1
|
Zhang N, Qin X, Liu J, Han K, Kang M, Zhu Z, Zhang D, Zhong F. Pan-cancer analysis and validation show GTF2E2's diagnostic, prognostic, and immunological roles in regulating ferroptosis in endometrial cancer. PLoS One 2025; 20:e0321983. [PMID: 40267151 PMCID: PMC12017540 DOI: 10.1371/journal.pone.0321983] [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: 12/18/2024] [Accepted: 03/11/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Transcription initiation factor IIE subunit beta (GTF2E2) is a crucial component of the RNA polymerase II transcription initiation complex. There is a lack of more detailed research on the biological function of GTF2E2 in pan-cancer. METHODS We conducted a comprehensive pan-cancer analysis using data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) project. Employing a multi-pronged approach with tools including R, Cytoscape, TISIDB, cBioPortal, STRING, GSCALite, and CancerSEA, we investigated GTF2E2's expression patterns, prognostic value, mutational landscape, functional enrichment, and immunological associations across 33 cancer types. Besides, we further validated the bioinformatic results through in vitro experiments in Uterine corpus endometrial carcinoma (UCEC), including western blotting (WB), cell proliferation assays and transwell. DCFH-DA, C11-BODIPY 581/591 and FeRhoNox-1 probes were performed to identify ferroptosis levels in vitro. RESULTS GTF2E2 expression was significantly elevated in most cancers compared to normal tissues, with notable diagnostic potential (AUC > 0.7) in 20 cancer types. GTF2E2 expression varied across molecular and immune subtypes and correlated with tumor stage and patient age in several cancers. Functional enrichment analyses highlighted GTF2E2's involvement in key cancer-related and immunological pathways. Notably, GTF2E2 promoted UCEC progression in vitro, and knockdown of GTF2E2 significantly inhibited the proliferation, migration and invasion of UCEC cells. Compared with the control group, GPX4 expression was down-regulated and ACSL4 expression was up-regulated in the GTF2E2-knockdown group. Knockdown of GTF2E2 also increased the intracellular levels of Fe2+, lipid peroxides (LPOs) and reactive oxygen species (ROS). CONCLUSIONS Our findings underscore GTF2E2's multifaceted roles in cancer biology, highlighting its potential as a diagnostic biomarker, prognostic indicator, and immunotherapeutic target across various malignancies. This investigation has the potential to contribute significantly to a deeper understanding of the substantial involvement of GTF2E2 in human malignancies, particularly UCEC.
Collapse
Affiliation(s)
- Nie Zhang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
- Graduate School of Anhui Medical University, Anhui, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Anhui, China
| | - Xuejin Qin
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Jingjing Liu
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Ke Han
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Manman Kang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Zhengchun Zhu
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Di Zhang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Fei Zhong
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
- Graduate School of Anhui Medical University, Anhui, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Anhui, China
| |
Collapse
|
2
|
Giri S, Batra L. Memory Cells in Infection and Autoimmunity: Mechanisms, Functions, and Therapeutic Implications. Vaccines (Basel) 2025; 13:205. [PMID: 40006751 PMCID: PMC11860616 DOI: 10.3390/vaccines13020205] [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: 01/08/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Memory cells are central to the adaptive immune system's ability to remember and respond effectively to previously encountered pathogens. While memory cells provide robust protection against infections, they can also contribute to autoimmunity when regulation fails. Here, we review the roles of memory T and B cells in infection and autoimmunity, focusing on their differentiation, activation, effector functions, and underlying regulatory mechanisms. We elaborate on the precise mechanisms by which memory cells contribute to autoimmune diseases, highlighting insights from current research on how pathogenic memory responses are formed and sustained in autoimmunity. Finally, we explore potential therapeutic strategies aimed at modulating memory cells to prevent or treat autoimmune disorders, including B cell-depleting therapies (e.g., Rituximab), T cell-targeting agents (e.g., Abatacept), and cytokine inhibitors (e.g., IL-17 or IL-23 blockers) that are currently used in diseases such as rheumatoid arthritis, multiple sclerosis, and psoriasis.
Collapse
Affiliation(s)
- Shilpi Giri
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lalit Batra
- Center for Predictive Medicine for Biodefence and Emerging Infectious Diseases, School of Medicine, University of Louisville, Louisville, KY 40222, USA;
| |
Collapse
|
3
|
Samiea A, Celis G, Yadav R, Rodda LB, Moreau JM. B cells in non-lymphoid tissues. Nat Rev Immunol 2025:10.1038/s41577-025-01137-6. [PMID: 39910240 DOI: 10.1038/s41577-025-01137-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
B cells have long been understood to be drivers of both humoral and cellular immunity. Recent advances underscore this importance but also indicate that in infection, inflammatory disease and cancer, B cells function directly at sites of inflammation and form tissue-resident memory populations. The spatial organization and cellular niches of tissue B cells have profound effects on their function and on disease outcome, as well as on patient response to therapy. Here we review the role of B cells in peripheral tissues in homeostasis and disease, and discuss the newly identified cellular and molecular signals that are involved in regulating their activity. We integrate emerging data from multi-omic human studies with experimental models to propose a framework for B cell function in tissue inflammation and homeostasis.
Collapse
Affiliation(s)
- Abrar Samiea
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - George Celis
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Rashi Yadav
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Lauren B Rodda
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA.
| | - Joshua M Moreau
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA.
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
4
|
Zhu W, Dong C, Wei L, Kim JK, Wang BZ. Inverted HA-EV immunization elicits stalk-specific influenza immunity and cross-protection in mice. Mol Ther 2025; 33:485-498. [PMID: 39741410 PMCID: PMC11852689 DOI: 10.1016/j.ymthe.2024.12.052] [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: 08/20/2024] [Revised: 12/02/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Enhancing protective immunity in the respiratory tract is crucial to combat influenza infection and transmission. Developing mucosal universal influenza vaccines requires effective delivery platforms to overcome the respiratory mucosal barrier and stimulate appropriate innate immune reactions, thereby bridging adaptive immune responses with minimal necessary inflammation. Meanwhile, the vaccine platforms must be biocompatible. This study employed cell-derived extracellular vesicles (EVs) as a mucosal universal influenza vaccine platform. By conjugating influenza hemagglutinin (HA) onto EV surfaces through HA-receptor interaction, we achieved an upside-down (inverted) influenza HA configuration that exposed the conserved HA stalk region while partially hiding the globular head domain. Intranasal immunization with the resulting EVs induced robust HA stalk- and virus-specific serum antibody and mucosal immune responses in mice, protecting against heterologous virus infection. Notably, EVs derived from the lung epithelial cell line A549 induced superior cross-reactive antibodies and enhanced protection upon intranasal immunization. EVs conjugating multivalent HA elicited broadly cross-reactive antibody and cellular responses against different influenza strains. Our results demonstrated that EVs conjugating multiple inverted HAs represented an effective strategy for developing a mucosal universal influenza vaccine.
