1
|
Løberg RI, Dewan AE, Kleppa L, du Pré MF, Sollid LM. A Mouse Model for Generation of Gut Lamina Propria Plasma Cells Specific for a Deamidated Gluten Peptide. Eur J Immunol 2025; 55:e202451658. [PMID: 40098349 DOI: 10.1002/eji.202451658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Celiac disease is an autoimmune enteropathy caused by aberrant immune responses to dietary gluten peptides. Plasma cells (PCs) reactive with deamidated gluten peptides (DGP) or transglutaminase 2 are abundant in celiac disease gut lesions, yet their role in disease pathogenesis remains unclear. Here, we present a mouse model that allows for exploring the role of DGP-specific IgA PCs. This model employs a novel immunoglobulin knock-in (Ig KI) mouse expressing a celiac-patient-derived anti-DGP B-cell receptor (BCR) that recognizes an immunodominant DGP epitope. In these mice, ∼80% of splenic B cells express the transgenic BCR. In co-culture experiments with transgenic DGP-specific B cells and transgenic gluten-specific CD4+ T cells, stimulation with DGP led to T-cell and B-cell proliferation. Mice carrying the celiac disease-associated human leukocyte antigen (HLA) allotype HLA-DQ2.5 developed DGP-specific small intestinal IgA PCs upon adoptive transfer of HLA-DQ2.5-expressing DGP-specific B cells and oral immunizations with DGP and cholera toxin (CT). However, covalent conjugation of DGP to CT was required for effective anti-DGP gut immunity. This novel mouse model provides an important tool for studying the role of PCs beyond antibody production in celiac disease.
Collapse
Affiliation(s)
- Runa I Løberg
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Alisa E Dewan
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Liv Kleppa
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - M Fleur du Pré
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| |
Collapse
|
2
|
Artola-Borán M, Kirsche L, Fallegger A, Leary P, Tanriover M, Goodwin T, Geiger G, Hapfelmeier S, Yousefi S, Simon HU, Arnold IC, Müller A. IgA facilitates the persistence of the mucosal pathogen Helicobacter pylori. Mucosal Immunol 2025; 18:232-247. [PMID: 39581230 DOI: 10.1016/j.mucimm.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024]
Abstract
IgA antibodies have an important role in clearing mucosal pathogens. In this study, we have examined the contribution of IgA to the immune control of the gastrointestinal bacterial pathogens Helicobacter pylori and Citrobacter rodentium. Both bacteria trigger a strong local IgA response that results in bacterial IgA coating in mice and in gastritis patients. Class switching to IgA depends on Peyer's patches, T-cells, eosinophils, and eosinophil-derived TGF-β in both models. In the case of H. pylori, IgA secretion and bacterial coating also depend on a functional bacterial type IV secretion system, which drives the generation of Th17 cells and the IL-17-dependent expression of the polymeric immunoglobulin receptor PIGR. IgA-/- mice are hypercolonized with C. rodentium in all examined tissues, suffer from more severe weight loss and develop more colitis. In contrast, H. pylori is controlled more efficiently in IgA-/- mice than their WT counterparts. The effects of IgA deficiency of the offspring can be compensated by maternal IgA delivered by WT foster mothers. We attribute the improved immune control observed in IgA-/- mice to IgA-mediated protection from complement killing, as H. pylori colonization is restored to wild type levels in a composite strain lacking both IgA and the central complement component C3. IgA antibodies can thus have protective or detrimental activities depending on the infectious agent.
Collapse
Affiliation(s)
- Mariela Artola-Borán
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Lydia Kirsche
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Angela Fallegger
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Peter Leary
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Functional Genomics Center Zürich, University of Zürich/ETHZ, Zürich, Switzerland
| | - Mine Tanriover
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Tanja Goodwin
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Gavin Geiger
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | | | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, 16816 Neuruppin, Germany
| | - Isabelle C Arnold
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Comprehensive Cancer Center Zürich, Zürich, Switzerland.
| |
Collapse
|
3
|
Carreto-Binaghi LE, Sztein MB, Booth JS. Role of cellular effectors in the induction and maintenance of IgA responses leading to protective immunity against enteric bacterial pathogens. Front Immunol 2024; 15:1446072. [PMID: 39324143 PMCID: PMC11422102 DOI: 10.3389/fimmu.2024.1446072] [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: 06/08/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
The mucosal immune system is a critical first line of defense to infectious diseases, as many pathogens enter the body through mucosal surfaces, disrupting the balanced interactions between mucosal cells, secretory molecules, and microbiota in this challenging microenvironment. The mucosal immune system comprises of a complex and integrated network that includes the gut-associated lymphoid tissues (GALT). One of its primary responses to microbes is the secretion of IgA, whose role in the mucosa is vital for preventing pathogen colonization, invasion and spread. The mechanisms involved in these key responses include neutralization of pathogens, immune exclusion, immune modulation, and cross-protection. The generation and maintenance of high affinity IgA responses require a delicate balance of multiple components, including B and T cell interactions, innate cells, the cytokine milieu (e.g., IL-21, IL-10, TGF-β), and other factors essential for intestinal homeostasis, including the gut microbiota. In this review, we will discuss the main cellular components (e.g., T cells, innate lymphoid cells, dendritic cells) in the gut microenvironment as mediators of important effector responses and as critical players in supporting B cells in eliciting and maintaining IgA production, particularly in the context of enteric infections and vaccination in humans. Understanding the mechanisms of humoral and cellular components in protection could guide and accelerate the development of more effective mucosal vaccines and therapeutic interventions to efficiently combat mucosal infections.
Collapse
Affiliation(s)
- Laura E. Carreto-Binaghi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Laboratorio de Inmunobiologia de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Tumor Immunology and Immunotherapy Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Jayaum S. Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
Siniscalco ER, Williams A, Eisenbarth SC. All roads lead to IgA: Mapping the many pathways of IgA induction in the gut. Immunol Rev 2024; 326:66-82. [PMID: 39046160 DOI: 10.1111/imr.13369] [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: 07/25/2024]
Abstract
The increasing prevalence of food allergy and related pathologies in recent years has underscored the need to understand the factors affecting adverse reactions to food. Food allergy is caused when food-specific IgE triggers the release of histamine from mast cells. However, other food-specific antibody isotypes exist as well, including IgG and IgA. IgA is the main antibody isotype in the gut and mediates noninflammatory reactions to toxins, commensal bacteria, and food antigens. It has also been thought to induce tolerance to food, thus antagonizing the role of food-specific IgE. However, this has remained unclear as food-specific IgA generation is poorly understood. Particularly, the location of IgA induction, the role of T cell help, and the fates of food-specific B cells remain elusive. In this review, we outline what is known about food-specific IgA induction and highlight areas requiring further study. We also explore how knowledge of food-specific IgA induction can be informed by and subsequently contribute to our overall knowledge of gut immunity.
