underlying chronic disease, systemic immune burden, including previous biologic therapy failure, and [epi]genetic background) and treatment related (e.g., dose and administration regimens, drug physical structure, photostability, temperature, and agitation). Here, we outline these elements in detail to enhance biopharmaceutical delivery and therapy for patients with IBD. Moreover, concurrent immunomodulator medication may reduce the risks of ADA generation, especially when using the chimeric drug infliximab. Summarizing the latest developments and knowledge in the field, this review aims to provide strategies to prevent ADA production and information on managing non-responsiveness or loss of response to biologics. Better understanding of the molecular mechanisms underlying the formation of ADAs and the critical factors influencing the immunogenicity of biopharmaceuticals may lead to improved health outcomes in the IBD community that may benefit both the individual patient and society through lower healthcare expenses.
Collapse
Affiliation(s)
- Ole Haagen Nielsen
- Department of Gastroenterology D112, Herlev Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 1, 2730 Herlev, Copenhagen, Denmark.
| | - Alexander Hammerhøj
- Department of Gastroenterology D112, Herlev Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 1, 2730 Herlev, Copenhagen, Denmark
| | - Mark Andrew Ainsworth
- Department of Gastroenterology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - John Gubatan
- Department of Gastroenterology & Hepatology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Geert D'Haens
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Datta M. Analysis of Germinal Center Reaction in Competitive Bone Marrow Chimeric Rag2 -/-γc -/-Mice. Methods Mol Biol 2025; 2909:19-29. [PMID: 40029512 DOI: 10.1007/978-1-0716-4442-3_2] [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] [Indexed: 03/05/2025]
Abstract
Germinal center (GC) reaction is crucial for the generation of high-affinity antibodies against any infection. To address the role of specific genes in GC, knockout mouse models are generally used. However, since GC is a multicellular event, complete knockout models cannot pinpoint the cell-intrinsic effect of certain genes in GC reaction. Here, we describe a detailed protocol for the analysis of GC in competitive bone marrow (BM) chimeric mice generated by transplanting 1:1 mixture of CD45.1 and CD45.2 positive donor bone marrow (BM) cells in immunodeficient Rag2-/-γc-/- mice as recipient host. We describe the method of immunization using sheep red blood cells (SRBC), detection of successful immunization by the antigen-specific antibody titer in the serum of the immunized mice and finally assessing the GC reaction in the spleen of the immunized mice by flow cytometry 10 days post-immunization. When the competitive chimera is made with wild type (WT) and a knockout (KO) donor BM, this method is suitable to address the cell type-specific (e.g., B-cell specific) role of the KO gene in GC.
Collapse
Affiliation(s)
- Moumita Datta
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
25
|
Wu N, Liu Y, Miao C, Yu Z, Ma G, Wu J. Enhancing the Deformability of the Adjuvant: Achieving Lymph Node Targeted Delivery to Elicit a Long-Lasting Immune Protection. Adv Healthc Mater 2025; 14:e2401520. [PMID: 39632365 DOI: 10.1002/adhm.202401520] [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: 04/25/2024] [Revised: 10/12/2024] [Indexed: 12/07/2024]
Abstract
A key challenge in vaccine development is to induce an effective and durable immune response. Live virus vaccines induce lifelong antibody responses; however, the immune responses induced by inactivated or subunit vaccines decrease gradually. Activation of the germinal center (GC) reaction, which generates long-lived plasma cells (LLPCs), is a key mediator of long-term antibody responses. To enhance the activation of GC, lymph node-targeted delivery of the vaccine is promoted by enhancing the deformability of the delivery vector. In this study, a double emulsion is designed with strong deformability and containing chitosan nanoparticles (CSNP) in the internal aqueous phase (WNP) for efficient antigen loading, called WNP/O/W. The flexible oil layer and the internally loaded positively charged particles endow the emulsion with strong deformability, continuously enrich model antigen ovalbumin (OVA) in the lymph nodes, activate germinal center B (GC B) cells and T follicular helper (TFH) cells, induce LLPCs, and obtain high-level antibody persistence for more than 5 months, which is significantly better than the traditional oil emulsion adjuvant. Concurrently, it also improves the immune-protective effect in aged mice. Altogether, these results indicate that WNP/O/W achieves lymph node targeted delivery by strengthening deformability, generating high-intensity antibody responses, and long-lasting immune protection.
Collapse
Affiliation(s)
- Nan Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yuyang Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemistry Engineering, Nanjing Tech University, Nanjing, 211816, P. R. China
| | - Chunyu Miao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Ziyi Yu
- School of Chemistry Engineering, Nanjing Tech University, Nanjing, 211816, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Jie Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| |
Collapse
|
26
|
Hadebe S. Adoptive Transfer of B Cells In Vivo for Assessment of Their Immune Function. Methods Mol Biol 2025; 2909:3-17. [PMID: 40029511 DOI: 10.1007/978-1-0716-4442-3_1] [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] [Indexed: 03/05/2025]
Abstract
B lymphocytes are a critical part of the adaptive immune response elicited by the immune system to fight various pathogens. The main effector function of the B lymphocytes is the ability to secrete antibodies, whether nonclass-switched immunoglobulin M (IgM) or class-switched immunoglobulin isotypes. To understand the function of B cells in vivo, mice are subjected to bone marrow depletion (using radiation or chemical radiation) before being adoptively transferred with donor bone marrow. Alternatively, B cells can be isolated from spleens and adoptively transferred to B-cell-deficient recipient mice to demonstrate function. In this chapter, we will outline the protocol used to isolate and transfer B cells to B-cell-deficient (μMT-/-) mice in vivo to study their functions.
Collapse
Affiliation(s)
- Sabelo Hadebe
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
27
|
Corinaldesi C, Holmes AB, Martire G, Tosato A, Rizzato D, Lovisa F, Gallingani I, Shen Q, Ferrone L, Harris M, Davies K, Molinaro L, Mortara U, Dei Tos AP, Ofori K, D'Amore ESG, Chiarle R, Ngan B, Carraro E, Pillon M, Hussein S, Bhagat G, Pizzi M, Mussolin L, Basso K. Single-cell transcriptomics of pediatric Burkitt lymphoma reveals intra-tumor heterogeneity and markers of therapy resistance. Leukemia 2025; 39:189-198. [PMID: 39424708 PMCID: PMC11717704 DOI: 10.1038/s41375-024-02431-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/08/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024]
Abstract
Burkitt lymphoma (BL) is the most frequent B-cell lymphoma in pediatric patients. While most patients are cured, a fraction of them are resistant to therapy. To investigate BL heterogeneity and the features distinguishing therapy responders (R) from non-responders (NR), we analyzed by single-cell (sc)-transcriptomics diagnostic EBV-negative BL specimens. Analysis of the non-tumor component revealed a predominance of immune cells and a small representation of fibroblasts, enriched in NR. Tumors displayed patient-specific features, as well as shared subpopulations that expressed transcripts related to cell cycle, signaling pathways and cell-of-origin signatures. Several transcripts were differentially expressed in R versus NR. The top candidate, Tropomyosin 2 (TPM2), a member of the tropomyosin actin filament binding protein family, was confirmed to be significantly higher in NR both at the transcript and protein level. Stratification of patients based on TPM2 expression at diagnosis significantly correlated with prognosis, independently of TP53 mutations. These results indicate that BL displays transcriptional heterogeneity and identify candidate biomarkers of therapy resistance.