Collapse
Affiliation(s)
- Wandi Zhu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Joo Kyung Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
5
|
Kirchenbaum GA, Pawelec G, Lehmann PV. The Importance of Monitoring Antigen-Specific Memory B Cells, and How ImmunoSpot Assays Are Suitable for This Task. Cells 2025; 14:223. [PMID: 39937014 PMCID: PMC11816810 DOI: 10.3390/cells14030223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/10/2025] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
Determining an individual's humoral immune reactivity to a pathogen, autoantigen, or environmental agent is traditionally accomplished through the assessment of specific antibody levels in blood. However, in many instances, titers of specific antibodies decline over time and thus do not faithfully reveal prior antigen exposure or establishment of immunological memory. To estimate an individual's humoral immune competence, it is therefore necessary to assess functional B cell memory. Here, we describe novel B cell ELISPOT and FluoroSpot assays (collectively referred to as ImmunoSpot) that can be rapidly developed and validated to characterize the memory B cell (Bmem) repertoire specific for any desired antigen ex vivo and at single-cell resolution. Moreover, multiplexed variants of the B cell FluoroSpot assay enable high-throughput testing of antigen-specific B cells secreting distinct antibody classes and/or IgG subclasses, with minimal cell material requirements. B cell ImmunoSpot assays also enable measurement of affinity distributions within the antigen-specific Bmem compartment and permit cross-reactivity measurements that can provide insights into Bmem established against future pathogen variants. Collectively, the ImmunoSpot® system presented here is highly reproducible, and can be readily validated for regulated tests. The newly gained ability to monitor the antigen-specific Bmem compartment should catalyze a more comprehensive understanding of humoral immunity in health and disease.
Collapse
Affiliation(s)
- Greg A. Kirchenbaum
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA;
| | - Graham Pawelec
- Department of Immunology, University of Tübingen, D-72076 Tübingen, Germany;
- Health Sciences North Research Institute, Sudbury, ON P3E 2H3, Canada
| | - Paul V. Lehmann
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA;
| |
Collapse
|
6
|
Sato Y, Inoue N, Osada E, Tsunemi Y, Nakashima D, Hirota T, Otori N, Yoshikawa M, Haruna SI, Nakayama T. Resident memory B cells are enriched in chronic rhinosinusitis with nasal polyps. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2025; 4:100349. [PMID: 39583038 PMCID: PMC11585886 DOI: 10.1016/j.jacig.2024.100349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/15/2024] [Accepted: 07/20/2024] [Indexed: 11/26/2024]
Abstract
Background Chronic rhinosinusitis with nasal polyps (CRSwNP) is a chronic nasal and sinonasal inflammatory disease. Recently, resident memory B (BRM) cells have been identified in the lungs, although not in the sinonasal mucosa. Objective Our aim was to characterize memory B-cell phenotypes with regard to patients with CRSwNP and identify BRM cells in both normal sinonosal mucosa and samples from patients with CRSwNP. Methods CD19+ B cells were isolated from patients with CRSwNP and analyzed using flow cytometry and immunohistochemistry. Results Although BRM cells were found in the normal sinonasal mucosa, their numbers and frequencies tended to be limited. These findings were confirmed on the basis of immunohistochemical analyses indicating an upregulation of CD69/CD45RB in tissue sections from patients with CRSwNP, although not in normal sinonasal mucosa. Accordingly, BRM cells were established to be enriched in the nasal polyps isolated from patients with CRSwNP. Conclusion Our findings in this study reveal that BRM cells can be detected in normal sinonasal mucosa, although they are significantly enriched in nasal polyps derived from patients with CRSwNP. These findings can contribute to gaining a more comprehensive understanding of the immune reactions associated with CRSwNP and facilitate the identification of potential therapeutic targets, such as anti-B-cell therapy.
Collapse
Affiliation(s)
- Yohei Sato
- Laboratory of Immune Cell Therapy, Project Research Unit, Jikei University School of Medicine, Tokyo, Japan
- Core Research Facilities, Jikei University School of Medicine, Tokyo, Japan
| | - Natsuki Inoue
- Division of Molecular Genetics, Jikei University School of Medicine, Tokyo, Japan
- Department of Otorhinolaryngology, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Erika Osada
- Core Research Facilities, Jikei University School of Medicine, Tokyo, Japan
| | - Yasuhiro Tsunemi
- Department of Otorhinolaryngology, Head, and Neck Surgery, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Daiki Nakashima
- Division of Molecular Genetics, Jikei University School of Medicine, Tokyo, Japan
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Tomomitsu Hirota
- Division of Molecular Genetics, Jikei University School of Medicine, Tokyo, Japan
| | - Nobuyoshi Otori
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Mamoru Yoshikawa
- Department of Otorhinolaryngology, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Shin-ichi Haruna
- Department of Otorhinolaryngology, Head, and Neck Surgery, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Tsuguhisa Nakayama
- Department of Otorhinolaryngology, Head, and Neck Surgery, Dokkyo Medical University School of Medicine, Tochigi, Japan
| |
Collapse
|
7
|
Lin Y, Liao X, Cao X, Zhang Z, Wang X, He X, Liao H, Ju B, Qi F, Xu H, Ren Z, Wang Y, Hu Z, Yang J, Fu YX, Zhao J, Zhang Z, Peng H. Sequential intranasal booster triggers class switching from intramuscularly primed IgG to mucosal IgA against SARS-CoV-2. J Clin Invest 2025; 135:e175233. [PMID: 39808503 PMCID: PMC11870729 DOI: 10.1172/jci175233] [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: 08/29/2023] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
The persistent emergence of COVID-19 variants and recurrent waves of infection worldwide underscores the urgent need for vaccines that effectively reduce viral transmission and prevent infections. Current intramuscular (IM) COVID-19 vaccines inadequately protect the upper respiratory mucosa. In response, we have developed a nonadjuvanted, IFN-armed SARS-CoV-2 fusion protein vaccine with IM priming and intranasal (IN) boost sequential immunization. Our study showed that this sequential vaccination strategy of the IM+IN significantly enhanced both upper respiratory and systemic antiviral immunity in a mouse model, characterized by the rapid increase in systemic and mucosal T and B cell responses, particularly the mucosal IgA antibody response. The IN boost triggered a swift secondary immune response, rapidly inducing antigen-specific IgA+ B cells. Further B cell receptor-seq (BCR-seq) analysis indicated that these IgA+ B cells primarily arose through direct class switching from preexisting IgG+ B cells in draining lymph nodes. Notably, our clinical studies revealed that the IN boost after IM vaccination elicited a robust systemic IgA antibody response in humans, as measured in serum. Thus, we believe that our cytokine-armed protein vaccine presents a promising strategy for inducing rapid and potent mucosal protection against respiratory viral infections.
Collapse
Affiliation(s)
- Yifan Lin
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xuejiao Liao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xuezhi Cao
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Zhaoyong Zhang
- 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, China
| | - Xiuye Wang
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Xiaomeng He
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | | | - Bin Ju
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Furong Qi
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hairong Xu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | - Yanqun Wang
- 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, China
| | | | | | - Yang-Xin Fu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Jincun Zhao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hua Peng
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Prosser AC, Klenerman P, Lucas M. Understanding Liver Transplantation Outcomes Through the Lens of Its Tissue-resident Immunobiome. Transplantation 2025:00007890-990000000-00973. [PMID: 39780303 DOI: 10.1097/tp.0000000000005303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Tissue-resident lymphocytes (TRLs) provide a front-line immunological defense mechanism uniquely placed to detect perturbations in tissue homeostasis. The heterogeneous TRL population spans the innate to adaptive immune continuum, with roles during normal physiology in homeostatic maintenance, tissue repair, pathogen detection, and rapid mounting of immune responses. TRLs are especially enriched in the liver, with every TRL subset represented, including liver-resident natural killer cells; tissue-resident memory B cells; conventional tissue-resident memory CD8, CD4, and regulatory T cells; and unconventional gamma-delta, natural killer, and mucosal-associated invariant T cells. The importance of donor- and recipient-derived TRLs after transplantation is becoming increasingly recognized, although it has not been examined in detail after liver transplantation. This review summarizes the evidence for the roles of TRLs in liver transplant immunology, focusing on their features, functions, and potential for their harnessing to improve transplant outcomes.