Collapse
Affiliation(s)
- Emily R Siniscalco
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Adam Williams
- Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Allergy and Immunology, The Department Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Stephanie C Eisenbarth
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Allergy and Immunology, The Department Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
5
|
Liu J, Stoler-Barak L, Hezroni-Bravyi H, Biram A, Lebon S, Davidzohn N, Kedmi M, Chemla M, Pilzer D, Cohen M, Brenner O, Biton M, Shulman Z. Turbinate-homing IgA-secreting cells originate in the nasal lymphoid tissues. Nature 2024; 632:637-646. [PMID: 39085603 DOI: 10.1038/s41586-024-07729-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/18/2024] [Indexed: 08/02/2024]
Abstract
Nasal vaccination elicits a humoral immune response that provides protection from airborne pathogens1, yet the origins and specific immune niches of antigen-specific IgA-secreting cells in the upper airways are unclear2. Here we define nasal glandular acinar structures and the turbinates as immunological niches that recruit IgA-secreting plasma cells from the nasal-associated lymphoid tissues (NALTs)3. Using intact organ imaging, we demonstrate that nasal vaccination induces B cell expansion in the subepithelial dome of the NALT, followed by invasion into commensal-bacteria-driven chronic germinal centres in a T cell-dependent manner. Initiation of the germinal centre response in the NALT requires pre-expansion of antigen-specific T cells, which interact with cognate B cells in interfollicular regions. NALT ablation and blockade of PSGL-1, which mediates interactions with endothelial cell selectins, demonstrated that NALT-derived IgA-expressing B cells home to the turbinate region through the circulation, where they are positioned primarily around glandular acinar structures. CCL28 expression was increased in the turbinates in response to vaccination and promoted homing of IgA+ B cells to this site. Thus, in response to nasal vaccination, the glandular acini and turbinates provide immunological niches that host NALT-derived IgA-secreting cells. These cellular events could be manipulated in vaccine design or in the treatment of upper airway allergic responses.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Liat Stoler-Barak
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Hadas Hezroni-Bravyi
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Adi Biram
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Sacha Lebon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Natalia Davidzohn
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Kedmi
- Department of Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
- The Nancy & Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | - Muriel Chemla
- Department of Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
- The Nancy & Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | - David Pilzer
- Department of Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
- The Nancy & Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | - Marina Cohen
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Ori Brenner
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Moshe Biton
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Shulman
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
6
|
Zhang B, Chen S, Yin X, McBride CD, Gertie JA, Yurieva M, Bielecka AA, Hoffmann B, Travis Hinson J, Grassmann J, Xu L, Siniscalco ER, Soldatenko A, Hoyt L, Joseph J, Norton EB, Uthaman G, Palm NW, Liu E, Eisenbarth SC, Williams A. Metabolic fitness of IgA + plasma cells in the gut requires DOCK8. Mucosal Immunol 2024; 17:431-449. [PMID: 38159726 PMCID: PMC11571232 DOI: 10.1016/j.mucimm.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/16/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Dedicator of cytokinesis 8 (DOCK8) mutations lead to a primary immunodeficiency associated with recurrent gastrointestinal infections and poor antibody responses but, paradoxically, heightened IgE to food antigens, suggesting that DOCK8 is central to immune homeostasis in the gut. Using Dock8-deficient mice, we found that DOCK8 was necessary for mucosal IgA production to multiple T cell-dependent antigens, including peanut and cholera toxin. Yet DOCK8 was not necessary in T cells for this phenotype. Instead, B cell-intrinsic DOCK8 was required for maintenance of antigen-specific IgA-secreting plasma cells (PCs) in the gut lamina propria. Unexpectedly, DOCK8 was not required for early B cell activation, migration, or IgA class switching. An unbiased interactome screen revealed novel protein partners involved in metabolism and apoptosis. Dock8-deficient IgA+ B cells had impaired cellular respiration and failed to engage glycolysis appropriately. These results demonstrate that maintenance of the IgA+ PC compartment requires DOCK8 and suggest that gut IgA+ PCs have unique metabolic requirements for long-term survival in the lamina propria.
Collapse
Affiliation(s)
- Biyan Zhang
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Shuting Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xiangyun Yin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Caleb D McBride
- The Department Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jake A Gertie
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Marina Yurieva
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Agata A Bielecka
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Microbial Immunoregulation, Helmholtz Center for Infection Research, 38124 Braunschweig, Germany
| | - Brian Hoffmann
- Mass Spectrometry and Protein Chemistry, The Jackson Laboratory for Genomic Medicine, Bar Harbor, ME 04609, USA
| | - J Travis Hinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA; Cardiology center, Department of Medicine, UConn Health, Farmington, CT, USA
| | - Jessica Grassmann
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Lan Xu
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Emily R Siniscalco
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Arielle Soldatenko
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Laura Hoyt
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Julie Joseph
- Department of Laboratory Medicine, USA; Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Elizabeth B Norton
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Gowthaman Uthaman
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Elise Liu
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; The Department Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Adam Williams
- The Department Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA; Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
7
|
Bemark M, Pitcher MJ, Dionisi C, Spencer J. Gut-associated lymphoid tissue: a microbiota-driven hub of B cell immunity. Trends Immunol 2024; 45:211-223. [PMID: 38402045 PMCID: PMC11227984 DOI: 10.1016/j.it.2024.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/26/2024]
Abstract
The diverse gut microbiota, which is associated with mucosal health and general wellbeing, maintains gut-associated lymphoid tissues (GALT) in a chronically activated state, including sustainment of germinal centers in a context of high antigenic load. This influences the rules for B cell engagement with antigen and the potential consequences. Recent data have highlighted differences between GALT and other lymphoid tissues. For example, GALT propagates IgA responses against glycans that show signs of having been generated in germinal centers. Other findings suggest that humans are among those species where GALT supports the diversification, propagation, and possibly selection of systemic B cells. Here, we review novel findings that identify GALT as distinctive, and able to support these processes.
Collapse
Affiliation(s)
- Mats Bemark
- Department of Translational Medicine - Human Immunology, Lund University, J Waldenströms gata 35, Malmö, Sweden; Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Michael J Pitcher
- Peter Gorer Department of Immunobiology, King's College London, Guy's Hospital Campus, St Thomas' Street, London SE1 9RT, UK
| | - Chiara Dionisi
- Peter Gorer Department of Immunobiology, King's College London, Guy's Hospital Campus, St Thomas' Street, London SE1 9RT, UK
| | - Jo Spencer
- Peter Gorer Department of Immunobiology, King's College London, Guy's Hospital Campus, St Thomas' Street, London SE1 9RT, UK.
| |
Collapse
|
8
|
De Giovanni M, Vykunta VS, Biram A, Chen KY, Taglinao H, An J, Sheppard D, Paidassi H, Cyster JG. Mast cells help organize the Peyer's patch niche for induction of IgA responses. Sci Immunol 2024; 9:eadj7363. [PMID: 38427721 PMCID: PMC11008922 DOI: 10.1126/sciimmunol.adj7363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 01/23/2024] [Indexed: 03/03/2024]
Abstract
Peyer's patches (PPs) are lymphoid structures situated adjacent to the intestinal epithelium that support B cell responses that give rise to many intestinal IgA-secreting cells. Induction of isotype switching to IgA in PPs requires interactions between B cells and TGFβ-activating conventional dendritic cells type 2 (cDC2s) in the subepithelial dome (SED). However, the mechanisms promoting cDC2 positioning in the SED are unclear. Here, we found that PP cDC2s express GPR35, a receptor that promotes cell migration in response to various metabolites, including 5-hydroxyindoleacetic acid (5-HIAA). In mice lacking GPR35, fewer cDC2s were found in the SED, and frequencies of IgA+ germinal center (GC) B cells were reduced. IgA plasma cells were reduced in both the PPs and lamina propria. These phenotypes were also observed in chimeric mice that lacked GPR35 selectively in cDCs. GPR35 deficiency led to reduced coating of commensal bacteria with IgA and reduced IgA responses to cholera toxin. Mast cells were present in the SED, and mast cell-deficient mice had reduced PP cDC2s and IgA+ cells. Ablation of tryptophan hydroxylase 1 (Tph1) in mast cells to prevent their production of 5-HIAA similarly led to reduced PP cDC2s and IgA responses. Thus, mast cell-guided positioning of GPR35+ cDC2s in the PP SED supports induction of intestinal IgA responses.
Collapse
Affiliation(s)
- Marco De Giovanni
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Vivasvan S. Vykunta
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Medical Scientist Training Program, School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Adi Biram
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kevin Y. Chen
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Medical Scientist Training Program, School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hanna Taglinao
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dean Sheppard
- Lung Biology Center, Department of Medicine, University of California San Francisco, 1550 4 Street, San Francisco, CA 94158, USA
| | - Helena Paidassi
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, France
| | - Jason G. Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
9
|
Pabst O, Nowosad CR. B cells and the intestinal microbiome in time, space and place. Semin Immunol 2023; 69:101806. [PMID: 37473559 DOI: 10.1016/j.smim.2023.101806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 07/08/2023] [Indexed: 07/22/2023]
Abstract
The gut immune system is shaped by the continuous interaction with the microbiota. Here we dissect temporal, spatial and contextual layers of gut B cell responses. The microbiota impacts on the selection of the developing pool of pre-immune B cells that serves as substrate for B cell activation, expansion and differentiation. However, various aspects of the gut B cell response display unique features. In particular, occurrence of somatically mutated B cells, chronic gut germinal centers in T cell-deficient settings and polyreactive binding of gut IgA to the microbiota questioned the nature and microbiota-specificity of gut germinal centers. We propose a model to reconcile these observations incorporating recent work demonstrating microbiota-specificity of gut germinal centers. We speculate that adjuvant effects of the microbiota might modify permissiveness for B cell to enter and exit gut germinal centers. We propose that separating aspects of time, space and place facilitate the occasionally puzzling discussion of gut B cell responses to the microbiota.