Collapse
Affiliation(s)
| | - Antony B Holmes
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Gaia Martire
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy
| | - Anna Tosato
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy
| | - Domenico Rizzato
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
| | - Federica Lovisa
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy
| | - Ilaria Gallingani
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy
| | - Qiong Shen
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Lavinia Ferrone
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy
| | - Marian Harris
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Luca Molinaro
- Department of Medical Science, University of Torino, Torino, Italy
| | - Umberto Mortara
- Department of Medical Science, University of Torino, Torino, Italy
| | - Angelo Paolo Dei Tos
- General Pathology and Cytopathology Unit, Department of Medicine-DMED, University-Hospital of Padova, Padova, Italy
| | - Kenneth Ofori
- Department of Pathology & Cell Biology, Columbia University, New York, NY, USA
| | | | - Roberto Chiarle
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- European Institute of Oncology IRCCS, Division of Hematopathology, Milan, Italy
| | - Bo Ngan
- Hospital for Sick Children (SickKids), Toronto, ON, Canada
| | - Elisa Carraro
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
| | - Marta Pillon
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy
| | - Shafinaz Hussein
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Govind Bhagat
- Department of Pathology & Cell Biology, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Marco Pizzi
- General Pathology and Cytopathology Unit, Department of Medicine-DMED, University-Hospital of Padova, Padova, Italy
| | - Lara Mussolin
- Maternal and Child Health Department, University-Hospital of Padova, Padova, Italy.
- Istituto di Ricerca Pediatrica Citta' della Speranza, Padova, Italy.
| | - Katia Basso
- Institute for Cancer Genetics, Columbia University, New York, NY, USA.
- Department of Pathology & Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
28
|
Meng X, Asadi-Asadabad S, Cao S, Song R, Lin Z, Safhi M, Qin Y, Tcheumi Tactoum E, Taudte V, Ekici A, Mielenz D, Wirtz S, Schett G, Bozec A. Metabolic rewiring controlled by HIF-1α tunes IgA-producing B-cell differentiation and intestinal inflammation. Cell Mol Immunol 2025; 22:54-67. [PMID: 39543372 PMCID: PMC11686098 DOI: 10.1038/s41423-024-01233-y] [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: 10/10/2023] [Accepted: 10/18/2024] [Indexed: 11/17/2024] Open
Abstract
Germinal centers where B cells undergo clonal expansion and antibody affinity maturation are hypoxic microenvironments. However, the function of hypoxia-inducible factor (HIF)-1α in immunoglobulin production remains incompletely characterized. Here, we demonstrated that B cells lacking HIF-1α exhibited significantly lower glycolytic metabolism and impaired IgA production. Loss of HIF-1α in B cells affects IgA-producing B-cell differentiation and exacerbates dextran sodium sulfate (DSS)-induced colitis. Conversely, promoting HIF-1α stabilization via a PHD inhibitor roxadustat enhances IgA class switching and alleviates intestinal inflammation. Mechanistically, HIF-1α facilitates IgA class switching through acetyl-coenzyme A (acetyl-CoA) accumulation, which is essential for histone H3K27 acetylation at the Sα region. Consequently, supplementation with acetyl-CoA improved defective IgA production in Hif1a-deficient B cells and limited experimental colitis. Collectively, these findings highlight the critical importance of HIF-1α in IgA class switching and the potential for targeting the HIF-1α-dependent metabolic‒epigenetic axis to treat inflammatory bowel diseases and other inflammatory disorders.
Collapse
Affiliation(s)
- Xianyi Meng
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Sahar Asadi-Asadabad
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Shan Cao
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Rui Song
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Zhen Lin
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Mohammed Safhi
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Yi Qin
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Estelle Tcheumi Tactoum
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Verena Taudte
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054, Germany
- Insitute of Laboratory Medicine, Philipps University of Marburg, Marburg, 35043, Germany
| | - Arif Ekici
- Institute of Human Genetics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Stefan Wirtz
- Department of Internal Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 90154, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany.
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany.
| |
Collapse
|
29
|
Ado S, Dong C, Attaf N, Moussa M, Carrier A, Milpied P, Navarro JM. FB5P-seq-mAbs: monoclonal antibody production from FB5P-seq libraries for integrative single-cell analysis of B cells. Front Immunol 2024; 15:1505971. [PMID: 39742275 PMCID: PMC11685048 DOI: 10.3389/fimmu.2024.1505971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/26/2024] [Indexed: 01/03/2025] Open
Abstract
Parallel analysis of phenotype, transcriptome and antigen receptor sequence in single B cells is a useful method for tracking B cell activation and maturation during immune responses. However, in most cases, the specificity and affinity of the B cell antigen receptor cannot be inferred from its sequence. Antibody cloning and expression from single B cells is then required for functional assays. Here we propose a method that integrates FACS-based 5'-end single-cell RNA sequencing (FB5P-seq) and monoclonal antibody cloning for integrative analysis of single B cells. Starting from a cell suspension, single B cells are FACS-sorted into 96-well plates for reverse transcription, cDNA barcoding and amplification. A fraction of the single-cell cDNA is used for preparing 5'-end RNA-seq libraries that are sequenced for retrieving transcriptome-wide gene expression and paired BCR sequences. The archived cDNA of selected cells of interest is used as input for cloning heavy and light chain variable regions into antibody expression plasmid vectors. The corresponding monoclonal antibodies are produced by transient transfection of a eukaryotic producing cell line and purified for functional assays. We provide detailed step-by-step instructions and describe results obtained on ovalbumin-specific murine germinal center B cells after immunization. Our method is robust, flexible, cost-effective, and applicable to different B cell types and species. We anticipate it will be useful for mapping antigen specificity and affinity of rare B cell subsets characterized by defined gene expression and/or antigen receptor sequence.
Collapse
Affiliation(s)
- Sakina Ado
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Chuang Dong
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Noudjoud Attaf
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Myriam Moussa
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Agathe Carrier
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- Paris-Saclay University, Inserm, Gustave Roussy, Tumour Immunology and Anti-Cancer Immunotherapy, Villejuif, France
| | - Pierre Milpied
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Jean-Marc Navarro
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| |
Collapse
|
30
|
Davis-Porada J, George AB, Lam N, Caron DP, Gray JI, Huang J, Hwu J, Wells SB, Matsumoto R, Kubota M, Lee Y, Morrison-Colvin R, Jensen IJ, Ural BB, Shaabani N, Weiskopf D, Grifoni A, Sette A, Szabo PA, Teijaro JR, Sims PA, Farber DL. Maintenance and functional regulation of immune memory to COVID-19 vaccines in tissues. Immunity 2024; 57:2895-2913.e8. [PMID: 39510068 PMCID: PMC11634668 DOI: 10.1016/j.immuni.2024.10.003] [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: 03/05/2024] [Revised: 06/28/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024]
Abstract
Memory T and B cells in tissues are essential for protective immunity. Here, we performed a comprehensive analysis of the tissue distribution, phenotype, durability, and transcriptional profile of COVID-19 mRNA vaccine-induced immune memory across blood, lymphoid organs, and lungs obtained from 63 vaccinated organ donors aged 23-86, some of whom experienced SARS-CoV-2 infection. Spike (S)-reactive memory T cells were detected in lymphoid organs and lungs and variably expressed tissue-resident markers based on infection history, and S-reactive B cells comprised class-switched memory cells resident in lymphoid organs. Compared with blood, S-reactive tissue memory T cells persisted for longer times post-vaccination and were more prevalent with age. S-reactive T cells displayed site-specific subset compositions and functions: regulatory cell profiles were enriched in tissues, while effector and cytolytic profiles were more abundant in circulation. Our findings reveal functional compartmentalization of vaccine-induced T cell memory where surveilling effectors and in situ regulatory responses confer protection with minimal tissue damage.
Collapse
Affiliation(s)
- Julia Davis-Porada
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alex B George
- Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Nora Lam
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Daniel P Caron
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Joshua I Gray
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jenny Huang
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jennifer Hwu
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Steven B Wells
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rei Matsumoto
- Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Masaru Kubota
- Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - YoonSeung Lee
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rory Morrison-Colvin
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Isaac J Jensen
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Basak B Ural
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Namir Shaabani
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center of Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Alba Grifoni
- Center of Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center of Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Peter A Szabo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - John R Teijaro
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
31
|
Mahdisoltani S, Murugan P, Chakraborty AK, Kardar M. Minimal framework for optimizing vaccination protocols targeting highly mutable pathogens. Phys Rev E 2024; 110:064137. [PMID: 39916243 DOI: 10.1103/physreve.110.064137] [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: 04/24/2024] [Accepted: 10/03/2024] [Indexed: 02/12/2025]
Abstract
A persistent public health challenge is identifying immunization schemes that are effective against highly mutable pathogens such as HIV and influenza viruses. To address this, we analyze a simplified model of affinity maturation, the Darwinian evolutionary process B cells undergo during immunization. The vaccination protocol determines the selection forces that steer affinity maturation to generate antibodies. We focus on identifying the optimal selection forces exerted by a generic time-dependent vaccination protocol to maximize the production of broadly neutralizing antibodies (bnAbs) that can protect against a broad spectrum of pathogen strains. The model utilizes a path integral representation and operator approximations within a mean-field limit and provides guiding principles for optimizing time-dependent vaccine-induced selection forces to enhance bnAb generation. We compare our analytical mean-field results with the outcomes of stochastic simulations, and we discuss their similarities and differences.