Collapse
Affiliation(s)
- Amy C Prosser
- Medical School, University of Western Australia, Perth, WA, Australia
| | - Paul Klenerman
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Michaela Lucas
- Medical School, University of Western Australia, Perth, WA, Australia
- Department of Immunology, PathWest Laboratory Medicine, Perth, WA, Australia
- Department of Immunology, Sir Charles Gairdner Hospital, Perth, WA, Australia
- Department of Immunology, Perth Children's Hospital, Perth, WA, Australia
| |
Collapse
|
9
|
Bai Z, Wan D, Lan T, Hong W, Dong H, Wei Y, Wei X. Nanoplatform Based Intranasal Vaccines: Current Progress and Clinical Challenges. ACS NANO 2024; 18:24650-24681. [PMID: 39185745 PMCID: PMC11394369 DOI: 10.1021/acsnano.3c10797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 08/27/2024]
Abstract
Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
Collapse
Affiliation(s)
| | | | | | - Weiqi Hong
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Haohao Dong
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Yuquan Wei
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Xiawei Wei
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
10
|
Shamseldin MM, Read KA, Hall JM, Tuazon JA, Brown JM, Guo M, Gupta YA, Deora R, Oestreich KJ, Dubey P. The adjuvant BcfA activates antigen presenting cells through TLR4 and supports T FH and T H1 while attenuating T H2 gene programming. Front Immunol 2024; 15:1439418. [PMID: 39267766 PMCID: PMC11390363 DOI: 10.3389/fimmu.2024.1439418] [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: 05/27/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024] Open
Abstract
Introduction Adjuvants added to subunit vaccines augment antigen-specific immune responses. One mechanism of adjuvant action is activation of pattern recognition receptors (PRRs) on innate immune cells. Bordetella colonization factor A (BcfA); an outer membrane protein with adjuvant function, activates TH1/TH17-polarized immune responses to protein antigens from Bordetella pertussis and SARS CoV-2. Unlike other adjuvants, BcfA does not elicit a TH2 response. Methods To understand the mechanism of BcfA-driven TH1/TH17 vs. TH2 activation, we screened PRRs to identify pathways activated by BcfA. We then tested the role of this receptor in the BcfA-mediated activation of bone marrow-derived dendritic cells (BMDCs) using mice with germline deletion of TLR4 to quantify upregulation of costimulatory molecule expression and cytokine production in vitro and in vivo. Activity was also tested on human PBMCs. Results PRR screening showed that BcfA activates antigen presenting cells through murine TLR4. BcfA-treated WT BMDCs upregulated expression of the costimulatory molecules CD40, CD80, and CD86 and produced IL-6, IL-12/23 p40, and TNF-α while TLR4 KO BMDCs were not activated. Furthermore, human PBMCs stimulated with BcfA produced IL-6. BcfA-stimulated murine BMDCs also exhibited increased uptake of the antigen DQ-OVA, supporting a role for BcfA in improving antigen presentation to T cells. BcfA further activated APCs in murine lungs. Using an in vitro TH cell polarization system, we found that BcfA-stimulated BMDC supernatant supported TFH and TH1 while suppressing TH2 gene programming. Conclusions Overall, these data provide mechanistic understanding of how this novel adjuvant activates immune responses.
Collapse
Affiliation(s)
- Mohamed M. Shamseldin
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Departments of Microbiology, The Ohio State University, Columbus, OH, United States
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University-Ain Helwan, Helwan, Egypt
| | - Kaitlin A. Read
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Jesse M. Hall
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Jasmine A. Tuazon
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Jessica M. Brown
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Myra Guo
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Yash A. Gupta
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Rajendar Deora
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Departments of Microbiology, The Ohio State University, Columbus, OH, United States
| | - Kenneth J. Oestreich
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States
| | - Purnima Dubey
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
11
|
Allam M, Hu T, Fang Z, Pi M, Singh A, Coskun AF. Spatial immunophenotyping using multiplexed imaging of immune follicles in secondary lymphoid tissues. PNAS NEXUS 2024; 3:pgae285. [PMID: 39108301 PMCID: PMC11299982 DOI: 10.1093/pnasnexus/pgae285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 07/09/2024] [Indexed: 08/10/2024]
Abstract
Secondary lymphoid organs (SLOs), including tonsils (TS), lymph nodes (LN), and Peyer's Patches, exhibit complementary immune functions. However, little is known about the spatial organization of immune cells and extracellular matrix (ECM) in the SLOs. Traditional imaging is limited to a few markers, confining our understanding of the differences between the SLOs. Herein, imaging mass cytometry addressed this gap by simultaneously profiling 25-plex proteins in SLO tissues at subcellular resolution. The antibody panel targeted immune, stromal, chemokine, epigenetic, and functional markers. For robust cell identification, a computational workflow SpatialVizPheno was developed to spatially phenotype 999,970 cells using two approaches, including manual gating and semi-supervised gating, iterative clustering, and annotation. LN exhibited the highest density of B cells while the intestinal tissues contained the highest proportion of regulatory and follicular helper T cells. SpatialVizPheno identified the most prevalent interaction between follicular dendritic cells and stromal cells (SCs), plasmablasts/plasma cells, and the SCs across the lymphoid tissues. Collagen-enriched regions were associated with the spatial orientation of B cell follicles in both TS and LN tissues, but not in intestinal lymphoid tissues. Such spatial differences of immunophenotypes and ECM in different SLO tissues can be used to quantify the relationship between cellular organization and ultimate immune responses.
Collapse
Affiliation(s)
- Mayar Allam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Thomas Hu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zhou Fang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Michelle Pi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ankur Singh
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30318, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
12
|
Zheng HY, Song TZ, Zheng YT. Immunobiology of COVID-19: Mechanistic and therapeutic insights from animal models. Zool Res 2024; 45:747-766. [PMID: 38894519 PMCID: PMC11298684 DOI: 10.24272/j.issn.2095-8137.2024.062] [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: 02/23/2024] [Accepted: 04/22/2024] [Indexed: 06/21/2024] Open
Abstract
The distribution of the immune system throughout the body complicates in vitro assessments of coronavirus disease 2019 (COVID-19) immunobiology, often resulting in a lack of reproducibility when extrapolated to the whole organism. Consequently, developing animal models is imperative for a comprehensive understanding of the pathology and immunology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This review summarizes current progress related to COVID-19 animal models, including non-human primates (NHPs), mice, and hamsters, with a focus on their roles in exploring the mechanisms of immunopathology, immune protection, and long-term effects of SARS-CoV-2 infection, as well as their application in immunoprevention and immunotherapy of SARS-CoV-2 infection. Differences among these animal models and their specific applications are also highlighted, as no single model can fully encapsulate all aspects of COVID-19. To effectively address the challenges posed by COVID-19, it is essential to select appropriate animal models that can accurately replicate both fatal and non-fatal infections with varying courses and severities. Optimizing animal model libraries and associated research tools is key to resolving the global COVID-19 pandemic, serving as a robust resource for future emerging infectious diseases.