Collapse
Affiliation(s)
- Oliver Pabst
- Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany.
| | - Carla R Nowosad
- Department of Pathology, NYU Grossman School of Medicine, New York University, New York, USA; Translational Immunology Center, NYU Grossman School of Medicine, New York University, New York, USA.
| |
Collapse
|
10
|
Hernandez-Cazares F, Maqueda-Alfaro RA, Lopez-Saucedo C, Martinez-Barnetche J, Yam-Puc JC, Estrada-Parra S, Flores-Romo L, Estrada-Garcia T. Elevated levels of enteric IgA in an unimmunised mouse model of Hyper IgM syndrome derived from gut-associated secondary lymph organs even in the absence of germinal centres. Front Cell Infect Microbiol 2023; 13:1172021. [PMID: 37457961 PMCID: PMC10339347 DOI: 10.3389/fcimb.2023.1172021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Patients with Human Hyper IgM syndromes (HIGM) developed pulmonary and gastrointestinal infections since infancy and most patients have mutations in the CD40 ligand (CD40L) gene. Most HIGM patients compared to healthy subjects have higher/similar IgM and lower IgG, and IgA serum concentrations but gut antibody concentrations are unknown. CD40L on activated T-cells interacts with CD40 on B-cells, essential for the formation of germinal centres (GCs) inside secondary lymphoid organs (SLOs), where high-affinity antibodies, long-lived antibody-secreting plasma cells, and memory B-cells, are produced. C57BL6-CD40 ligand deficient mice (C57BL6-cd40l -/-), are a model of HIGM, because serum immunoglobulin concentrations parallel levels observed in HIGM patients and have higher faecal IgA concentrations. In mice, TGFβ and other cytokines induce IgA production. Aims To compare and evaluate B-cell populations and IgA-producing plasma cells in peritoneal lavage, non-gut-associated SLOs, spleen/inguinal lymph nodes (ILN), and gut-associated SLOs, mesenteric lymph nodes (MLN)/Peyer´s patches (PP) of unimmunised C57BL6-cd40l -/- and C57BL6-wild-type (WT) mice. Material and methods Peritoneal lavages, spleens, ILN, MLN, and PP from 8-10 weeks old C57BL6-cd40l -/- and WT mice, were obtained. Organ cryosections were analysed by immunofluorescence and B-cell populations and IgA-positive plasma cell suspensions by flow cytometry. Results In unimmunised WT mice, GCs were only observed in the gut-associated SLOs, but GCs were absent in all C57BL6-cd40l -/- SLOs. PP and MLN of C57BL6-cd40l -/- mice exhibited a significantly higher number of IgA-producing cells than WT mice. In the spleen and ILN of C57BL6-cd40l- /- mice IgA-producing cells significantly decreased, while IgM-positive plasma cells increased. C57BL6-cd40l -/- B-1 cells were more abundant in all analysed SLOs, whereas in WT mice most B-1 cells were contained within the peritoneal cavity. C57BL6-cd40l -/- B-cells in MLN expressed a higher TGFβ receptor-1 than WT mice. Mouse strains small intestine microvilli (MV), have a similar frequency of IgA-positive cells. Discussion Together our results confirm the role of PP and MLN as gut inductive sites, whose characteristic features are to initiate an IgA preferential immune response production in these anatomical sites even in the absence of GCs. IgA antibodies play a pivotal role in neutralising, eliminating, and regulating potential pathogens and microorganisms in the gut.
Collapse
Affiliation(s)
| | | | | | - Jesus Martinez-Barnetche
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, Mexico
| | | | - Sergio Estrada-Parra
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | | | | |
Collapse
|
11
|
Sollid LM, Iversen R. Tango of B cells with T cells in the making of secretory antibodies to gut bacteria. Nat Rev Gastroenterol Hepatol 2023; 20:120-128. [PMID: 36056203 DOI: 10.1038/s41575-022-00674-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/27/2022] [Indexed: 02/03/2023]
Abstract
Polymeric IgA and IgM are transported across the epithelial barrier from plasma cells in the lamina propria to exert a function in the gut lumen as secretory antibodies. Many secretory antibodies are reactive with the gut bacteria, and mounting evidence suggests that these antibodies are important for the host to control gut bacterial communities. However, we have incomplete knowledge of how bacteria-reactive secretory antibodies are formed. Antibodies from gut plasma cells often show bacterial cross-species reactivity, putting the degree of specificity behind anti-bacterial antibody responses into question. Such cross-species reactive antibodies frequently recognize non-genome-encoded membrane glycan structures. On the other hand, the T cell epitopes are peptides encoded in the bacterial genomes, thereby allowing a higher degree of predictable specificity on the T cell side of anti-bacterial immune responses. In this Perspective, we argue that the production of bacteria-reactive secretory antibodies is mainly controlled by the antigen specificity of T cells, which provide help to B cells. To be able to harness this system (for instance, for manipulation with vaccines), we need to obtain insight into the bacterial epitopes recognized by T cells in addition to characterizing the reactivity of the antibodies.
Collapse
Affiliation(s)
- Ludvig M Sollid
- K.G. Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway. .,Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| | - Rasmus Iversen
- K.G. Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway. .,Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| |
Collapse
|
12
|
Canales-Herrerias P, Uzzan M, Seki A, Czepielewski RS, Verstockt B, Livanos A, Raso F, Dunn A, Dai D, Wang A, Al-taie Z, Martin J, Ko HM, Tokuyama M, Tankelevich M, Meringer H, Cossarini F, Jha D, Krek A, Paulsen JD, Nakadar MZ, Wong J, Erlich EC, Onufer EJ, Helmink BA, Sharma K, Rosenstein A, Chung G, Dawson T, Juarez J, Yajnik V, Cerutti A, Faith J, Suarez-Farinas M, Argmann C, Petralia F, Randolph GJ, Polydorides AD, Reboldi A, Colombel JF, Mehandru S. Gut-associated lymphoid tissue attrition associates with response to anti-α4β7 therapy in ulcerative colitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524731. [PMID: 36711839 PMCID: PMC9882272 DOI: 10.1101/2023.01.19.524731] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Targeting the α4β7-MAdCAM-1 axis with vedolizumab (VDZ) is a front-line therapeutic paradigm in ulcerative colitis (UC). However, mechanism(s) of action (MOA) of VDZ remain relatively undefined. Here, we examined three distinct cohorts of patients with UC (n=83, n=60, and n=21), to determine the effect of VDZ on the mucosal and peripheral immune system. Transcriptomic studies with protein level validation were used to study drug MOA using conventional and transgenic murine models. We found a significant decrease in colonic and ileal naïve B and T cells and circulating gut-homing plasmablasts (β7+) in VDZ-treated patients, pointing to gut-associated lymphoid tissue (GALT) targeting by VDZ. Murine Peyer's patches (PP) demonstrated a significant loss cellularity associated with reduction in follicular B cells, including a unique population of epithelium-associated B cells, following anti-α4β7 antibody (mAb) administration. Photoconvertible (KikGR) mice unequivocally demonstrated impaired cellular entry into PPs in anti-α4β7 mAb treated mice. In VDZ-treated, but not anti-tumor necrosis factor-treated UC patients, lymphoid aggregate size was significantly reduced in treatment responders compared to non-responders, with an independent validation cohort further confirming these data. GALT targeting represents a novel MOA of α4β7-targeted therapies, with major implications for this therapeutic paradigm in UC, and for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Pablo Canales-Herrerias
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mathieu Uzzan
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Paris Est Créteil University UPEC, Assistance Publique-Hôpitaux de Paris (AP-HP), Henri Mondor Hospital, Gastroenterology department, Fédération Hospitalo-Universitaire TRUE InnovaTive theRapy for immUne disordErs, Créteil F-94010, France
| | - Akihiro Seki
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Bram Verstockt
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
- Translational Research in Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Alexandra Livanos
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fiona Raso
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alexandra Dunn
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Dai
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew Wang
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zainab Al-taie
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jerome Martin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nantes Université, CHU Nantes, Inserm, Centre de Recherche Translationelle en Transplantation et Immunologie, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Laboratoire d’Immunologie, CIMNA, Nantes, France
| | - Huaibin M. Ko
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Minami Tokuyama
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Tankelevich
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hadar Meringer
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francesca Cossarini
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Divya Jha
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Azra Krek
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John D. Paulsen
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M. Zuber Nakadar
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua Wong
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emma C. Erlich
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, USA
| | - Emily J. Onufer
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Beth A. Helmink
- Department of Surgery, Section of Surgical Oncology, Washington University School of Medicine, St. Louis, MO
| | - Keshav Sharma
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam Rosenstein
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Grace Chung
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Travis Dawson
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Andrea Cerutti
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Translational Clinical Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Jeremiah Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mayte Suarez-Farinas
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gwendalyn J. Randolph
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexandros D. Polydorides
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jean Frederic Colombel
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
13
|
Takeuchi T, Ohno H. IgA in human health and diseases: Potential regulator of commensal microbiota. Front Immunol 2022; 13:1024330. [PMID: 36439192 PMCID: PMC9685418 DOI: 10.3389/fimmu.2022.1024330] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/27/2022] [Indexed: 07/29/2023] Open
Abstract
Gut microbiota has extensive and tremendous impacts on human physiology and pathology. The regulation of microbiota is therefore a cardinal problem for the mutualistic relationship, as both microbial overgrowth and excessive immune reactions toward them could potentially be detrimental to host homeostasis. Growing evidence suggests that IgA, the most dominant secretory immunoglobulin in the intestine, regulates the colonization of commensal microbiota, and consequently, the microbiota-mediated intestinal and extra-intestinal diseases. In this review, we discuss the interactions between IgA and gut microbiota particularly relevant to human pathophysiology. We review current knowledge about how IgA regulates gut microbiota in humans and about the molecular mechanisms behind this interaction. We further discuss the potential role of IgA in regulating human diseases by extrapolating experimental findings, suggesting that IgA can be a future therapeutic strategy that functionally modulates gut microbiota.