Collapse
Affiliation(s)
- Saeed Mahdisoltani
- Massachusetts Institute of Technology, Department of Chemical Engineering, Cambridge, Massachusetts 02139, USA
- Massachusetts Institute of Technology, Institute for Medical Engineering and Science, Cambridge, Massachusetts 02139, USA
- Massachusetts Institute of Technology, Department of Physics, Cambridge, Massachusetts 02139, USA
| | - Pranav Murugan
- Massachusetts Institute of Technology, Department of Physics, Cambridge, Massachusetts 02139, USA
- Massachusetts Institute of Technology, Department of Electrical Engineering and Computer Science, Cambridge, Massachusetts 02139, USA
| | - Arup K Chakraborty
- Massachusetts Institute of Technology, Department of Chemical Engineering, Cambridge, Massachusetts 02139, USA
- Massachusetts Institute of Technology, Institute for Medical Engineering and Science, Cambridge, Massachusetts 02139, USA
- Massachusetts Institute of Technology, Department of Physics, Cambridge, Massachusetts 02139, USA
- Massachusetts Institute of Technology, Ragon Institute of Massachusetts General Hospital, and Harvard University, Cambridge, Massachusetts 02139, USA
- Massachusetts Institute of Technology, Department of Chemistry, Cambridge, Massachusetts 02139, USA
| | - Mehran Kardar
- Massachusetts Institute of Technology, Department of Physics, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
32
|
Juillard S, Karakeussian-Rimbaud A, Normand MH, Turgeon J, Veilleux-Trinh C, C Robitaille A, Rauch J, Chruscinski A, Grandvaux N, Boilard É, Hébert MJ, Dieudé M. Vascular injury derived apoptotic exosome-like vesicles trigger autoimmunity. J Transl Autoimmun 2024; 9:100250. [PMID: 39286649 PMCID: PMC11402544 DOI: 10.1016/j.jtauto.2024.100250] [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: 02/19/2024] [Revised: 07/15/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024] Open
Abstract
According to a central tenet of classical immune theory, a healthy immune system must avoid self-reactive lymphocyte clones but we now know that B cells repertoire exhibit some level of autoreactivity. These autoreactive B cells are thought to rely on self-ligands for their clonal selection and survival. Here, we confirm that healthy mice exhibit self-reactive B cell clones that can be stimulated in vitro by agonists of toll-like receptor (TLR) 1/2, TLR4, TLR7 and TLR9 to secrete anti-LG3/perlecan. LG3/perlecan is an antigen packaged in exosome-like structures released by apoptotic endothelial cells (ApoExos) upon vascular injury. We demonstrate that the injection of ApoExos in healthy animals activates the IL-23/IL-17 pro-inflammatory and autoimmune axis, and produces several autoantibodies, including anti-LG3 autoantibodies and hallmark autoantibodies found in systemic lupus erythematosus. We also identify γδT cells as key mediators of the maturation of ApoExos-induced autoantibodies in healthy mice. Altogether we show that ApoExos released by apoptotic endothelial cells display immune-mediating functions that can stimulate the B cells in the normal repertoire to produce autoantibodies. Our work also identifies TLR activation and γδT cells as important modulators of the humoral autoimmune response induced by ApoExos.
Collapse
Affiliation(s)
- Sandrine Juillard
- Centre de Recherche Du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, R12.218, 900 Rue St-Denis, Montréal, QC, H2X 0A9, Canada
- Université de Montréal, 2900 Bd Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada
- Canadian Donation and Transplantation Research Program (CDTRP), University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Annie Karakeussian-Rimbaud
- Centre de Recherche Du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, R12.218, 900 Rue St-Denis, Montréal, QC, H2X 0A9, Canada
| | - Marie-Hélène Normand
- Centre de Recherche Du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, R12.218, 900 Rue St-Denis, Montréal, QC, H2X 0A9, Canada
- Université de Montréal, 2900 Bd Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada
- Canadian Donation and Transplantation Research Program (CDTRP), University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Julie Turgeon
- Centre de Recherche Du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, R12.218, 900 Rue St-Denis, Montréal, QC, H2X 0A9, Canada
- Canadian Donation and Transplantation Research Program (CDTRP), University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Charlotte Veilleux-Trinh
- Centre de Recherche Du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, R12.218, 900 Rue St-Denis, Montréal, QC, H2X 0A9, Canada
| | - Alexa C Robitaille
- Centre de Recherche Du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, R12.218, 900 Rue St-Denis, Montréal, QC, H2X 0A9, Canada
- Université de Montréal, 2900 Bd Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Joyce Rauch
- Division of Rheumatology, Research Institute of the McGill University Health Centre (RI MUHC), 1001 Bd Décarie, Montréal, QC, H4A 3J1, Canada
| | | | - Nathalie Grandvaux
- Centre de Recherche Du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, R12.218, 900 Rue St-Denis, Montréal, QC, H2X 0A9, Canada
- Université de Montréal, 2900 Bd Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Éric Boilard
- Centre de Recherche Du CHU de Québec, Université Laval, 2705 Bd Laurier, Québec, QC, G1V 4G2, Canada
| | - Marie-Josée Hébert
- Centre de Recherche Du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, R12.218, 900 Rue St-Denis, Montréal, QC, H2X 0A9, Canada
- Université de Montréal, 2900 Bd Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada
- Canadian Donation and Transplantation Research Program (CDTRP), University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Mélanie Dieudé
- Centre de Recherche Du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, R12.218, 900 Rue St-Denis, Montréal, QC, H2X 0A9, Canada
- Université de Montréal, 2900 Bd Édouard-Montpetit, Montréal, QC, H3T 1J4, Canada
- Canadian Donation and Transplantation Research Program (CDTRP), University of Alberta, Edmonton, AB, T6G 2E1, Canada
- Medical Affairs and Innovation, Héma-Québec, 1070 Avenue des Sciences-de-la-Vie, Québec, QC, G1V 5C3, Canada
| |
Collapse
|
33
|
Holmes S, Li H, Shen X, Martin M, Tuck R, Chen Y, Giorgi EE, Kirshner HF, Berry M, Van Italie E, Venkatayogi S, Martin Beem JS, Edwards RJ, Mansouri K, Singh A, Kuykendall C, Gurley T, Anthony Moody M, DeNayer N, Demarco T, Denny TN, Wang Y, Evangelous TD, Clinton JT, Hora B, Wagh K, Seaman MS, Saunders KO, Solomotis N, Misamore J, Lewis MG, Wiehe K, Montefiori DC, Shaw GM, Williams WB. Neonatal immunity associated with heterologous HIV-1 neutralizing antibody induction in SHIV-infected Rhesus Macaques. Nat Commun 2024; 15:10302. [PMID: 39604409 PMCID: PMC11603298 DOI: 10.1038/s41467-024-54753-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
The details of the pediatric immune system that supports induction of antibodies capable of neutralizing geographically-diverse or heterologous HIV-1 is currently unclear. Here we explore the pediatric immune environment in neonatal macaque undergoing Simian-HIV infection. Simian-HIV infection of 11 pairs of therapy-naive dams and infant rhesus macaques for 24 months results in heterologous HIV-1 neutralizing antibodies in 64% of young macaques compared to 18% of adult macaques. Heterologous HIV-1 neutralizing antibodies emerge by 12 months post-infection in young macaques, in association with lower expression of immunosuppressive genes, fewer germinal center CD4 + T regulatory cells, and a lower ratio of CD4 + T follicular regulatory to helper cells. Antibodies from peripheral blood B cells in two young macaques following SHIV infection neutralize 13% of 119 heterologous HIV-1 strains and map to regions of canonical broadly neutralizing antibody epitopes on the envelope surface protein. Here we show that pediatric immunity to SHIV infection in a macaque model may inform vaccine strategies to induce effective HIV-1 neutralizing antibodies in infants and children prior to viral exposure.