Collapse
Affiliation(s)
- Hong-Yi Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Tian-Zhang Song
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Yong-Tang Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China. E-mail:
| |
Collapse
|
13
|
MacLean AJ, Bonifacio JP, Oram SL, Mohsen MO, Bachmann MF, Arnon TI. Regulation of pulmonary plasma cell responses during secondary infection with influenza virus. J Exp Med 2024; 221:e20232014. [PMID: 38661717 PMCID: PMC11044945 DOI: 10.1084/jem.20232014] [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: 11/02/2023] [Revised: 03/05/2024] [Accepted: 04/01/2024] [Indexed: 04/26/2024] Open
Abstract
During secondary infection with influenza virus, plasma cells (PCs) develop within the lung, providing a local source of antibodies. However, the site and mechanisms that regulate this process are poorly defined. Here, we show that while circulating memory B cells entered the lung during rechallenge and were activated within inducible bronchus-associated lymphoid tissues (iBALTs), resident memory B (BRM) cells responded earlier, and their activation occurred in a different niche: directly near infected alveoli. This process required NK cells but was largely independent of CD4 and CD8 T cells. Innate stimuli induced by virus-like particles containing ssRNA triggered BRM cell differentiation in the absence of cognate antigen, suggesting a low threshold of activation. In contrast, expansion of PCs in iBALTs took longer to develop and was critically dependent on CD4 T cells. Our work demonstrates that spatially distinct mechanisms evolved to support pulmonary secondary PC responses, and it reveals a specialized function for BRM cells as guardians of the alveoli.
Collapse
Affiliation(s)
| | | | - Sophia L. Oram
- University of Oxford, Kennedy Institute of Rheumatology, Oxford, UK
| | - Mona O. Mohsen
- Department of Bio Medical Research, University of Bern, Rheumatology, Immunology and Allergology, Bern, Switzerland
| | - Martin F. Bachmann
- Nuffield Department of Medicine, University of Oxford, The Jenner Institute, Oxford, UK
- Department of Bio Medical Research, University of Bern, Rheumatology, Immunology and Allergology, Bern, Switzerland
| | - Tal I. Arnon
- University of Oxford, Kennedy Institute of Rheumatology, Oxford, UK
| |
Collapse
|
14
|
Torrance BL, Cadar AN, Panier HA, Martin DE, Lorenzo EC, Jellison ER, Bartley JM, Haynes L. Cellular senescence is a double-edged sword in regulating aged immune responses to influenza. Aging Cell 2024; 23:e14162. [PMID: 38689516 PMCID: PMC11258475 DOI: 10.1111/acel.14162] [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: 05/19/2023] [Revised: 03/10/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024] Open
Abstract
Clearance of senescent cells has demonstrated therapeutic potential in the context of chronic age-related diseases. Little is known, however, how clearing senescent cells affects the ability to respond to an acute infection and form quality immunological memory. We aimed to probe the effects of clearing senescent cells in aged mice on the immune response to influenza (flu) infection. We utilized a p16 trimodality reporter mouse model (p16-3MR) to allow for identification and selective clearance of p16-expressing cells upon administration of ganciclovir (GCV). While p16-expressing cells may exacerbate dysfunctional responses to a primary infection, our data suggest they may play a role in fostering memory cell generation. We demonstrate that although clearance of p16-expressing cells enhanced viral clearance, this also severely limited antibody production in the lungs of flu-infected aged mice. 30 days later, there were fewer flu-specific CD8 memory T cells and lower levels of flu-specific antibodies in the lungs of GCV-treated mice. Furthermore, GCV-treated mice were unable to mount an optimal memory response and demonstrated increased viral load following heterosubtypic challenge. These results suggest that targeting senescent cells may potentiate primary responses while limiting the ability to form durable and protective immune memory with age.
Collapse
Affiliation(s)
- Blake L. Torrance
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Andreia N. Cadar
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Hunter A. Panier
- Department of MedicineUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Dominique E. Martin
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Erica C. Lorenzo
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Evan R. Jellison
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Jenna M. Bartley
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Laura Haynes
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
- Department of ImmunologyUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| |
Collapse
|
15
|
Lopez CE, Zacharias ZR, Ross KA, Narasimhan B, Waldschmidt TJ, Legge KL. Polyanhydride nanovaccine against H3N2 influenza A virus generates mucosal resident and systemic immunity promoting protection. NPJ Vaccines 2024; 9:96. [PMID: 38822003 PMCID: PMC11143372 DOI: 10.1038/s41541-024-00883-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 05/07/2024] [Indexed: 06/02/2024] Open
Abstract
Influenza A virus (IAV) causes significant morbidity and mortality worldwide due to seasonal epidemics and periodic pandemics. The antigenic drift/shift of IAV continually gives rise to new strains and subtypes, aiding IAV in circumventing previously established immunity. As a result, there has been substantial interest in developing a broadly protective IAV vaccine that induces, durable immunity against multiple IAVs. Previously, a polyanhydride nanoparticle-based vaccine or nanovaccine (IAV-nanovax) encapsulating H1N1 IAV antigens was reported, which induced pulmonary B and T cell immunity and resulted in cross-strain protection against IAV. A key feature of IAV-nanovax is its ability to easily incorporate diverse proteins/payloads, potentially increasing its ability to provide broad protection against IAV and/or other pathogens. Due to human susceptibility to both H1N1 and H3N2 IAV, several H3N2 nanovaccines were formulated herein with multiple IAV antigens to examine the "plug-and-play" nature of the polyanhydride nanovaccine platform and determine their ability to induce humoral and cellular immunity and broad-based protection similar to IAV-nanovax. The H3N2-based IAV nanovaccine formulations induced systemic and mucosal B cell responses which were associated with antigen-specific antibodies. Additionally, systemic and lung-tissue resident CD4 and CD8 T cell responses were enhanced post-vaccination. These immune responses corresponded with protection against both homologous and heterosubtypic IAV infection. Overall, these results demonstrate the plug-and-play nature of the polyanhydride nanovaccine platform and its ability to generate immunity and protection against IAV utilizing diverse antigenic payloads.
Collapse
Affiliation(s)
- Christopher E Lopez
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Zeb R Zacharias
- Interdisciplinary Immunology Graduate Program, Department of Pathology, University of Iowa, Iowa City, IA, USA
| | | | - Balaji Narasimhan
- Nanovaccine Institute, Iowa State University, Ames, IA, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Thomas J Waldschmidt
- Interdisciplinary Immunology Graduate Program, Department of Pathology, University of Iowa, Iowa City, IA, USA
- Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Kevin L Legge
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA.
- Interdisciplinary Immunology Graduate Program, Department of Pathology, University of Iowa, Iowa City, IA, USA.
- Nanovaccine Institute, Iowa State University, Ames, IA, USA.
| |
Collapse
|
16
|
Costa-da-Silva AC, Villapudua CU, Hoffman MP, Aure MH. Immunomodulation of salivary gland function due to cancer therapy. Oral Dis 2024:10.1111/odi.14972. [PMID: 38696474 PMCID: PMC11530405 DOI: 10.1111/odi.14972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 05/04/2024]
Abstract
Functional salivary glands (SG) are essential for maintaining oral health, and salivary dysfunction is a persistent major clinical challenge. Several cancer therapies also have off-target effects leading to SG dysfunction. Recent advances highlight the role of SG immune populations in homeostasis, dysfunction and gland regeneration. Here, we review what is known about SG immune populations during development and postnatal homeostasis. We summarize recent findings of immune cell involvement in SG dysfunction following cancer treatments such as irradiation (IR) for head and neck cancers, immune transplant leading to graft-versus-host-disease (GVHD) and immune checkpoint inhibitor (ICI) treatment. The role of immune cells in SG in both homeostasis and disease, is an emerging field of research that may provide important clues to organ dysfunction and lead to novel therapeutic targets.