Collapse
Affiliation(s)
- Tadashi Takeuchi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| |
Collapse
|
14
|
Gribonika I, Strömberg A, Lebrero-Fernandez C, Schön K, Moon J, Bemark M, Lycke N. Peyer's patch T H17 cells are dispensable for gut IgA responses to oral immunization. Sci Immunol 2022; 7:eabc5500. [PMID: 35776804 DOI: 10.1126/sciimmunol.abc5500] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
T helper 17 (TH17) cells located at the Peyer's patch (PP) inductive site and at the lamina propria effector site of the intestinal immune system are responsive to both pathogenic and commensal bacteria. Their plasticity to convert into follicular helper T (TFH) cells has been proposed to be central to gut immunoglobulin A (IgA) responses. Here, we used an IL-17A fate reporter mouse and an MHC-II tetramer to analyze antigen-specific CD4+ T cell subsets and isolate them for single-cell RNA sequencing after oral immunization with cholera toxin and ovalbumin. We found a TFH-dominated response with only rare antigen-specific TH17 cells (<8%) in the PP. A clonotypic analysis provided little support that clonotypes were shared between TFH and TH17 cells, arguing against TH17 plasticity as a major contributor to TFH differentiation. Two mouse models of TH17 deficiency confirmed that gut IgA responses to oral immunization do not require TH17 cells, with CD4CreRorcfl/fl mice exhibiting normal germinal centers in PP and unperturbed total IgA production in the intestine.
Collapse
Affiliation(s)
- Inta Gribonika
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Anneli Strömberg
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Cristina Lebrero-Fernandez
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Karin Schön
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - James Moon
- Center for Immunology and Inflammatory Diseases and Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Mats Bemark
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Immunology and Transfusion Medicine, Gothenburg, Sweden
| | - Nils Lycke
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
15
|
Bos A, van Egmond M, Mebius R. The role of retinoic acid in the production of immunoglobulin A. Mucosal Immunol 2022; 15:562-572. [PMID: 35418672 DOI: 10.1038/s41385-022-00509-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/09/2022] [Accepted: 03/26/2022] [Indexed: 02/04/2023]
Abstract
Vitamin A and its derivative retinoic acid (RA) play important roles in the regulation of mucosal immunity. The effect of vitamin A metabolism on T lymphocyte immunity has been well documented, but its role in mucosal B lymphocyte regulation is less well described. Intestinal immunoglobulin A (IgA) is key in orchestrating a balanced gut microbiota composition. Here, we describe the contribution of RA to IgA class switching in tissues including the lamina propria, mesenteric lymph nodes, Peyer's patches and isolated lymphoid follicles. RA can either indirectly skew T cells or directly affect B cell differentiation. IgA levels in healthy individuals are under the control of the metabolism of vitamin A, providing a steady supply of RA. However, IgA levels are altered in inflammatory bowel disease patients, making control of the metabolism of vitamin A a potential therapeutic target. Thus, dietary vitamin A is a key player in regulating IgA production within the intestine, acting via multiple immunological pathways.
Collapse
Affiliation(s)
- Amelie Bos
- Amsterdam UMC, Department of Molecular Cell Biology and Immunology, Research Institute of Amsterdam Institute for Infection and Immunity, Vrije Universiteit, Amsterdam, The Netherlands
| | - Marjolein van Egmond
- Amsterdam UMC, Department of Molecular Cell Biology and Immunology, Research Institute of Amsterdam Institute for Infection and Immunity, Vrije Universiteit, Amsterdam, The Netherlands.,Amsterdam UMC, Department of Surgery, Research Institute of Amsterdam Institute for Infection and Immunity, Vrije Universiteit, Amsterdam, The Netherlands
| | - Reina Mebius
- Amsterdam UMC, Department of Molecular Cell Biology and Immunology, Research Institute of Amsterdam Institute for Infection and Immunity, Vrije Universiteit, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Boothby MR, Brookens SK, Raybuck AL, Cho SH. Supplying the trip to antibody production-nutrients, signaling, and the programming of cellular metabolism in the mature B lineage. Cell Mol Immunol 2022; 19:352-369. [PMID: 34782762 PMCID: PMC8591438 DOI: 10.1038/s41423-021-00782-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/16/2021] [Indexed: 12/26/2022] Open
Abstract
The COVID pandemic has refreshed and expanded recognition of the vital role that sustained antibody (Ab) secretion plays in our immune defenses against microbes and of the importance of vaccines that elicit Ab protection against infection. With this backdrop, it is especially timely to review aspects of the molecular programming that govern how the cells that secrete Abs arise, persist, and meet the challenge of secreting vast amounts of these glycoproteins. Whereas plasmablasts and plasma cells (PCs) are the primary sources of secreted Abs, the process leading to the existence of these cell types starts with naive B lymphocytes that proliferate and differentiate toward several potential fates. At each step, cells reside in specific microenvironments in which they not only receive signals from cytokines and other cell surface receptors but also draw on the interstitium for nutrients. Nutrients in turn influence flux through intermediary metabolism and sensor enzymes that regulate gene transcription, translation, and metabolism. This review will focus on nutrient supply and how sensor mechanisms influence distinct cellular stages that lead to PCs and their adaptations as factories dedicated to Ab secretion. Salient findings of this group and others, sometimes exhibiting differences, will be summarized with regard to the journey to a distinctive metabolic program in PCs.