Collapse
Affiliation(s)
- Sommer Holmes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Hui Li
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Mitchell Martin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Ryan Tuck
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Yue Chen
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | | | | | - Madison Berry
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Elizabeth Van Italie
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Sravani Venkatayogi
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Joshua S Martin Beem
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Robert J Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Katayoun Mansouri
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Ajay Singh
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cindy Kuykendall
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Thaddeus Gurley
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Nicole DeNayer
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Todd Demarco
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Thomas N Denny
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Yunfei Wang
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Tyler D Evangelous
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - John T Clinton
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Bhavna Hora
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kshitij Wagh
- Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Michael S Seaman
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | | | | | | | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - David C Montefiori
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - George M Shaw
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wilton B Williams
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
34
|
Wang L, Wan J, He W, Wang Z, Wu Q, Zhou M, Fu ZF, Zhao L. IL-7 promotes mRNA vaccine-induced long-term immunity. J Nanobiotechnology 2024; 22:716. [PMID: 39550592 PMCID: PMC11568559 DOI: 10.1186/s12951-024-02993-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024] Open
Abstract
Messenger RNA (mRNA) vaccines are a key technology in combating existing and emerging infectious diseases. However, improving the immunogenicity and durability of mRNA vaccines remains a challenge. To elicit optimal immune responses, integrating antigen-encoded mRNA and immunostimulatory adjuvants into a single formulation is a promising approach to enhancing the efficacy of mRNA vaccines. Here, we report an adjuvant strategy to enhance the efficacy of mRNA vaccines by co-loading mRNA encoding the antigen (rabies virus glycoprotein, RABV-G) and mRNA encoding IL-7 into lipid nanoparticles, achieving co-delivery to the same antigen-presenting cells. A single immunization with G&IL-7 mRNA vaccine elicited robust humoral immune responses in mice and conferred complete protection against RABV challenge. Notably, the high levels of neutralizing antibody induced by the G&IL-7 mRNA vaccine were maintained for at least 6 months, providing mice with long-term significant and complete protection against RABV. Additionally, IL-7 also enhanced antibody responses against the SARS-CoV-2. These data demonstrate that IL-7 is a potent mRNA vaccine adjuvant that can provide the required immune stimulation in various mRNA vaccine formulations.
Collapse
Affiliation(s)
- Lingli Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jiawu Wan
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenna He
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zongmei Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Qiong Wu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ming Zhou
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhen F Fu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ling Zhao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.
- Hubei Hongshan Laboratory, Wuhan, 430070, China.
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China.
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
35
|
Zhang X, Ma C, Lu Y, Wang J, Yun H, Jiang H, Wu M, Feng X, Gai W, Xu G, Deng H, Feng J, Liu W, Shi T, Cheng Q, Zhang J. Rack1 regulates B-cell development and function by binding to and stabilizing the transcription factor Pax5. Cell Mol Immunol 2024; 21:1282-1295. [PMID: 39256480 PMCID: PMC11528059 DOI: 10.1038/s41423-024-01213-2] [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: 12/24/2023] [Accepted: 08/26/2024] [Indexed: 09/12/2024] Open
Abstract
The transcription factor Pax5 activates genes essential for B-cell development and function. However, the regulation of Pax5 expression remains elusive. The adaptor Rack1 can interact with multiple transcription factors and modulate their activation and/or stability. However, its role in the transcriptional control of B-cell fates is largely unknown. Here, we show that CD19-driven Rack1 deficiency leads to pro-B accumulation and a simultaneous reduction in B cells at later developmental stages. The generation of bone marrow chimeras indicates a cell-intrinsic role of Rack1 in B-cell homeostasis. Moreover, Rack1 augments BCR and TLR signaling in mature B cells. On the basis of the aberrant expression of Pax5-regulated genes, including CD19, upon Rack1 deficiency, further exploration revealed that Rack1 maintains the protein level of Pax5 through direct interaction and consequently prevents Pax5 ubiquitination. Accordingly, Mb1-driven Rack1 deficiency almost completely blocks B-cell development at the pro-B-cell stage. Ectopic expression of Pax5 in Rack1-deficient pro-B cells partially rescues B-cell development. Thus, Rack1 regulates B-cell development and function through, at least partially, binding to and stabilizing Pax5.
Collapse
Affiliation(s)
- Xueting Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Chenke Ma
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yuchen Lu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jing Wang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hongfang Yun
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hui Jiang
- Beijing Institute of Basic Medical Sciences, Beijing, China
- University of South China, Hengyang Medical School, Hengyang, China
| | - Mengyao Wu
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Anhui Medical University, Hefei, China
| | - Xiaoyao Feng
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Henan University Joint National Laboratory for Antibody Drug Engineering, Kaifeng, China
| | - Wenbin Gai
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Guanglei Xu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hongbin Deng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiannan Feng
- Henan University Joint National Laboratory for Antibody Drug Engineering, Kaifeng, China
- National Engineering Research Center for Emergence Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Wanli Liu
- Institute of Life Sciences, Tsinghua University, Beijing, China
| | - Taoxing Shi
- Beijing Institute of Basic Medical Sciences, Beijing, China.
| | - Qianqian Cheng
- Beijing Institute of Basic Medical Sciences, Beijing, China.
| | - Jiyan Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, China.
- University of South China, Hengyang Medical School, Hengyang, China.
- Anhui Medical University, Hefei, China.
- Henan University Joint National Laboratory for Antibody Drug Engineering, Kaifeng, China.
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
36
|
Lam N, Lee Y, Farber DL. A guide to adaptive immune memory. Nat Rev Immunol 2024; 24:810-829. [PMID: 38831162 DOI: 10.1038/s41577-024-01040-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 06/05/2024]
Abstract
Immune memory - comprising T cells, B cells and plasma cells and their secreted antibodies - is crucial for human survival. It enables the rapid and effective clearance of a pathogen after re-exposure, to minimize damage to the host. When antigen-experienced, memory T cells become activated, they proliferate and produce effector molecules at faster rates and in greater magnitudes than antigen-inexperienced, naive cells. Similarly, memory B cells become activated and differentiate into antibody-secreting cells more rapidly than naive B cells, and they undergo processes that increase their affinity for antigen. The ability of T cells and B cells to form memory cells after antigen exposure is the rationale behind vaccination. Understanding immune memory not only is crucial for the design of more-efficacious vaccines but also has important implications for immunotherapies in infectious disease and cancer. This 'guide to' article provides an overview of the current understanding of the phenotype, function, location, and pathways for the generation, maintenance and protective capacity of memory T cells and memory B cells.
Collapse
Affiliation(s)
- Nora Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - YoonSeung Lee
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
37
|
Montiel-Armendariz A, Roe K, Lagos-Orellana J, MartinezCastro LV, Lacy-Hulbert A, Acharya M. B cell αv integrin regulates germinal center derived lung-resident IgA B cell responses following influenza virus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587969. [PMID: 39386536 PMCID: PMC11463618 DOI: 10.1101/2024.04.03.587969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Emerging studies have highlighted the importance of tissue-resident B cells in the lungs, for protective immunity against respiratory viruses. However, the mechanisms controlling generation and maintenance of such tissue-resident B cells at respiratory sites remain obscure. We have previously shown that αv integrins limit B cell responses to antigens containing Toll-like receptor ligands, and that deletion of B cell αv integrins, in mice, enhances germinal center (GC)-derived long-lived B cell responses after systemic immunization with viral antigens. Here we investigated whether αv also regulates B cell responses at the respiratory tract during viral infection. Our data show that αv integrin restricts tissue-resident B cell responses in the airway, and that deletion of B cell αv promotes generation of lung-resident IgA B cell responses following influenza A virus (IAV) infection. Investigating the mechanism for this, we found that loss of B cell αv, promotes persistence of GC reactions locally in the lungs, which leads to increases in lung-resident IgA+ memory B cells, cross-reactive to antigenic variants. Thus, these studies reveal how IgA B cells are maintained in the lungs and point to a new strategy to improve the durability of lung-resident IgA B cell responses for IAV vaccine efficacy.