Collapse
Affiliation(s)
- Ana C. Costa-da-Silva
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Carlos U. Villapudua
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Matthew P. Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Marit H. Aure
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
17
|
Tembo G, Mayuni M, Kamng'ona R, Chimgoneko L, Chiwala G, Sichone S, Galafa B, Thole F, Mkandawire C, Chirwa AE, Nsomba E, Nkhoma V, Ngoliwa C, Toto N, Makhaza L, Muyaya A, Kudowa E, Henrion MYR, Dula D, Morton B, Chikaonda T, Gordon SB, Jambo KC. Poor association between 13-valent pneumococcal conjugate vaccine-induced serum and mucosal antibody responses with experimental Streptococcus pneumoniae serotype 6B colonisation. Vaccine 2024; 42:2975-2982. [PMID: 38570270 PMCID: PMC11056720 DOI: 10.1016/j.vaccine.2024.03.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/23/2024] [Accepted: 03/21/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Pneumococcal carriage is the primary reservoir for transmissionand a prerequisite for invasive pneumococcal disease. Pneumococcal Conjugate Vaccine 13 (PCV13) showed a 62% efficacy in protection against experimental Streptococcus pneumoniae serotype 6B (Spn6B) carriage in a controlled human infection model (CHIM) of healthy Malawian adults. We, therefore, measured humoral responses to experimental challenge and PCV-13 vaccination and determined the association with protection against pneumococcal carriage. METHODS We vaccinated 204 young, healthy Malawian adults with PCV13 or placebo and nasally inoculated them with Spn6B at least four weeks post-vaccination to establish carriage. We collected peripheral blood and nasal lining fluid at baseline, 4 weeks post-vaccination (7 days pre-inoculation), 2, 7, 14 and > 1 year post-inoculation. We measured the concentration of anti-serotype 6B Capsular Polysaccharide (CPS) Immunoglobulin G (IgG) and IgA antibodies in serum and nasal lining fluid using the World Health Organization (WHO) standardised enzyme-linked immunosorbent assay (ELISA). RESULTS PCV13-vaccinated adults had higher serum IgG and nasal IgG/IgA anti-Spn6B CPS-specific binding antibodies than placebo recipients 4 to 6 weeks post-vaccination, which persisted for at least a year after vaccination. Nasal challenge with Spn6B did not significantly alter serum or nasal anti-CPS IgG binding antibody titers with or without experimental pneumococcal carriage. Pre-challenge titers of PCV13-induced serum IgG and nasal IgG/IgA anti-Spn6B CPS binding antibodies did not significantly differ between those that got experimentally colonised by Spn6B compared to those that did not. CONCLUSION This study demonstrates that despite high PCV13 efficacy against experimental Spn6B carriage in young, healthy Malawian adults, robust vaccine-induced systemic and mucosal anti-Spn6B CPS binding antibodies did not directly relate to protection.
Collapse
Affiliation(s)
- G Tembo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi.
| | - M Mayuni
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - R Kamng'ona
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - L Chimgoneko
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - G Chiwala
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - S Sichone
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - B Galafa
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - F Thole
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - C Mkandawire
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - A E Chirwa
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - E Nsomba
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - V Nkhoma
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - C Ngoliwa
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - N Toto
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - L Makhaza
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - A Muyaya
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - E Kudowa
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - M Y R Henrion
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Liverpool School of Tropical Medicine, Clinical Sciences Department, Pembroke Place, Liverpool, UK
| | - D Dula
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - B Morton
- Liverpool School of Tropical Medicine, Clinical Sciences Department, Pembroke Place, Liverpool, UK
| | - T Chikaonda
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - S B Gordon
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Liverpool School of Tropical Medicine, Clinical Sciences Department, Pembroke Place, Liverpool, UK
| | - K C Jambo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Liverpool School of Tropical Medicine, Clinical Sciences Department, Pembroke Place, Liverpool, UK.
| |
Collapse
|
18
|
Etesami NS, Barker KA, Shenoy AT, De Ana CL, Arafa EI, Grifno GN, Matschulat AM, Vannini ME, Pihl RMF, Breen MP, Soucy AM, Goltry WN, Ha CT, Betsuyaku H, Browning JL, Varelas X, Traber KE, Jones MR, Quinton LJ, Maglione PJ, Nia HT, Belkina AC, Mizgerd JP. B cells in the pneumococcus-infected lung are heterogeneous and require CD4 + T cell help including CD40L to become resident memory B cells. Front Immunol 2024; 15:1382638. [PMID: 38715601 PMCID: PMC11074383 DOI: 10.3389/fimmu.2024.1382638] [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: 02/06/2024] [Accepted: 04/01/2024] [Indexed: 05/12/2024] Open
Abstract
Recovery from respiratory pneumococcal infections generates lung-localized protection against heterotypic bacteria, mediated by resident memory lymphocytes. Optimal protection in mice requires re-exposure to pneumococcus within days of initial infection. Serial surface marker phenotyping of B cell populations in a model of pneumococcal heterotypic immunity revealed that bacterial re-exposure stimulates the immediate accumulation of dynamic and heterogeneous populations of B cells in the lung, and is essential for the establishment of lung resident memory B (BRM) cells. The B cells in the early wave were activated, proliferating locally, and associated with both CD4+ T cells and CXCL13. Antagonist- and antibody-mediated interventions were implemented during this early timeframe to demonstrate that lymphocyte recirculation, CD4+ cells, and CD40 ligand (CD40L) signaling were all needed for lung BRM cell establishment, whereas CXCL13 signaling was not. While most prominent as aggregates in the loose connective tissue of bronchovascular bundles, morphometry and live lung imaging analyses showed that lung BRM cells were equally numerous as single cells dispersed throughout the alveolar septae. We propose that CD40L signaling from antigen-stimulated CD4+ T cells in the infected lung is critical to establishment of local BRM cells, which subsequently protect the airways and parenchyma against future potential infections.
Collapse
Affiliation(s)
- Neelou S. Etesami
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Kimberly A. Barker
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Anukul T. Shenoy
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Carolina Lyon De Ana
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Emad I. Arafa
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Gabrielle N. Grifno
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, MA, United States
| | - Adeline M. Matschulat
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Michael E. Vannini
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Riley M. F. Pihl
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Michael P. Breen
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Alicia M. Soucy
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Wesley N. Goltry
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Catherine T. Ha
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Hanae Betsuyaku
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Jeffrey L. Browning
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Xaralabos Varelas
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Katrina E. Traber
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Matthew R. Jones
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Lee J. Quinton
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Paul J. Maglione
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Hadi T. Nia
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, MA, United States
| | - Anna C. Belkina
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Flow Cytometry Core Facility, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Joseph P. Mizgerd
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
19
|
El Mahdaoui S, Hansen MM, von Essen MR, Hvalkof VH, Holm Hansen R, Mahler MR, Jennum P, Sellebjerg F, Romme Christensen J. CD11c + B cells in relapsing-remitting multiple sclerosis and effects of anti-CD20 therapy. Ann Clin Transl Neurol 2024; 11:926-937. [PMID: 38332555 PMCID: PMC11021659 DOI: 10.1002/acn3.52009] [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: 10/12/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
OBJECTIVES B cells are important in the pathogenesis of multiple sclerosis. It is yet unknown which subsets may be involved, but atypical B cells have been proposed as mediators of autoimmunity. In this study, we investigated differences in B-cell subsets between controls and patients with untreated and anti-CD20-treated multiple sclerosis. METHODS We recruited 155 participants for an exploratory cohort comprising peripheral blood and cerebrospinal fluid, and a validation cohort comprising peripheral blood. Flow cytometry was used to characterize B-cell phenotypes and effector functions of CD11c+ atypical B cells. RESULTS There were no differences in circulating B cells between controls and untreated multiple sclerosis. As expected, anti-CD20-treated patients had a markedly lower B-cell count. Of B cells remaining after treatment, we observed higher proportions of CD11c+ B cells and plasmablasts. CD11c+ B cells were expanded in cerebrospinal fluid compared to peripheral blood in controls and untreated multiple sclerosis. Surprisingly, the proportion of CD11c+ cerebrospinal fluid B cells was higher in controls and after anti-CD20 therapy than in untreated multiple sclerosis. Apart from the presence of plasmablasts, the cerebrospinal fluid B-cell composition after anti-CD20 therapy resembled that of controls. CD11c+ B cells demonstrated a high potential for both proinflammatory and regulatory cytokine production. INTERPRETATION The study demonstrates that CD11c+ B cells and plasmablasts are less efficiently depleted by anti-CD20 therapy, and that CD11c+ B cells comprise a phenotypically and functionally distinct, albeit heterogenous, B-cell subset with the capacity of exerting both proinflammatory and regulatory functions.