Collapse
Affiliation(s)
- Mark R Boothby
- Department of Pathology, Microbiology & Immunology, Molecular Pathogenesis Division, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Medicine, Rheumatology & Immunology Division, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Cancer Biology Program, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Institute of Infection, Inflammation, and Immunology, Nashville, TN, 37232, USA.
| | - Shawna K Brookens
- Department of Pathology, Microbiology & Immunology, Molecular Pathogenesis Division, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Cancer Biology Program, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ariel L Raybuck
- Department of Pathology, Microbiology & Immunology, Molecular Pathogenesis Division, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sung Hoon Cho
- Department of Pathology, Microbiology & Immunology, Molecular Pathogenesis Division, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Institute of Infection, Inflammation, and Immunology, Nashville, TN, 37232, USA
| |
Collapse
|
17
|
Fleming A, Castro‐Dopico T, Clatworthy MR. B cell class switching in intestinal immunity in health and disease. Scand J Immunol 2022; 95:e13139. [PMID: 34978077 PMCID: PMC9285483 DOI: 10.1111/sji.13139] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/31/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022]
Abstract
The gastrointestinal tract is colonized by trillions of commensal microorganisms that collectively form the microbiome and make essential contributions to organism homeostasis. The intestinal immune system must tolerate these beneficial commensals, whilst preventing pathogenic organisms from systemic spread. Humoral immunity plays a key role in this process, with large quantities of immunoglobulin (Ig)A secreted into the lumen on a daily basis, regulating the microbiome and preventing bacteria from encroaching on the epithelium. However, there is an increasing appreciation of the role of IgG antibodies in intestinal immunity, including beneficial effects in neonatal immune development, pathogen and tumour resistance, but also of pathological effects in driving chronic inflammation in inflammatory bowel disease (IBD). These antibody isotypes differ in effector function, with IgG exhibiting more proinflammatory capabilities compared with IgA. Therefore, the process that leads to the generation of different antibody isotypes, class-switch recombination (CSR), requires careful regulation and is orchestrated by the immunological cues generated by the prevalent local challenge. In general, an initiating signal such as CD40 ligation on B cells leads to the induction of activation-induced cytidine deaminase (AID), but a second cytokine-mediated signal determines which Ig heavy chain is expressed. Whilst the cytokines driving intestinal IgA responses are well-studied, there is less clarity on how IgG responses are generated in the intestine, and how these cues might become dysfunctional in IBD. Here, we review the key mechanisms regulating class switching to IgA vs IgG in the intestine, processes that could be therapeutically manipulated in infection and IBD.
Collapse
Affiliation(s)
- Aaron Fleming
- Molecular Immunity UnitDepartment of MedicineCambridge Institute of Therapeutic Immunology and Infectious DiseasesUniversity of CambridgeCambridgeUK
| | - Tomas Castro‐Dopico
- Molecular Immunity UnitDepartment of MedicineCambridge Institute of Therapeutic Immunology and Infectious DiseasesUniversity of CambridgeCambridgeUK
- The Francis Crick InstituteLondonUK
| | - Menna R. Clatworthy
- Molecular Immunity UnitDepartment of MedicineCambridge Institute of Therapeutic Immunology and Infectious DiseasesUniversity of CambridgeCambridgeUK
- Cellular GeneticsWellcome Trust Sanger InstituteHinxtonUK
- NIHR Cambridge Biomedical Research CentreCambridgeUK
| |
Collapse
|
18
|
Bondt A, Dingess KA, Hoek M, van Rijswijck DMH, Heck AJR. A Direct MS-Based Approach to Profile Human Milk Secretory Immunoglobulin A (IgA1) Reveals Donor-Specific Clonal Repertoires With High Longitudinal Stability. Front Immunol 2021; 12:789748. [PMID: 34938298 PMCID: PMC8685336 DOI: 10.3389/fimmu.2021.789748] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/17/2021] [Indexed: 12/29/2022] Open
Abstract
Recently, a mass spectrometry-based approach was introduced to directly assess the IgG1 immunoglobulin clonal repertoires in plasma. Here we expanded upon this approach by describing a mass spectrometry-based technique to assess specifically the clonal repertoire of another important class of immunoglobulin molecules, IgA1, and show it is efficiently and robustly applicable to either milk or plasma samples. Focusing on two individual healthy donors, whose milk was sampled longitudinally during the first 16 weeks of lactation, we demonstrate that the total repertoire of milk sIgA1 is dominated by only 50-500 clones, even though the human body theoretically can generate several orders of magnitude more clones. We show that in each donor the sIgA1 repertoire only changes marginally and quite gradually over the monitored 16-week period of lactation. Furthermore, the observed overlap in clonal repertoires between the two individual donors is close to non-existent. Mothers provide protection to their newborn infants directly by the transfer of antibodies via breastfeeding. The approach introduced here, can be used to visualize the clonal repertoire transferred from mother to infant and to detect changes in-time in that repertoire adapting to changes in maternal physiology.
Collapse
Affiliation(s)
- Albert Bondt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Kelly A Dingess
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Max Hoek
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Danique M H van Rijswijck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Netherlands Proteomics Center, Utrecht, Netherlands
| |
Collapse
|
19
|
Response kinetics of different classes of antibodies to SARS-CoV2 infection in the Japanese population: The IgA and IgG titers increased earlier than the IgM titers. Int Immunopharmacol 2021; 103:108491. [PMID: 34954559 PMCID: PMC8687758 DOI: 10.1016/j.intimp.2021.108491] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022]
Abstract
To better understand the immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in individuals with COVID-19, it is important to investigate the kinetics of the antibody responses and their associations with the clinical course in different populations, since there seem to be considerable differences between Western and Asian populations in the clinical features and spread of COVID-19. In this study, we serially measured the serum titers of IgM, IgG and IgA antibodies generated against the nucleocapsid protein (NCP), S1 subunit of the spike protein (S1), and receptor-binding domain in the S1 subunit (RBD) of SARS-CoV-2 in Japanese individuals with COVID-19. Among the IgM, IgG, and IgA antibodies, IgA antibodies against all of the aforementioned viral proteins were the first to appear after the infection, and IgG and/or IgA seroconversion often preceded IgM seroconversion. In regard to the timeline of the antibody responses to the different viral proteins (NCP, S1 and RBD), IgA against NCP appeared than IgA against S1 or RBD, while IgM and IgG against S1 appeared earlier than IgM/IgG against NCP or RBD. The IgG responses to all three viral proteins and responses of all three antibody classes to S1 and RBD were sustained for longer durations than the IgA/IgM responses to all three viral proteins and responses of all three antibody classes to NCP, respectively. The seroconversion of IgA against NCP occurred later and less frequently in patients with mild COVID-19. These results suggest possible differences in the antibody responses to SARS-CoV-2 antigens between the Japanese and Western populations.
Collapse
|
20
|
Takeuchi T, Ohno H. Reciprocal regulation of IgA and the gut microbiota: a key mutualism in the intestine. Int Immunol 2021; 33:781-786. [PMID: 34346497 DOI: 10.1093/intimm/dxab049] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/02/2021] [Indexed: 12/17/2022] Open
Abstract
The mammalian intestine is home to trillions of microbes, and their colonization contributes to host physiology through the production of indispensable metabolites and competition against pathogens. However, it is also important to balance this symbiotic relationship, as overgrowth and translocation of microbes could trigger a fatal infection. IgA is the major immunoglobulin class produced and secreted in the intestine and is considered to play a pivotal role in maintaining homeostasis. In this review, we summarize recent studies exploring the interactions between IgA and the gut microbiota, and explain how different types of IgA could coexist to regulate the gut microbiota. In particular, we discuss two important aspects of IgA in controlling the gut microbes: function and specificity. Differences in these two aspects appear attributable to how IgA is induced and are associated with the functions of IgA as well. Together, our review delineates a recent understanding of IgA-microbiome interactions and proposes a future direction to clarify its complexity.
Collapse
Affiliation(s)
- Tadashi Takeuchi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Intestinal Ecosystem, Yokohama, Japan.,Department of Microbiology and Immunology, Keio University Graduate School of Medicine, Tokyo, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Intestinal Ecosystem, Yokohama, Japan
| |
Collapse
|
21
|
Bemark M, Angeletti D. Know your enemy or find your friend?-Induction of IgA at mucosal surfaces. Immunol Rev 2021; 303:83-102. [PMID: 34331314 PMCID: PMC7612940 DOI: 10.1111/imr.13014] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/15/2022]
Abstract
Most antibodies produced in the body are of the IgA class. The dominant cell population producing them are plasma cells within the lamina propria of the gastrointestinal tract, but many IgA-producing cells are also found in the airways, within mammary tissues, the urogenital tract and inside the bone marrow. Most IgA antibodies are transported into the lumen by epithelial cells as part of the mucosal secretions, but they are also present in serum and other body fluids. A large part of the commensal microbiota in the gut is covered with IgA antibodies, and it has been demonstrated that this plays a role in maintaining a healthy balance between the host and the bacteria. However, IgA antibodies also play important roles in neutralizing pathogens in the gastrointestinal tract and the upper airways. The distinction between the two roles of IgA - protective and balance-maintaining - not only has implications on function but also on how the production is regulated. Here, we discuss these issues with a special focus on gut and airways.