Collapse
Affiliation(s)
| | - Kelsey Roe
- Seattle Children’s Research Institute, Seattle, WA
| | | | | | | | - Mridu Acharya
- Seattle Children’s Research Institute, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
| |
Collapse
|
38
|
Slavny P, Hegde M, Doerner A, Parthiban K, McCafferty J, Zielonka S, Hoet R. Advancements in mammalian display technology for therapeutic antibody development and beyond: current landscape, challenges, and future prospects. Front Immunol 2024; 15:1469329. [PMID: 39381002 PMCID: PMC11459229 DOI: 10.3389/fimmu.2024.1469329] [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: 07/23/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
The evolving development landscape of biotherapeutics and their growing complexity from simple antibodies into bi- and multi-specific molecules necessitates sophisticated discovery and engineering platforms. This review focuses on mammalian display technology as a potential solution to the pressing challenges in biotherapeutic development. We provide a comparative analysis with established methodologies, highlighting key aspects of mammalian display technology, including genetic engineering, construction of display libraries, and its pivotal role in hit selection and/or developability engineering. The review delves into the mechanisms underpinning developability-driven selection via mammalian display and their broader implications. Applications beyond antibody discovery are also explored, alongside advancements towards function-first screening technologies, precision genome engineering and AI/ML-enhanced libraries, situating them in the context of mammalian display. Overall, the review provides a comprehensive overview of the current mammalian display technology landscape, underscores the expansive potential of the technology for biotherapeutic development, addresses the critical challenges for the full realisation of this potential, and examines advances in related disciplines that might impact the future application of mammalian display technologies.
Collapse
Affiliation(s)
- Peter Slavny
- Discovery & Engineering Division, Iontas Ltd./FairJourney Biologics, Cambridge, United Kingdom
| | - Manjunath Hegde
- Technology Division, Iontas/FairJourney Biologics, Cambridge, United Kingdom
| | - Achim Doerner
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Kothai Parthiban
- Discovery & Engineering Division, Iontas Ltd./FairJourney Biologics, Cambridge, United Kingdom
| | - John McCafferty
- Maxion Therapeutics, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Stefan Zielonka
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Rene Hoet
- Technology Division, Iontas/FairJourney Biologics, Cambridge, United Kingdom
- Technology Division, FairJourney Biologics, Porto, Portugal
| |
Collapse
|
39
|
Jiang H, Wang S. Physical extraction of antigen and information. Proc Natl Acad Sci U S A 2024; 121:e2320537121. [PMID: 39302963 PMCID: PMC11441497 DOI: 10.1073/pnas.2320537121] [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: 11/21/2023] [Accepted: 07/18/2024] [Indexed: 09/22/2024] Open
Abstract
To respond and adapt, cells use surface receptors to sense environmental cues. While biochemical signal processing inside the cell is studied in depth, less is known about how physical processes during cell-cell contact impact signal acquisition. New experiments found that fast-evolving immune B cells in germinal centers (GCs) apply force to acquire antigen clusters prior to internalization, suggesting adaptive benefits of physical information extraction. We present a theory of stochastic antigen transfer and show that maximizing information gain via physical extraction can explain the dramatic phenotypic transition from naive to GC B cells-attenuated receptor signaling, enhanced force usage, and decentralized contact architecture. Our model suggests that binding-lifetime measurement and physical extraction serve as complementary modes of antigen recognition, greatly extending the dynamic range of affinity discrimination when combined. This physical-information framework further predicts that the optimal size of receptor clusters decreases as affinity improves, rationalizing the use of a multifocal synaptic pattern seen in GC B cells. By linking extraction dynamics to selection fidelity via discriminatory performance, we propose that cells may physically enhance information acquisition to sustain adaptive evolution.
Collapse
Affiliation(s)
- Hongda Jiang
- Department of Physics and Astronomy, University of California, Los Angeles, CA90095
| | - Shenshen Wang
- Department of Physics and Astronomy, University of California, Los Angeles, CA90095
| |
Collapse
|
40
|
Bhagchandani SH, Yang L, Lam JH, Maiorino L, Ben-Akiva E, Rodrigues KA, Romanov A, Suh H, Aung A, Wu S, Wadhera A, Chakraborty AK, Irvine DJ. Two-dose priming immunization amplifies humoral immunity by synchronizing vaccine delivery with the germinal center response. Sci Immunol 2024; 9:eadl3755. [PMID: 39303017 PMCID: PMC11492009 DOI: 10.1126/sciimmunol.adl3755] [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: 10/14/2023] [Accepted: 08/29/2024] [Indexed: 09/22/2024]
Abstract
Prolonging exposure to subunit vaccines during the primary immune response enhances humoral immunity. Escalating-dose immunization (EDI), administering vaccines every other day in an increasing pattern over 2 weeks, is particularly effective but challenging to implement clinically. Here, using an HIV Env trimer/saponin adjuvant vaccine, we explored simplified EDI regimens and found that a two-shot regimen administering 20% of the vaccine followed by the remaining 80% of the dose 7 days later increased TFH responses 6-fold, antigen-specific germinal center (GC) B cells 10-fold, and serum antibody titers 10-fold compared with bolus immunization. Computational modeling of TFH priming and the GC response suggested that enhanced activation/antigen loading on dendritic cells and increased capture of antigen delivered in the second dose by follicular dendritic cells contribute to these effects, predictions we verified experimentally. These results suggest that a two-shot priming approach can be used to substantially enhance responses to subunit vaccines.