Collapse
Affiliation(s)
- Sahla El Mahdaoui
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Marie Mathilde Hansen
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Marina Rode von Essen
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Victoria Hyslop Hvalkof
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Rikke Holm Hansen
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Mie Reith Mahler
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| | - Poul Jennum
- Department of NeurologyDanish Center for Sleep Medicine, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagen2200Denmark
| | - Finn Sellebjerg
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagen2200Denmark
| | - Jeppe Romme Christensen
- Department of NeurologyDanish Multiple Sclerosis Center, Copenhagen University Hospital – RigshospitaletGlostrup2600Denmark
| |
Collapse
|
20
|
Lee EG, Oh JE. From neglect to spotlight: the underappreciated role of B cells in cutaneous inflammatory diseases. Front Immunol 2024; 15:1328785. [PMID: 38426103 PMCID: PMC10902158 DOI: 10.3389/fimmu.2024.1328785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
The skin, covering our entire body as its largest organ, manifests enormous complexities and a profound interplay of systemic and local responses. In this heterogeneous domain, B cells were considered strangers. Yet, recent studies have highlighted their existence in the skin and their distinct role in modulating cutaneous immunity across various immune contexts. Accumulating evidence is progressively shedding light on the significance of B cells in maintaining skin health and in skin disorders. Herein, we integrate current insights on the systemic and local contributions of B cells in three prevalent inflammatory skin conditions: Pemphigus Vulgaris (PV), Systemic Lupus Erythematosus (SLE), and Atopic Dermatitis (AD), underscoring the previously underappreciated importance of B cells within skin immunity. Moreover, we address the potential adverse effects of current treatments used for skin diseases, emphasizing their unintentional consequences on B cells. These comprehensive approaches may pave the way for innovative therapeutic strategies that effectively address the intricate nature of skin disorders.
Collapse
Affiliation(s)
- Eun-Gang Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ji Eun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- BioMedical Research Center, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
21
|
Hartmannsberger B, Scriba S, Guidolin C, Becker J, Mehling K, Doppler K, Sommer C, Rittner HL. Transient immune activation without loss of intraepidermal innervation and associated Schwann cells in patients with complex regional pain syndrome. J Neuroinflammation 2024; 21:23. [PMID: 38233858 PMCID: PMC10792943 DOI: 10.1186/s12974-023-02969-6] [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: 09/06/2023] [Accepted: 11/22/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Complex regional pain syndrome (CRPS) develops after injury and is characterized by disproportionate pain, oedema, and functional loss. CRPS has clinical signs of neuropathy as well as neurogenic inflammation. Here, we asked whether skin biopsies could be used to differentiate the contribution of these two systems to ultimately guide therapy. To this end, the cutaneous sensory system including nerve fibres and the recently described nociceptive Schwann cells as well as the cutaneous immune system were analysed. METHODS We systematically deep-phenotyped CRPS patients and immunolabelled glabrous skin biopsies from the affected ipsilateral and non-affected contralateral finger of 19 acute (< 12 months) and 6 chronic (> 12 months after trauma) CRPS patients as well as 25 sex- and age-matched healthy controls (HC). Murine foot pads harvested one week after sham or chronic constriction injury were immunolabelled to assess intraepidermal Schwann cells. RESULTS Intraepidermal Schwann cells were detected in human skin of the finger-but their density was much lower compared to mice. Acute and chronic CRPS patients suffered from moderate to severe CRPS symptoms and corresponding pain. Most patients had CRPS type I in the warm category. Their cutaneous neuroglial complex was completely unaffected despite sensory plus signs, e.g. allodynia and hyperalgesia. Cutaneous innate sentinel immune cells, e.g. mast cells and Langerhans cells, infiltrated or proliferated ipsilaterally independently of each other-but only in acute CRPS. No additional adaptive immune cells, e.g. T cells and plasma cells, infiltrated the skin. CONCLUSIONS Diagnostic skin punch biopsies could be used to diagnose individual pathophysiology in a very heterogenous disease like acute CRPS to guide tailored treatment in the future. Since numbers of inflammatory cells and pain did not necessarily correlate, more in-depth analysis of individual patients is necessary.
Collapse
Affiliation(s)
- Beate Hartmannsberger
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University of Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Sabrina Scriba
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University of Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Carolina Guidolin
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University of Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Juliane Becker
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University of Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Katharina Mehling
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University of Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Kathrin Doppler
- Department of Neurology, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Claudia Sommer
- Department of Neurology, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Heike L Rittner
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University of Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany.
| |
Collapse
|
22
|
Wagoner ZW, Mitul MT, Wagar LE. Using Ex Vivo Tonsil Organoids to Study Memory B Cells. Methods Mol Biol 2024; 2826:3-13. [PMID: 39017881 DOI: 10.1007/978-1-0716-3950-4_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: 07/18/2024]
Abstract
Tools to study memory B cell (MBC) development and function are needed to understand their role in supporting sustained protection against recurrent infections. While human MBCs are traditionally measured using blood, there is a growing interest in elucidating their behavior within lymphoid tissues, which are the main sites where adaptive immune responses are orchestrated. In this chapter, we introduce a high-throughput organoid system that is derived from primary human lymphoid tissues. The approach can recapitulate many hallmarks of successful adaptive immune responses and capture inter-individual variation in response to a variety of stimuli. Lymphoid tissue organoids enable characterization of pre-existing antigen-specific MBCs within an entirely human system and can provide valuable insights into MBC dynamics.
Collapse
Affiliation(s)
- Zachary W Wagoner
- Department of Physiology and Biophysics, Institute for Immunology, Center for Virus Research, and Vaccine R&D Center, University of California Irvine, Irvine, CA, USA
| | - Mahina Tabassum Mitul
- Department of Physiology and Biophysics, Institute for Immunology, Center for Virus Research, and Vaccine R&D Center, University of California Irvine, Irvine, CA, USA
| | - Lisa E Wagar
- Department of Physiology and Biophysics, Institute for Immunology, Center for Virus Research, and Vaccine R&D Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
23
|
Neziraj T, Siewert L, Pössnecker E, Pröbstel AK. Therapeutic targeting of gut-originating regulatory B cells in neuroinflammatory diseases. Eur J Immunol 2023; 53:e2250033. [PMID: 37624875 DOI: 10.1002/eji.202250033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/29/2023] [Accepted: 08/23/2023] [Indexed: 08/27/2023]
Abstract
Regulatory B cells (Bregs) are immunosuppressive cells that support immunological tolerance by the production of IL-10, IL-35, and TGF-β. Bregs arise from different developmental stages in response to inflammatory stimuli. In that regard, mounting evidence points towards a direct influence of gut microbiota on mucosal B cell development, activation, and regulation in health and disease. While an increasing number of diseases are associated with alterations in gut microbiome (dysbiosis), little is known about the role of microbiota on Breg development and induction in neuroinflammatory disorders. Notably, gut-originating, IL-10- and IgA-producing regulatory plasma cells have recently been demonstrated to egress from the gut to suppress inflammation in the CNS raising fundamental questions about the triggers and functions of mucosal-originating Bregs in systemic inflammation. Advancing our understanding of Bregs in neuroinflammatory diseases could lead to novel therapeutic approaches. Here, we summarize the main aspects of Breg differentiation and functions and evidence about their involvement in neuroinflammatory diseases. Further, we highlight current data of gut-originating Bregs and their microbial interactions and discuss future microbiota-regulatory B cell-targeted therapies in immune-mediated diseases.