Collapse
Affiliation(s)
- Mats Bemark
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Davide Angeletti
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
22
|
Wishnie AJ, Chwat-Edelstein T, Attaway M, Vuong BQ. BCR Affinity Influences T-B Interactions and B Cell Development in Secondary Lymphoid Organs. Front Immunol 2021; 12:703918. [PMID: 34381455 PMCID: PMC8350505 DOI: 10.3389/fimmu.2021.703918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 07/07/2021] [Indexed: 11/13/2022] Open
Abstract
B cells produce high-affinity immunoglobulins (Igs), or antibodies, to eliminate foreign pathogens. Mature, naïve B cells expressing an antigen-specific cell surface Ig, or B cell receptor (BCR), are directed toward either an extrafollicular (EF) or germinal center (GC) response upon antigen binding. B cell interactions with CD4+ pre-T follicular helper (pre-Tfh) cells at the T-B border and effector Tfh cells in the B cell follicle and GC control B cell development in response to antigen. Here, we review recent studies demonstrating the role of B cell receptor (BCR) affinity in modulating T-B interactions and the subsequent differentiation of B cells in the EF and GC response. Overall, these studies demonstrate that B cells expressing high affinity BCRs preferentially differentiate into antibody secreting cells (ASCs) while those expressing low affinity BCRs undergo further affinity maturation or differentiate into memory B cells (MBCs).
Collapse
Affiliation(s)
- Alec J Wishnie
- Biology PhD Program, Graduate Center, The City University of New York, New York, NY, United States.,Department of Biology, The City College of New York, New York, NY, United States
| | - Tzippora Chwat-Edelstein
- Department of Biology, The City College of New York, New York, NY, United States.,Macaulay Honors College, New York, NY, United States
| | - Mary Attaway
- Department of Biology, The City College of New York, New York, NY, United States
| | - Bao Q Vuong
- Biology PhD Program, Graduate Center, The City University of New York, New York, NY, United States.,Department of Biology, The City College of New York, New York, NY, United States
| |
Collapse
|
23
|
Human gut-associated lymphoid tissues (GALT); diversity, structure, and function. Mucosal Immunol 2021; 14:793-802. [PMID: 33753873 DOI: 10.1038/s41385-021-00389-4] [Citation(s) in RCA: 211] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 02/07/2023]
Abstract
Gut-associated lymphoid tissues (GALT) are the key antigen sampling and adaptive immune inductive sites within the intestinal wall. Human GALT includes the multi-follicular Peyer's patches of the ileum, the vermiform appendix, and the numerous isolated lymphoid follicles (ILF) which are distributed along the length of the intestine. Our current understanding of GALT diversity and function derives primarily from studies in mice, and the relevance of many of these findings to human GALT remains unclear. Here we review our current understanding of human GALT diversity, structure, and composition as well as their potential for regulating intestinal immune responses during homeostasis and inflammatory bowel disease (IBD). Finally, we outline some key remaining questions regarding human GALT, the answers to which will advance our understanding of intestinal immune responses and provide potential opportunities to improve the treatment of intestinal diseases.
Collapse
|
24
|
Elsner RA, Shlomchik MJ. Germinal Center and Extrafollicular B Cell Responses in Vaccination, Immunity, and Autoimmunity. Immunity 2021; 53:1136-1150. [PMID: 33326765 DOI: 10.1016/j.immuni.2020.11.006] [Citation(s) in RCA: 300] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/19/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Activated B cells participate in either extrafollicular (EF) or germinal center (GC) responses. Canonical responses are composed of a short wave of plasmablasts (PBs) arising from EF sites, followed by GC producing somatically mutated memory B cells (MBC) and long-lived plasma cells. However, somatic hypermutation (SHM) and affinity maturation can take place at both sites, and a substantial fraction of MBC are produced prior to GC formation. Infection responses range from GC responses that persist for months to persistent EF responses with dominant suppression of GCs. Here, we review the current understanding of the functional output of EF and GC responses and the molecular switches promoting them. We discuss the signals that regulate the magnitude and duration of these responses, and outline gaps in knowledge and important areas of inquiry. Understanding such molecular switches will be critical for vaccine development, interpretation of vaccine efficacy and the treatment for autoimmune diseases.
Collapse
Affiliation(s)
- Rebecca A Elsner
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Mark J Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15216, USA.
| |
Collapse
|
25
|
Biram A, Winter E, Denton AE, Zaretsky I, Dassa B, Bemark M, Linterman MA, Yaari G, Shulman Z. B Cell Diversification Is Uncoupled from SAP-Mediated Selection Forces in Chronic Germinal Centers within Peyer's Patches. Cell Rep 2021; 30:1910-1922.e5. [PMID: 32049020 PMCID: PMC7016508 DOI: 10.1016/j.celrep.2020.01.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/24/2019] [Accepted: 01/08/2020] [Indexed: 12/17/2022] Open
Abstract
Antibodies secreted within the intestinal tract provide protection from the invasion of microbes into the host tissues. Germinal center (GC) formation in lymph nodes and spleen strictly requires SLAM-associated protein (SAP)-mediated T cell functions; however, it is not known whether this mechanism plays a similar role in mucosal-associated lymphoid tissues. Here, we find that in Peyer’s patches (PPs), SAP-mediated T cell help is required for promoting B cell selection in GCs, but not for clonal diversification. PPs of SAP-deficient mice host chronic GCs that are absent in T cell-deficient mice. GC B cells in SAP-deficient mice express AID and Bcl6 and generate plasma cells in proportion to the GC size. Single-cell IgA sequencing analysis reveals that these mice host few diversified clones that were subjected to mild selection forces. These findings demonstrate that T cell-derived help to B cells in PPs includes SAP-dependent and SAP-independent functions. Chronic germinal centers in Peyer’s patches are formed in SAP-deficient mice SAP-independent germinal centers arise in response to influenza infection Few highly diversified clones dominate the SAP-independent germinal centers Germinal center B cells in SAP-deficient mice are subjected to mild selection forces
Collapse
Affiliation(s)
- Adi Biram
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eitan Winter
- Faculty of Engineering, Bar Ilan University, Ramat Gan 52900, Israel
| | - Alice E Denton
- Laboratory of Lymphocyte Signaling and Development, Babraham Institute, Cambridge CB22 3AT, UK
| | - Irina Zaretsky
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Bareket Dassa
- Department of Life Science Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Mats Bemark
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg SE-405 30, Sweden
| | - Michelle A Linterman
- Laboratory of Lymphocyte Signaling and Development, Babraham Institute, Cambridge CB22 3AT, UK
| | - Gur Yaari
- Faculty of Engineering, Bar Ilan University, Ramat Gan 52900, Israel
| | - Ziv Shulman
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
26
|
Abstract
Among antibodies, IgA is unique because it has evolved to be secreted onto mucosal surfaces. The structure of IgA and the associated secretory component allow IgA to survive the highly proteolytic environment of mucosal surfaces but also substantially limit IgA's ability to activate effector functions on immune cells. Despite these characteristics, IgA is critical for both preventing enteric infections and shaping the local microbiome. IgA's function is determined by a distinct antigen-binding repertoire, composed of antibodies with a variety of specificities, from permissive polyspecificity to cross-reactivity to exquisite specificity to a single epitope, which act together to regulate intestinal bacteria. Development of the unique function and specificities of IgA is shaped by local cues provided by the gut-associated lymphoid tissue, driven by the constantly changing environment of the intestine and microbiota.