Collapse
Affiliation(s)
- Sachin H Bhagchandani
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Leerang Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Jonathan H Lam
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Laura Maiorino
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Elana Ben-Akiva
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Kristen A Rodrigues
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Anna Romanov
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Heikyung Suh
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Aereas Aung
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Shengwei Wu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Anika Wadhera
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Arup K Chakraborty
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Department of Physics, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Darrell J Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
41
|
Hébert V, Novarino J, Maho-Vaillant M, Perals C, Calbo S, Golinski ML, Martinez F, Joly P, Fazilleau N. The emergence of circulating activated autoreactive desmoglein 3-specific follicular regulatory T cells is associated with long-term efficacy of rituximab in patients with pemphigus vulgaris. Br J Dermatol 2024; 191:605-615. [PMID: 38848544 DOI: 10.1093/bjd/ljae220] [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: 05/16/2023] [Revised: 05/17/2024] [Accepted: 05/18/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Pemphigus vulgaris (PV) is characterized by autoantibodies targeting keratinocyte adhesion proteins desmoglein (Dsg) 1 and 3, and by the human leukocyte antigen (HLA) predisposition allele HLA-DRB1*0402. Treatment using rituximab (RTX) combined with short-term corticosteroids (CS) allows disease control and long-lasting remission. OBJECTIVES The principal aim of this study was to evaluate the impact of RTX on the circulating subpopulations of Dsg3-specific T lymphocytes that specifically regulate B-cell responses: follicular helper T (Tfh) and follicular regulatory T (Tfr) lymphocytes. METHODS Using the HLA-DRB1*0402 tetramer loaded with the Dsg3 immunodominant peptide, we used flow cytometry to analyse the frequency, polarization and activation status of blood Dsg3-specific follicular T-cell populations at baseline, month (M) 6 and long-term follow-up (M60-90) from patients with PV. RESULTS At baseline, we observed a predominance of Tfh1* and Tfh17 subsets and an underrepresentation of the Tfh2 subset among autoreactive Dsg3-specific Tfh cells compared with nonautoreactive Tfh cells. RTX treatment induced a decrease of autoreactive Tfh cells with no effect on their polarization during follow-up. In parallel, we observed the emergence of a Dsg3-specific Tfr subpopulation with a significant overexpression of the surface activation markers PD1, ICOS and CD25 that was not observed at the surface of autoreactive Tfh and nonautoreactive Tfr cells of the same patients with PV. In contrast, very few Dsg3-specific Tfr cells were observed in patients with PV who were treated with CS alone. CONCLUSIONS Here we show that the emergence of circulating autoreactive Dsg3-specific Tfr cells is associated with the long-term efficacy of RTX in patients with PV. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Vivien Hébert
- Department of Dermatology, French Reference Center for Auto Immune Blistering Diseases, Rouen University Hospital, Normandie University, Rouen, France
- Normandie University, UNIROUEN, Inserm, U1234, FOCIS Center of Excellence PAn'THER, Rouen University Hospital, Department of Immunology and Biotherapy, Rouen, France
- INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France
| | - Julien Novarino
- INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France
| | - Maud Maho-Vaillant
- Normandie University, UNIROUEN, Inserm, U1234, FOCIS Center of Excellence PAn'THER, Rouen University Hospital, Department of Immunology and Biotherapy, Rouen, France
| | - Corine Perals
- INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France
| | - Sébastien Calbo
- Normandie University, UNIROUEN, Inserm, U1234, FOCIS Center of Excellence PAn'THER, Rouen University Hospital, Department of Immunology and Biotherapy, Rouen, France
| | - Marie-Laure Golinski
- Normandie University, UNIROUEN, Inserm, U1234, FOCIS Center of Excellence PAn'THER, Rouen University Hospital, Department of Immunology and Biotherapy, Rouen, France
| | - Fanny Martinez
- INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France
| | - Pascal Joly
- Department of Dermatology, French Reference Center for Auto Immune Blistering Diseases, Rouen University Hospital, Normandie University, Rouen, France
- Normandie University, UNIROUEN, Inserm, U1234, FOCIS Center of Excellence PAn'THER, Rouen University Hospital, Department of Immunology and Biotherapy, Rouen, France
| | - Nicolas Fazilleau
- INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France
| |
Collapse
|
42
|
Franco C, Cornejo A, Rodríguez M, García A, Belisario I, Mayora S, Garzaro DJ, Jaspe RC, Hidalgo M, Parra N, Liprandi F, Zambrano JL, Rangel HR, Pujol FH. Sputnik V-Induced Antibodies against SARS-CoV-2 Variants during the Dissemination of the Gamma Variant in Venezuela. Viruses 2024; 16:1480. [PMID: 39339956 PMCID: PMC11437404 DOI: 10.3390/v16091480] [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: 08/23/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
The COVID-19 pandemic was characterized by the emergence and succession of SARS-CoV-2 variants able to evade the antibody response induced by natural infection and vaccination. To evaluate the IgG reactivity and neutralizing capacity of the serum of individuals vaccinated with Sputnik V (105 volunteers vaccinated) against different viral variants. IgG reactivity to the Spike protein (S) was evaluated by ELISA. A plaque reduction neutralization test was performed using different viral variant isolates. At 42 days post-vaccination, the frequency of recognition and reactivity to the S protein of the Omicron variant was lower compared to that of the other variants. In general, a higher average neutralization titer was seen against the ancestral variant compared to the variants, especially Omicron. However, some sera exhibited a higher neutralization titer to the Gamma variant compared to the ancestral variant, suggesting unapparent exposure during the clinical trial. Antibodies induced by Sputnik V can recognize, persist, and neutralize SARS-CoV-2 variants, with Omicron being the one that best evades this response. These results represent a unique report on the humoral response induced by a globally lesser-studied vaccine in terms of efficacy and immune escape, offering insights into developing vaccines targeting unknown coronaviruses.
Collapse
Affiliation(s)
- Christopher Franco
- Laboratorio de Virología Celular, Centro de Microbiología y Biología Celular, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020, Venezuela
| | - Alejandro Cornejo
- Laboratorio de Bioquímica Celular, Centro de Microbiología y Biología Celular, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020, Venezuela
| | - Mariajosé Rodríguez
- Laboratorio de Virología Molecular, Centro de Microbiología y Biología Celular, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020, Venezuela
| | - Alexis García
- Instituto de Inmunología, Universidad Central de Venezuela (UCV), Caracas 1020, Venezuela
| | - Inirida Belisario
- Instituto de Inmunología, Universidad Central de Venezuela (UCV), Caracas 1020, Venezuela
| | - Soriuska Mayora
- Instituto de Inmunología, Universidad Central de Venezuela (UCV), Caracas 1020, Venezuela
| | - Domingo José Garzaro
- Laboratorio de Virología Molecular, Centro de Microbiología y Biología Celular, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020, Venezuela
| | - Rossana Celeste Jaspe
- Laboratorio de Virología Molecular, Centro de Microbiología y Biología Celular, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020, Venezuela
| | - Mariana Hidalgo
- Laboratorio de Inmunoparasitología, Centro de Microbiología y Biología Celular, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020, Venezuela
| | - Nereida Parra
- Laboratorio de Fisiología de Parásitos, Centro Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020, Venezuela
| | - Ferdinando Liprandi
- Laboratorio de Biología de Virus, Centro de Microbiología y Biología Celular, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020, Venezuela
| | - José Luis Zambrano
- Laboratorio de Virología Celular, Centro de Microbiología y Biología Celular, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020, Venezuela
| | - Héctor Rafael Rangel
- Laboratorio de Virología Molecular, Centro de Microbiología y Biología Celular, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020, Venezuela
| | - Flor Helene Pujol
- Laboratorio de Virología Molecular, Centro de Microbiología y Biología Celular, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020, Venezuela
| |
Collapse
|
43
|
Weber LL, Reiman D, Roddur MS, Qi Y, El-Kebir M, Khan AA. Isotype-aware inference of B cell clonal lineage trees from single-cell sequencing data. CELL GENOMICS 2024; 4:100637. [PMID: 39208795 PMCID: PMC11480863 DOI: 10.1016/j.xgen.2024.100637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/19/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) enables comprehensive characterization of the micro-evolutionary processes of B cells during an adaptive immune response, capturing features of somatic hypermutation (SHM) and class switch recombination (CSR). Existing phylogenetic approaches for reconstructing B cell evolution have primarily focused on the SHM process alone. Here, we present tree inference of B cell clonal lineages (TRIBAL), an algorithm designed to optimally reconstruct the evolutionary history of B cell clonal lineages undergoing both SHM and CSR from scRNA-seq data. Through simulations, we demonstrate that TRIBAL produces more comprehensive and accurate B cell lineage trees compared to existing methods. Using real-world datasets, TRIBAL successfully recapitulates expected biological trends in a model affinity maturation system while reconstructing evolutionary histories with more parsimonious class switching than state-of-the-art methods. Thus, TRIBAL significantly improves B cell lineage tracing, useful for modeling vaccine responses, disease progression, and the identification of therapeutic antibodies.
Collapse
Affiliation(s)
- Leah L Weber
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Derek Reiman
- Toyota Technological Institute at Chicago, Chicago, IL 60637, USA
| | - Mrinmoy S Roddur
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yuanyuan Qi
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Mohammed El-Kebir
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Aly A Khan
- Toyota Technological Institute at Chicago, Chicago, IL 60637, USA; Department of Pathology, University of Chicago, Chicago, IL 60637, USA; Chan Zuckerberg Biohub Chicago, Chicago, IL 60642, USA.