Collapse
Affiliation(s)
- Tradite Neziraj
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Lena Siewert
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Elisabeth Pössnecker
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Anne-Katrin Pröbstel
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
24
|
Mitsi E, Diniz MO, Reiné J, Collins AM, Robinson RE, Hyder-Wright A, Farrar M, Liatsikos K, Hamilton J, Onyema O, Urban BC, Solórzano C, Belij-Rammerstorfer S, Sheehan E, Lambe T, Draper SJ, Weiskopf D, Sette A, Maini MK, Ferreira DM. Respiratory mucosal immune memory to SARS-CoV-2 after infection and vaccination. Nat Commun 2023; 14:6815. [PMID: 37884506 PMCID: PMC10603102 DOI: 10.1038/s41467-023-42433-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
Respiratory mucosal immunity induced by vaccination is vital for protection from coronavirus infection in animal models. In humans, the capacity of peripheral vaccination to generate sustained immunity in the lung mucosa, and how this is influenced by prior SARS-CoV-2 infection, is unknown. Here we show using bronchoalveolar lavage samples that donors with history of both infection and vaccination have more airway mucosal SARS-CoV-2 antibodies and memory B cells than those only vaccinated. Infection also induces populations of airway spike-specific memory CD4+ and CD8+ T cells that are not expanded by vaccination alone. Airway mucosal T cells induced by infection have a distinct hierarchy of antigen specificity compared to the periphery. Spike-specific T cells persist in the lung mucosa for 7 months after the last immunising event. Thus, peripheral vaccination alone does not appear to induce durable lung mucosal immunity against SARS-CoV-2, supporting an argument for the need for vaccines targeting the airways.
Collapse
Affiliation(s)
- Elena Mitsi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Mariana O Diniz
- Division of Infection and Immunity and Institute of Immunity and Transplantation, UCL, London, UK
| | - Jesús Reiné
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Andrea M Collins
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Ryan E Robinson
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Angela Hyder-Wright
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Madlen Farrar
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | - Josh Hamilton
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Onyia Onyema
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Britta C Urban
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Carla Solórzano
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | - Emma Sheehan
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, USA
| | - Mala K Maini
- Division of Infection and Immunity and Institute of Immunity and Transplantation, UCL, London, UK
| | - Daniela M Ferreira
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
- Department of Clinical Science, Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
25
|
Kim CM, Manhart LE, Wood GE. Serum and Urethral Antibody Response in Mycoplasma genitalium -Infected Men. Sex Transm Dis 2023; 50:e26-e29. [PMID: 37432986 PMCID: PMC10527067 DOI: 10.1097/olq.0000000000001845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
ABSTRACT The antibody response to Mycoplasma genitalium in serum and urethral secretions of men with nongonococcal urethritis was examined longitudinally. Serum and urethral antibodies reacted primarily with the MgpB and MgpC adhesins. Serum antibodies persisted throughout follow-up, whereas urethral antibodies waned despite organism persistence. Declining antibodies may facilitate chronic infection.
Collapse
Affiliation(s)
- Caroline M. Kim
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA
| | - Lisa E. Manhart
- Center for AIDS and STD and Department of Epidemiology, University of Washington, Seattle, WA
| | - Gwendolyn E. Wood
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA
| |
Collapse
|
26
|
Longet S, Paul S. Pivotal role of tissue-resident memory lymphocytes in the control of mucosal infections: can mucosal vaccination induce protective tissue-resident memory T and B cells? Front Immunol 2023; 14:1216402. [PMID: 37753095 PMCID: PMC10518612 DOI: 10.3389/fimmu.2023.1216402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Affiliation(s)
- Stephanie Longet
- Centre International de Recherche en Infectiologie, Team Groupe sur l'immunité des muqueuses et agents pathogènes (GIMAP), Université Jean Monnet, Université Claude Bernard Lyon, Inserm, Saint-Etienne, France
| | - Stephane Paul
- Centre International de Recherche en Infectiologie, Team Groupe sur l'immunité des muqueuses et agents pathogènes (GIMAP), Université Jean Monnet, Université Claude Bernard Lyon, Inserm, Saint-Etienne, France
- Centre d'investigation clinique (CIC) 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
- Immunology Department, iBiothera Reference Center, University Hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
27
|
Dickson A, Geerling E, Stone ET, Hassert M, Steffen TL, Makkena T, Smither M, Schwetye KE, Zhang J, Georges B, Roberts MS, Suschak JJ, Pinto AK, Brien JD. The role of vaccination route with an adenovirus-vectored vaccine in protection, viral control, and transmission in the SARS-CoV-2/K18-hACE2 mouse infection model. Front Immunol 2023; 14:1188392. [PMID: 37662899 PMCID: PMC10469340 DOI: 10.3389/fimmu.2023.1188392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/22/2023] [Indexed: 09/05/2023] Open
Abstract
Introduction Vaccination is the most effective mechanism to prevent severe COVID-19. However, breakthrough infections and subsequent transmission of SARS-CoV-2 remain a significant problem. Intranasal vaccination has the potential to be more effective in preventing disease and limiting transmission between individuals as it induces potent responses at mucosal sites. Methods Utilizing a replication-deficient adenovirus serotype 5-vectored vaccine expressing the SARS-CoV-2 RBD (AdCOVID) in homozygous and heterozygous transgenic K18-hACE2, we investigated the impact of the route of administration on vaccine immunogenicity, SARS-CoV-2 transmission, and survival. Results Mice vaccinated with AdCOVID via the intramuscular or intranasal route and subsequently challenged with SARS-CoV-2 showed that animals vaccinated intranasally had improved cellular and mucosal antibody responses. Additionally, intranasally vaccinated animals had significantly better viremic control, and protection from lethal infection compared to intramuscularly vaccinated animals. Notably, in a novel transmission model, intranasal vaccination reduced viral transmission to naïve co-housed mice compared to intramuscular vaccination. Discussion Our data provide convincing evidence for the use of intranasal vaccination in protecting against SARS-CoV-2 infection and transmission.