Collapse
Affiliation(s)
- Timothy W Hand
- R.K. Mellon Institute for Pediatric Research, Department of Pediatrics, Division of Infectious Diseases, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania 15224, USA;
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA;
| |
Collapse
|
27
|
Bai Y, Huang F, Zhang R, Ma Q, Dong L, Su D, Chi J, Zhang M. Longan pulp polysaccharide protects against cyclophosphamide-induced immunosuppression in mice by promoting intestinal secretory IgA synthesis. Food Funct 2021; 11:2738-2748. [PMID: 32175536 DOI: 10.1039/c9fo02780g] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This study aimed to explore the effect of longan pulp polysaccharide (LP) on the systemic immunity and intestinal mucosal immunity of immunosuppressive mice. The synthesis process and secretion of intestinal secretory IgA (SIgA) were investigated. Results showed that LP increased the thymus index, spleen index, and serum IgA level in cyclophosphamide (CTX)-treated mice. SIgA secretion in the intestinal lumen was increased by LP as well. The underlying mechanism comes down to the facts as follow: LP increased intestinal cytokines expression and TGFβRII that is associated with pathways of IgA class switch recombination (CSR). By improving protein expression of mucosal addressin cell-adhesion molecule-1 (MAdCAM-1) and integrin α4β7, LP was beneficial to gut homing of IgA+ plasma cells. LP increased IgA, polymeric immunoglobulin receptor (pIgR), and secretory component (SC) to fortify the SIgA secretion. This study suggested that moderate consumption of LP is helpful for improving systemic immunity and intestinal mucosal immunity via promotion of intestinal SIgA to strengthen the mucosal barrier.
Collapse
Affiliation(s)
- Yajuan Bai
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Fei Huang
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Ruifen Zhang
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Qin Ma
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Lihong Dong
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Dongxiao Su
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, PR China
| | - Jianwei Chi
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Mingwei Zhang
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| |
Collapse
|
28
|
Zhang B, Liu E, Gertie JA, Joseph J, Xu L, Pinker EY, Waizman DA, Catanzaro J, Hamza KH, Lahl K, Gowthaman U, Eisenbarth SC. Divergent T follicular helper cell requirement for IgA and IgE production to peanut during allergic sensitization. Sci Immunol 2020; 5:5/47/eaay2754. [PMID: 32385053 DOI: 10.1126/sciimmunol.aay2754] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 03/24/2020] [Indexed: 12/14/2022]
Abstract
Immunoglobulin A (IgA) is the dominant antibody isotype in the gut and has been shown to regulate microbiota. Mucosal IgA is also widely believed to prevent food allergens from penetrating the gut lining. Even though recent work has elucidated how bacteria-reactive IgA is induced, little is known about how IgA to food antigens is regulated. Although IgA is presumed to be induced in a healthy gut at steady state via dietary exposure, our data do not support this premise. We found that daily food exposure only induced low-level, cross-reactive IgA in a minority of mice. In contrast, induction of significant levels of peanut-specific IgA strictly required a mucosal adjuvant. Although induction of peanut-specific IgA required T cells and CD40L, it was T follicular helper (TFH) cell, germinal center, and T follicular regulatory (TFR) cell-independent. In contrast, IgG1 and IgE production to peanut required TFH cells. These data suggest an alternative paradigm in which the cellular mechanism of IgA production to food antigens is distinct from IgE and IgG1. We developed an equivalent assay to study this process in stool samples from healthy, nonallergic humans, which revealed substantial levels of peanut-specific IgA that were stable over time. Similar to mice, patients with loss of CD40L function had impaired titers of gut peanut-specific IgA. This work challenges two widely believed but untested paradigms about antibody production to dietary antigens: (i) the steady state/tolerogenic response to food antigens includes IgA production and (ii) TFH cells drive food-specific gut IgA.
Collapse
Affiliation(s)
- Biyan Zhang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Elise Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA.,Section of Rheumatology, Allergy and Immunology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Jake A Gertie
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Julie Joseph
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Lan Xu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Elisha Y Pinker
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Columbia University, New York, NY 10027, USA
| | - Daniel A Waizman
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Jason Catanzaro
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06520, USA.,Section of Pulmonology, Allergy, Immunology and Sleep Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Kedir Hussen Hamza
- Department for Experimental Medicine, Immunology Section, Lund University, Lund 221 84, Sweden
| | - Katharina Lahl
- Department for Experimental Medicine, Immunology Section, Lund University, Lund 221 84, Sweden.,Division of Biopharma, Institute for Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Uthaman Gowthaman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA. .,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA.,Section of Rheumatology, Allergy and Immunology, Yale University School of Medicine, New Haven, CT, 06520, USA
| |
Collapse
|
29
|
Pietrzak B, Tomela K, Olejnik-Schmidt A, Mackiewicz A, Schmidt M. Secretory IgA in Intestinal Mucosal Secretions as an Adaptive Barrier against Microbial Cells. Int J Mol Sci 2020; 21:ijms21239254. [PMID: 33291586 PMCID: PMC7731431 DOI: 10.3390/ijms21239254] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Secretory IgA (SIgA) is the dominant antibody class in mucosal secretions. The majority of plasma cells producing IgA are located within mucosal membranes lining the intestines. SIgA protects against the adhesion of pathogens and their penetration into the intestinal barrier. Moreover, SIgA regulates gut microbiota composition and provides intestinal homeostasis. In this review, we present mechanisms of SIgA generation: T cell-dependent and -independent; in different non-organized and organized lymphoid structures in intestinal lamina propria (i.e., Peyer’s patches and isolated lymphoid follicles). We also summarize recent advances in understanding of SIgA functions in intestinal mucosal secretions with focus on its role in regulating gut microbiota composition and generation of tolerogenic responses toward its members.
Collapse
Affiliation(s)
- Bernadeta Pietrzak
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego, 60-627 Poznań, Poland;
- Correspondence: (B.P.); (M.S.)
| | - Katarzyna Tomela
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 8 Rokietnicka Street, 60-806 Poznań, Poland; (K.T.); (A.M.)
| | - Agnieszka Olejnik-Schmidt
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego, 60-627 Poznań, Poland;
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 8 Rokietnicka Street, 60-806 Poznań, Poland; (K.T.); (A.M.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznań, Poland
| | - Marcin Schmidt
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego, 60-627 Poznań, Poland;
- Correspondence: (B.P.); (M.S.)
| |
Collapse
|
30
|
Tang T, Cheng X, Truong B, Sun L, Yang X, Wang H. Molecular basis and therapeutic implications of CD40/CD40L immune checkpoint. Pharmacol Ther 2020; 219:107709. [PMID: 33091428 DOI: 10.1016/j.pharmthera.2020.107709] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/15/2020] [Indexed: 12/22/2022]
Abstract
The CD40 receptor and its ligand CD40L is one of the most critical molecular pairs of the stimulatory immune checkpoints. Both CD40 and CD40L have a membrane form and a soluble form generated by proteolytic cleavage or alternative splicing. CD40 and CD40L are widely expressed in various types of cells, among which B cells and myeloid cells constitutively express high levels of CD40, and T cells and platelets express high levels of CD40L upon activation. CD40L self-assembles into functional trimers which induce CD40 trimerization and downstream signaling. The canonical CD40/CD40L signaling is mediated by recruitment of TRAFs and NF-κB activation, which is supplemented by signal pathways such as PI3K/AKT, MAPKs and JAK3/STATs. CD40/CD40L immune checkpoint leads to activation of both innate and adaptive immune cells via two-way signaling. CD40/CD40L interaction also participates in regulating thrombosis, tissue inflammation, hematopoiesis and tumor cell fate. Because of its essential role in immune activation, CD40/CD40L interaction has been regarded as an attractive immunotherapy target. In recent years, significant advance has been made in CD40/CD40L-targeted therapy. Various types of agents, including agonistic/antagonistic monoclonal antibodies, cellular vaccines, adenoviral vectors and protein antagonist, have been developed and evaluated in early-stage clinical trials for treating malignancies, autoimmune diseases and allograft rejection. In general, these agents have demonstrated favorable safety and some of them show promising clinical efficacy. The mechanisms of benefits include immune cell activation and tumor cell lysis/apoptosis in malignancies, or immune cell inactivation in autoimmune diseases and allograft rejection. This review provides a comprehensive overview of the structure, processing, cellular expression pattern, signaling and effector function of CD40/CD40L checkpoint molecules. In addition, we summarize the progress, targeted diseases and outcomes of current ongoing and completed clinical trials of CD40/CD40L-targeted therapy.