| |
Collapse
|
44
|
Seefeld ML, Templeton EL, Lehtinen JM, Sinclair N, Yadav D, Hartwell BL. Harnessing the potential of the NALT and BALT as targets for immunomodulation using engineering strategies to enhance mucosal uptake. Front Immunol 2024; 15:1419527. [PMID: 39286244 PMCID: PMC11403286 DOI: 10.3389/fimmu.2024.1419527] [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: 04/18/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Mucosal barrier tissues and their mucosal associated lymphoid tissues (MALT) are attractive targets for vaccines and immunotherapies due to their roles in both priming and regulating adaptive immune responses. The upper and lower respiratory mucosae, in particular, possess unique properties: a vast surface area responsible for frontline protection against inhaled pathogens but also simultaneous tight regulation of homeostasis against a continuous backdrop of non-pathogenic antigen exposure. Within the upper and lower respiratory tract, the nasal and bronchial associated lymphoid tissues (NALT and BALT, respectively) are key sites where antigen-specific immune responses are orchestrated against inhaled antigens, serving as critical training grounds for adaptive immunity. Many infectious diseases are transmitted via respiratory mucosal sites, highlighting the need for vaccines that can activate resident frontline immune protection in these tissues to block infection. While traditional parenteral vaccines that are injected tend to elicit weak immunity in mucosal tissues, mucosal vaccines (i.e., that are administered intranasally) are capable of eliciting both systemic and mucosal immunity in tandem by initiating immune responses in the MALT. In contrast, administering antigen to mucosal tissues in the absence of adjuvant or costimulatory signals can instead induce antigen-specific tolerance by exploiting regulatory mechanisms inherent to MALT, holding potential for mucosal immunotherapies to treat autoimmunity. Yet despite being well motivated by mucosal biology, development of both mucosal subunit vaccines and immunotherapies has historically been plagued by poor drug delivery across mucosal barriers, resulting in weak efficacy, short-lived responses, and to-date a lack of clinical translation. Development of engineering strategies that can overcome barriers to mucosal delivery are thus critical for translation of mucosal subunit vaccines and immunotherapies. This review covers engineering strategies to enhance mucosal uptake via active targeting and passive transport mechanisms, with a parallel focus on mechanisms of immune activation and regulation in the respiratory mucosa. By combining engineering strategies for enhanced mucosal delivery with a better understanding of immune mechanisms in the NALT and BALT, we hope to illustrate the potential of these mucosal sites as targets for immunomodulation.
Collapse
Affiliation(s)
- Madison L Seefeld
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Erin L Templeton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Justin M Lehtinen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Noah Sinclair
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Daman Yadav
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Brittany L Hartwell
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
45
|
Mohammad Piri S, Amin Habibi M, Shool S, Khazaeli Najafabadi M, Ahmadpour S, Alemi F, Aria Nejadghaderi S, Shokri P, Abdi M, Asghari N, Amir Asef-Agah S, Tavakolpour S. Role of T follicular helper cells in autoimmune rheumatic Diseases: A systematic review on immunopathogenesis and response to treatment. Hum Immunol 2024; 85:110838. [PMID: 38970880 DOI: 10.1016/j.humimm.2024.110838] [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: 02/07/2024] [Revised: 06/09/2024] [Accepted: 06/28/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND T follicular helper (Tfh) cells are a subdivision of T helper cells involved in antigen-specific B cell immunity. Tfh cells play an essential role in the interaction of T cells/B cells in the germinal centers (GC), and dysregulation of Tfh actions can offer pathogenic autoantibody formation and lead to the development of autoimmune diseases. This study seeks to evaluate changes in Tfh frequency and its related cytokines in autoimmune disease, its association with disease phase, severity, prognosis, and the effect of immunosuppressive treatment on the Tfh population. METHOD The study adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 Statement. Electronic databases, including PubMed, Scopus, Web of Science, and Embase, were systematically searched for potentially eligible studies up to January 1, 2024. RESULTS We identified 4998 articles in the initial search, from which 1686 similar titles were removed. A total of 3312 articles were initially screened, and 3051 articles were excluded by title/abstract screening. A total of 261 studies were considered for full-text assessment, and 205 articles were excluded by reason. Finally, a total of 56 studies were included in our review. CONCLUSION The population of Tfh cells is generally higher in autoimmune diseases versus Health control. Moreover, the number of Tfh cells is associated with the disease severity and can be considered for determining the prognosis of studies. Also, peripheral blood circulating Tfh (cTfh) cells are an available sample that can be used as an indicator for diagnosing diseases.
Collapse
Affiliation(s)
- Seyed Mohammad Piri
- Department of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sina Shool
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Sajjad Ahmadpour
- Patient Safety Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Fakhroddin Alemi
- School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Seyed Aria Nejadghaderi
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Pourya Shokri
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohtaram Abdi
- Student Research Committe, Faculty of Medicine, North Khorasan University of Medical Sciences, Bonjnurd, Iran.
| | - Negin Asghari
- Student Research Committe, Faculty of Medicine, North Khorasan University of Medical Sciences, Bonjnurd, Iran.
| | - Seyed Amir Asef-Agah
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | |
Collapse
|
46
|
Wang Y, Wang Q, He F, Qiao N, Li X, Wei L, Sun L, Dai W, Li Y, Pang X, Hu J, Huang C, Yang G, Pang C, Hu Z, Xing M, Wan C, Zhou D. Age-dependent decrease of circulating T follicular helper cells correlates with disease severity in elderly patients with COVID-19. Clin Immunol 2024; 266:110329. [PMID: 39067679 DOI: 10.1016/j.clim.2024.110329] [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: 04/29/2024] [Revised: 07/04/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Overwhelming evidence has shown that aging is a significant risk factor for COVID-19-related hospitalizations, death and other adverse health outcomes. Particular T cell subsets that susceptible to aging and associated with COVID-19 disease severity requires further elucidation. Our study recruited 57 elderly patients with acute COVID-19 and 27 convalescent donors. Adaptive immunity was assessed across the COVID-19 severity spectrum. Patients underwent age-dependent CD4+ T lymphopenia, preferential loss of circulating T follicular regulatory cells (cTfh) subsets including cTfh-em, cTfh-cm, cTfh1, cTfh2, cTfh17 and circulating T follicular regulatory cells (cTfr), which regulated antibody production through different pathways and correlated with COVID-19 severity, were observed. Moreover, vaccination improved cTfh-cm, cTfh2, cTfr proportion and promoted NAb production. In conclusion, the elderly had gone through age-dependent cTfh subsets deficiency, which impeded NAb production and enabled aggravation of COVID-19 to critical illness, whereas SARS-CoV-2 vaccine inoculation helped to rejuvenate cTfh, cTfr and intensify NAb responses.
Collapse
Affiliation(s)
- Yihan Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Qiu Wang
- Department of Physical and Rehabilitation Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China
| | - Furong He
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Nan Qiao
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Xuejun Li
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Liqun Wei
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Lingjin Sun
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Weiqian Dai
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Ying Li
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Xueyang Pang
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Jiayi Hu
- Department of Clinical Medicine, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Chuan Huang
- Department of Physical and Rehabilitation Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China
| | - Guangchen Yang
- Department of Physical and Rehabilitation Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China
| | - Chongjie Pang
- Department of Infectious Diseases, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China
| | - Zhidong Hu
- Department of Clinical Laboratory, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China
| | - Man Xing
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China.
| | - Chunxiao Wan
- Department of Physical and Rehabilitation Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China.
| | - Dongming Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China; Shanghai Public Health Clinical Center, Fudan University, No. 2901 Caolang Road, Shanghai 201508, China.
| |
Collapse
|
47
|
Novak J, Reily C, Steers NJ, Schumann T, Rizk DV, Julian BA, Kiryluk K, Gharavi AG, Green TJ. Emerging Biochemical and Immunologic Mechanisms in the Pathogenesis of IgA Nephropathy. Semin Nephrol 2024; 44:151565. [PMID: 40087124 PMCID: PMC11972156 DOI: 10.1016/j.semnephrol.2025.151565] [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: 03/16/2025]
Abstract
IgA nephropathy is a mesangioproliferative glomerular disease with significant morbidity and mortality. Most patients with IgA nephropathy develop kidney failure in their lifetime, reducing their life expectancy by a decade. Since its first description in 1968, it has been established that kidneys of IgA nephropathy patients are injured as "innocent bystanders" by nephritogenic IgA1-containing immune complexes. Results from clinical, biochemical, immunologic, and genetic studies suggest a multistep pathogenetic mechanism. In genetically predisposed individuals, this process results in formation of circulating immune complexes due to the binding of IgG/IgA autoantibodies to the polymeric IgA1 molecules with incomplete O-glycosylation. This event is followed by the addition of other proteins, such as complement C3, resulting in the formation of nephritogenic immune complexes. These complexes are not effectively removed from the circulation, and some of them pass through the fenestration of glomerular endothelial cells to enter the mesangial space and activate mesangial cells. It is thought that the process is initiated by soluble immune complexes and that their accumulation results in the formation of immunodeposits that further amplify glomerular injury. Here we summarize current understanding of the pathogenesis of IgA nephropathy and discuss experimental model systems that can inform development of new therapeutic strategies and targets.