Collapse
Affiliation(s)
- Alexandria Dickson
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - E. Taylor Stone
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Mariah Hassert
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Tara L. Steffen
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Taneesh Makkena
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Madeleine Smither
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Katherine E. Schwetye
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | | | | | | | | | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| |
Collapse
|
28
|
Zhu W, Park J, Pho T, Wei L, Dong C, Kim J, Ma Y, Champion JA, Wang BZ. ISCOMs/MPLA-Adjuvanted SDAD Protein Nanoparticles Induce Improved Mucosal Immune Responses and Cross-Protection in Mice. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301801. [PMID: 37162451 PMCID: PMC10524461 DOI: 10.1002/smll.202301801] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/20/2023] [Indexed: 05/11/2023]
Abstract
The epidemics caused by the influenza virus are a serious threat to public health and the economy. Adding appropriate adjuvants to improve immunogenicity and finding effective mucosal vaccines to combat respiratory infection at the portal of virus entry are important strategies to boost protection. In this study, a novel type of core/shell protein nanoparticle consisting of influenza nucleoprotein (NP) as the core and NA1-M2e or NA2-M2e fusion proteins as the coating antigens by SDAD hetero-bifunctional crosslinking is exploited. Immune-stimulating complexes (ISCOMs)/monophosphoryl lipid A (MPLA) adjuvants further boost the NP/NA-M2e SDAD protein nanoparticle-induced immune responses when administered intramuscularly. The ISCOMs/MPLA-adjuvanted protein nanoparticles are delivered through the intranasal route to validate the application as mucosal vaccines. ISCOMs/MPLA-adjuvanted nanoparticles induce significantly strengthened antigen-specific antibody responses, cytokine-secreting splenocytes in the systemic compartment, and higher levels of antigen-specific IgA and IgG in the local mucosa. Meanwhile, significantly expanded lung resident memory (RM) T and B cells (TRM /BRM ) and alveolar macrophages population are observed in ISCOMs/MPLA-adjuvanted nanoparticle-immunized mice with a 100% survival rate after homogeneous and heterogeneous H3N2 viral challenges. Taken together, ISCOMs/MPLA-adjuvanted protein nanoparticles could improve strong systemic and mucosal immune responses conferring protection in different immunization routes.
Collapse
Affiliation(s)
- Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Jaeyoung Park
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Thomas Pho
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Bioengineering Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Joo Kim
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Julie A. Champion
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Bioengineering Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
29
|
Jung HE, Ku KB, Kang BH, Park JH, Kim HC, Kim KD, Lee HK. Intranasal delivery of an adenovirus-vector vaccine co-expressing a modified spike protein and a genetic adjuvant confers lasting mucosal immunity against SARS-CoV-2. Antiviral Res 2023; 216:105656. [PMID: 37327877 PMCID: PMC10265935 DOI: 10.1016/j.antiviral.2023.105656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 06/18/2023]
Abstract
The ongoing COVID-19 pandemic caused by SARS-CoV-2 infection has threatened global health. Since the first case of infection was reported in December 2019, SARS-CoV-2 has rapidly spread worldwide and caused millions of deaths. As vaccination is the best way to protect the host from invading pathogens, several vaccines have been developed to prevent the infection of SARS-CoV-2, saving numerous lives thus far. However, SARS-CoV-2 constantly changes its antigens, resulting in escape from vaccine-induced protection, and the longevity of immunity induced by vaccines remains an issue. Additionally, traditional intramuscular COVID-19 vaccines are insufficient at evoking mucosal-specific immune responses. Because the respiratory tract is the primary route of SARS-CoV-2 entry, the need for mucosal vaccines is strong. Using an adenoviral (Ad) vector platform, we generated Ad5-S.Mod, a recombinant COVID-19 vaccine that encodes modified-spike (S) antigen and the genetic adjuvant human CXCL9. Intranasal delivery of Ad5-S.Mod elicited superior airway humoral and T-cell responses over traditional intramuscular vaccines and protected mice from lethal SARS-CoV-2 infection. cDC1 cells were required for the generation of antigen-specific CD8+ T-cell responses and CD8+ tissue-resident memory T-cell development in intranasal Ad5-S.Mod vaccinated mice. Furthermore, we confirmed the efficacy of the intranasal Ad5-S.Mod vaccine in terms of transcriptional changes and identified lung macrophages as a key supporter of maintenance of lung-resident memory T and B cells. Our study demonstrates Ad5-S.Mod has the potential to confer protective immunity against SARS-CoV-2 and that lung macrophages support the maintenance of vaccine-induced tissue-resident memory lymphocytes.
Collapse
Affiliation(s)
- Hi Eun Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Keun Bon Ku
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea; Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Byeong Hoon Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jang Hyun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hyeon Cheol Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Kyun-Do Kim
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea; Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
30
|
Nakahashi-Ouchida R, Fujihashi K, Kurashima Y, Yuki Y, Kiyono H. Nasal vaccines: solutions for respiratory infectious diseases. Trends Mol Med 2023; 29:124-140. [PMID: 36435633 DOI: 10.1016/j.molmed.2022.10.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/07/2022] [Accepted: 10/26/2022] [Indexed: 11/25/2022]
Abstract
Nasal vaccines induce pathogen-specific dual protective immunity at mucosal surfaces and systemically throughout the body. Consequently, nasal vaccines both prevent pathogen invasion and reduce disease severity. Because of these features, nasal vaccines are considered to be a next-generation tool for preventing respiratory infectious diseases, including COVID-19. However, nasal vaccines must overcome key safety concerns given the anatomic proximity of the central nervous system (CNS) via the olfactory bulbs which lie next to the nasal cavity. This review summarizes current efforts to develop safe and effective nasal vaccines and delivery systems, as well as their clinical applications for the prevention of respiratory infections. We also discuss various concerns regarding the safety of nasal vaccines and introduce a system for evaluating them.
Collapse
Affiliation(s)
- Rika Nakahashi-Ouchida
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, Chiba University, Chiba, Japan
| | - Kohtaro Fujihashi
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, Chiba University, Chiba, Japan; Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yosuke Kurashima
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, Chiba University, Chiba, Japan; Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan; Institute for Advanced Academic Research, Chiba University, Chiba, Japan; Chiba University-University of California San Diego (CU-UCSD) Center for Mucosal Immunology, Allergy, and Vaccines (cMAV), Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
| | - Yoshikazu Yuki
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; HanaVax Inc., Tokyo, Japan
| | - Hiroshi Kiyono
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, Chiba University, Chiba, Japan; Institute for Advanced Academic Research, Chiba University, Chiba, Japan; Chiba University-University of California San Diego (CU-UCSD) Center for Mucosal Immunology, Allergy, and Vaccines (cMAV), Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA; Future Medicine Education and Research Organization, Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Chiba University, Chiba, Japan.
| |
Collapse
|
31
|
Bertrand Y, Sánchez-Montalvo A, Hox V, Froidure A, Pilette C. IgA-producing B cells in lung homeostasis and disease. Front Immunol 2023; 14:1117749. [PMID: 36936934 PMCID: PMC10014553 DOI: 10.3389/fimmu.2023.1117749] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
Immunoglobulin A (IgA) is the most abundant Ig in mucosae where it plays key roles in host defense against pathogens and in mucosal immunoregulation. Whereas intense research has established the different roles of secretory IgA in the gut, its function has been much less studied in the lung. This review will first summarize the state-of-the-art knowledge on the distribution and phenotype of IgA+ B cells in the human lung in both homeostasis and disease. Second, it will analyze the studies looking at cellular and molecular mechanisms of homing and priming of IgA+ B cells in the lung, notably following immunization. Lastly, published data on observations related to IgA and IgA+ B cells in lung and airway disease such as asthma, cystic fibrosis, idiopathic pulmonary fibrosis, or chronic rhinosinusitis, will be discussed. Collectively it provides the state-of-the-art of our current understanding of the biology of IgA-producing cells in the airways and identifies gaps that future research should address in order to improve mucosal protection against lung infections and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Youri Bertrand
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
| | - Alba Sánchez-Montalvo
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Allergy and Clinical Immunology Research Group, Department of Microbiology, Immunology and Transplantation, Katholieke universiteit (KU) Leuven, Leuven, Belgium
| | - Valérie Hox
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Department of Otorhinolaryngology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Antoine Froidure
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Service de Pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Charles Pilette
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Service de Pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- *Correspondence: Charles Pilette,
| |
Collapse
|