Collapse
Affiliation(s)
- Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL.
| | - Colin Reily
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL; Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Nicholas J Steers
- Division of Nephrology, Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | | | - Dana V Rizk
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Bruce A Julian
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Ali G Gharavi
- Division of Nephrology, Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Todd J Green
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
48
|
Hamada R, Yonezawa A, Matsumoto K, Mitani T, Takagi T, Muto A, Igarashi K, Naito Y, Higashimura Y. BTB and CNC homology 1 deficiency disrupts intestinal IgA secretion through regulation of polymeric immunoglobulin receptor expression. Am J Physiol Gastrointest Liver Physiol 2024; 327:G414-G423. [PMID: 38981617 DOI: 10.1152/ajpgi.00215.2023] [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: 09/26/2023] [Revised: 06/11/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
Immunoglobulin A (IgA)-mediated mucosal immunity is important for the host because it contributes to reducing infection risk and to establishing host-microbe symbiosis. BTB and CNC homology 1 (Bach1) is a transcriptional repressor with physiological and pathophysiological functions that are of particular interest for their relation to gastrointestinal diseases. However, Bach1 effects on IgA-mediated mucosal immunity remain unknown. For this study using Bach1-deficient (Bach1-/-) mice, we investigated the function of Bach1 in IgA-mediated mucosal immunity. Intestinal mucosa, feces, and plasma IgA were examined using immunosorbent assay. After cell suspensions were prepared from Peyer's patches and colonic lamina propria, they were examined using flow cytometry. The expression level of polymeric immunoglobulin receptor (pIgR), which plays an important role in the transepithelial transport of IgA, was evaluated using Western blotting, quantitative real-time PCR, and immunohistochemistry. Although no changes in the proportions of IgA-producing cells were observed, the amounts of IgA in the intestinal mucosa were increased in Bach1-/- mice. Furthermore, plasma IgA was increased in Bach1-/- mice, but fecal IgA was decreased, indicating that Bach1-/- mice have abnormal secretion of IgA into the intestinal lumen. In fact, Bach1 deficiency reduced pIgR expression in colonic mucosa at both the protein and mRNA levels. In the human intestinal epithelial cell line LS174T, suppression of Bach1 reduced pIgR mRNA stability. In contrast, the overexpression of Bach1 increased pIgR mRNA stability. These results demonstrate that Bach1 deficiency causes abnormal secretion of IgA into the intestinal lumen via suppression of pIgR expression.NEW & NOTEWORTHY The transcriptional repressor Bach1 has been implicated in diverse intestinal functions, but the effects of Bach1 on IgA-mediated mucosal immunity remain unclear. We demonstrate here that Bach1 deficiency causes abnormal secretion of IgA into the intestinal lumen, although the proportions of IgA-producing cells were not altered. Furthermore, Bach1 regulates the expression of pIgR, which plays an important role in the transepithelial transport of IgA, at the posttranscriptional level.
Collapse
Affiliation(s)
- Riku Hamada
- Department of Food Science, Ishikawa Prefectural University, Nonoichi, Japan
| | - Akari Yonezawa
- Department of Food Science, Ishikawa Prefectural University, Nonoichi, Japan
| | - Kenji Matsumoto
- Department of Food Science, Ishikawa Prefectural University, Nonoichi, Japan
| | - Takakazu Mitani
- Division of Food Science and Biotechnology, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Tomohisa Takagi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akihiko Muto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuki Higashimura
- Department of Food Science, Ishikawa Prefectural University, Nonoichi, Japan
| |
Collapse
|
49
|
Rodrigues KA, Zhang YJ, Aung A, Morgan DM, Maiorino L, Yousefpour P, Gibson G, Ozorowski G, Gregory JR, Amlashi P, Buckley M, Ward AB, Schief WR, Love JC, Irvine DJ. Vaccines combining slow delivery and follicle targeting of antigens increase germinal center B cell clonal diversity and clonal expansion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608655. [PMID: 39229011 PMCID: PMC11370361 DOI: 10.1101/2024.08.19.608655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Vaccines incorporating slow delivery, multivalent antigen display, or immunomodulation through adjuvants have an important role to play in shaping the humoral immune response. Here we analyzed mechanisms of action of a clinically relevant combination adjuvant strategy, where phosphoserine (pSer)-tagged immunogens bound to aluminum hydroxide (alum) adjuvant (promoting prolonged antigen delivery to draining lymph nodes) are combined with a potent saponin nanoparticle adjuvant termed SMNP (which alters lymph flow and antigen entry into lymph nodes). When employed with a stabilized HIV Env trimer antigen in mice, this combined adjuvant approach promoted substantial enhancements in germinal center (GC) and antibody responses relative to either adjuvant alone. Using scRNA-seq and scBCR-seq, we found that the alum-pSer/SMNP combination both increased the diversity of GC B cell clones and increased GC B cell clonal expansion, coincident with increases in the expression of Myc and the proportion of S-phase GC B cells. To gain insight into the source of these changes in the GC response, we analyzed antigen biodistribution and structural integrity in draining lymph nodes and found that the combination adjuvant approach, but not alum-pSer delivery or SMNP alone, promoted accumulation of highly intact antigen on follicular dendritic cells, reflecting an integration of the slow antigen delivery and altered lymph node uptake effects of these two adjuvants. These results demonstrate how adjuvants with complementary mechanisms of action impacting vaccine biodistribution and kinetics can synergize to enhance humoral immunity.
Collapse
Affiliation(s)
- Kristen A. Rodrigues
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Harvard-MIT Health Sciences and Technology Program, Institute for Medical Engineering and Science; Massachusetts Institute of Technology, Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Yiming J. Zhang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
| | - Aereas Aung
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
| | - Duncan M. Morgan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Department of Chemical Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
| | - Laura Maiorino
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Parisa Yousefpour
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Grace Gibson
- Department of Integrative, Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Gabriel Ozorowski
- Department of Integrative, Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Justin R. Gregory
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Parastoo Amlashi
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - Maureen Buckley
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
| | - Andrew B. Ward
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
- Department of Integrative, Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA 92037 USA
| | - William R. Schief
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - J. Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Department of Chemical Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
| | - Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University; Cambridge, MA 02139 USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute; La Jolla, CA 92037 USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139 USA
- Howard Hughes Medical Institute; Chevy Chase, MD 20815 USA
| |
Collapse
|
50
|
Kim W. Germinal Center Response to mRNA Vaccination and Impact of Immunological Imprinting on Subsequent Vaccination. Immune Netw 2024; 24:e28. [PMID: 39246619 PMCID: PMC11377948 DOI: 10.4110/in.2024.24.e28] [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: 01/30/2024] [Revised: 04/04/2024] [Accepted: 04/29/2024] [Indexed: 09/10/2024] Open
Abstract
Vaccines are the most effective intervention currently available, offering protective immunity against targeted pathogens. The emergence of the coronavirus disease 2019 pandemic has prompted rapid development and deployment of lipid nanoparticle encapsulated, mRNA-based vaccines. While these vaccines have demonstrated remarkable immunogenicity, concerns persist regarding their ability to confer durable protective immunity to continuously evolving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. This review focuses on human B cell responses induced by SARS-CoV-2 mRNA vaccination, with particular emphasis on the crucial role of germinal center reactions in shaping enduring protective immunity. Additionally, we explored observations of immunological imprinting and dynamics of recalled pre-existing immunity following variants of concern-based booster vaccination. Insights from this review contribute to comprehensive understanding B cell responses to mRNA vaccination in humans, thereby refining vaccination strategies for optimal and sustained protection against evolving coronavirus variants.
Collapse
Affiliation(s)
- Wooseob Kim
- Department of Microbiology, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| |
Collapse
|