Collapse
Affiliation(s)
- TingTing Tang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Billy Truong
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - LiZhe Sun
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Department of Cardiovascular Medicine, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - XiaoFeng Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Hong Wang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA; Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA.
| |
Collapse
|
31
|
Biram A, Shulman Z. T cell help to B cells: Cognate and atypical interactions in peripheral and intestinal lymphoid tissues. Immunol Rev 2020; 296:36-47. [PMID: 32557712 DOI: 10.1111/imr.12890] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
Enduring immunity against harmful pathogens depends on the generation of immunological memory. Serum immunoglobulins are constantly secreted by long-lived antibody-producing cells, which provide extended protection from recurrent exposures. These cells originate mainly from germinal center structures, wherein B cells introduce mutations to their immunoglobulin genes followed by affinity-based selection. Generation of high-affinity antibodies relies on physical contacts between T and B cells, a process that facilitates the delivery of fate decision signals. T-B cellular engagements are mediated through interactions between the T cell receptor and its cognate peptide presented on B cell major histocompatibility class II molecules. Here, we describe the cellular and molecular aspects of these cognate T-B interactions, and highlight exceptional cases, especially those arising at intestinal lymphoid organs, at which T cells provide help to B cells in an atypical manner, independent of T cell specificity.
Collapse
Affiliation(s)
- Adi Biram
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Shulman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
32
|
The Effects of Secretory IgA in the Mucosal Immune System. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2032057. [PMID: 31998782 PMCID: PMC6970489 DOI: 10.1155/2020/2032057] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/14/2019] [Indexed: 12/18/2022]
Abstract
Immunoglobulin A (IgA) is the most abundant antibody isotype in the mucosal immune system. Structurally, IgA in the mucosal surface is a polymeric structure, while serum IgA is monomeric. Secretory IgA (sIgA) is one of the polymeric IgAs composed of dimeric IgA, J chain, and secretory component (SC). Most of sIgAs were generated by gut and have effects in situ. Besides the function of “immune exclusion,” a nonspecific immune role, recent studies found it also played an important role in the specific immunity and immunoregulation. Thanks to the critical role of sIgA during the mucosal immune system homeostasis between commensal microorganisms and pathogens; it has been an important field exploring the relationship between sIgA and commensal microorganisms.
Collapse
|
33
|
Chen Z, Wholey WY, Hassani Najafabadi A, Moon JJ, Grigorova I, Chackerian B, Cheng W. Self-Antigens Displayed on Liposomal Nanoparticles above a Threshold of Epitope Density Elicit Class-Switched Autoreactive Antibodies Independent of T Cell Help. THE JOURNAL OF IMMUNOLOGY 2019; 204:335-347. [PMID: 31836655 DOI: 10.4049/jimmunol.1801677] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 11/11/2019] [Indexed: 12/12/2022]
Abstract
Epitope density has a profound impact on B cell responses to particulate Ags, the molecular mechanisms of which remain to be explored. To dissect the role of epitope density in this process, we have synthesized a series of liposomal particles, similar to the size of viruses, that display a model self-antigen peptide at defined surface densities. Immunization of C57BL/6J mice using these particles elicited both IgM and class-switched IgG1, IgG2b, and IgG3 autoreactive Abs that depended on the epitope density. In C57BL/6 gene knockout mice lacking either functional TCRs or MHC class II molecules on B cells, the liposomal particles also elicited IgM, IgG1, IgG2b, and IgG3 responses that were comparable in magnitudes to wild-type mice, suggesting that this B cell response was independent of cognate T cell help. Notably, the titer of the IgG in wild-type animals could be increased by more than 200-fold upon replacement of liposomes with bacteriophage Qβ virus-like particles that displayed the same self-antigen peptide at comparable epitope densities. This enhancement was lost almost completely in gene knockout mice lacking either TCRs or MHC class II molecules on B cells. In conclusion, epitope density above a threshold on particulate Ags can serve as a stand-alone signal to trigger secretion of autoreactive and class-switched IgG in vivo in the absence of cognate T cell help or any adjuvants. The extraordinary immunogenicity of Qβ viral-like particles relies, in large part, on their ability to effectively recruit T cell help after B cell activation.
Collapse
Affiliation(s)
- Zhilin Chen
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109
| | - Wei-Yun Wholey
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109
| | | | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Irina Grigorova
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Bryce Chackerian
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico, Albuquerque, NM 87131; and
| | - Wei Cheng
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109; .,Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
34
|
Tezuka H, Ohteki T. Regulation of IgA Production by Intestinal Dendritic Cells and Related Cells. Front Immunol 2019; 10:1891. [PMID: 31456802 PMCID: PMC6700333 DOI: 10.3389/fimmu.2019.01891] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/26/2019] [Indexed: 12/31/2022] Open
Abstract
The intestinal mucosa is a physiological barrier for most microbes, including both commensal bacteria and invading pathogens. Under homeostatic conditions, immunoglobulin A (IgA) is the major immunoglobulin isotype in the intestinal mucosa. Microbes stimulate the production of IgA, which controls bacterial translocation and neutralizes bacterial toxins at the intestinal mucosal surface. In the intestinal mucosa, dendritic cells (DCs), specialized antigen-presenting cells, regulate both T-cell-dependent (TD) and -independent (TI) immune responses. The intestinal DCs are a heterogeneous population that includes unique subsets that induce IgA synthesis in B cells. The characteristics of intestinal DCs are strongly influenced by the microenvironment, including the presence of commensal bacterial metabolites and epithelial cell-derived soluble factors. In this review, we summarize the ontogeny, classification, and function of intestinal DCs and how the intestinal microenvironment conditions DCs and their precursors to become the mucosal phenotype, in particular to regulate IgA production, after they arrive at the intestine. Understanding the mechanism of IgA synthesis could provide insights for designing effective mucosal vaccines.
Collapse
Affiliation(s)
- Hiroyuki Tezuka
- Department of Cellular Function Analysis, Research Promotion and Support Headquarters, Fujita Health University, Aichi, Japan
| | - Toshiaki Ohteki
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
35
|
Peyer’s patches contain abundant isotype-switched B cells with activated phenotypes and are inductive sites for T-independent anti-DNA IgA. Immunol Lett 2019; 211:53-59. [DOI: 10.1016/j.imlet.2019.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 12/19/2022]
|
36
|
Stebegg M, Silva-Cayetano A, Innocentin S, Jenkins TP, Cantacessi C, Gilbert C, Linterman MA. Heterochronic faecal transplantation boosts gut germinal centres in aged mice. Nat Commun 2019; 10:2443. [PMID: 31164642 PMCID: PMC6547660 DOI: 10.1038/s41467-019-10430-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/10/2019] [Indexed: 12/21/2022] Open
Abstract
Ageing is a complex multifactorial process associated with a plethora of disorders, which contribute significantly to morbidity worldwide. One of the organs significantly affected by age is the gut. Age-dependent changes of the gut-associated microbiome have been linked to increased frailty and systemic inflammation. This change in microbial composition with age occurs in parallel with a decline in function of the gut immune system; however, it is not clear whether there is a causal link between the two. Here we report that the defective germinal centre reaction in Peyer's patches of aged mice can be rescued by faecal transfers from younger adults into aged mice and by immunisations with cholera toxin, without affecting germinal centre reactions in peripheral lymph nodes. This demonstrates that the poor germinal centre reaction in aged animals is not irreversible, and that it is possible to improve this response in older individuals by providing appropriate stimuli.
Collapse
Affiliation(s)
- Marisa Stebegg
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Alyssa Silva-Cayetano
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Silvia Innocentin
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Timothy P Jenkins
- Department of Veterinary Medicine, Madingley Road, Cambridge, CB3 0ES, UK
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, Madingley Road, Cambridge, CB3 0ES, UK
| | - Colin Gilbert
- Biological Services Unit, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Michelle A Linterman
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
37
|
Komban RJ, Strömberg A, Biram A, Cervin J, Lebrero-Fernández C, Mabbott N, Yrlid U, Shulman Z, Bemark M, Lycke N. Activated Peyer's patch B cells sample antigen directly from M cells in the subepithelial dome. Nat Commun 2019; 10:2423. [PMID: 31160559 PMCID: PMC6547658 DOI: 10.1038/s41467-019-10144-w] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/23/2019] [Indexed: 01/13/2023] Open
Abstract
The germinal center (GC) reaction in Peyer's patches (PP) requires continuous access to antigens, but how this is achieved is not known. Here we show that activated antigen-specific CCR6+CCR1+GL7- B cells make close contact with M cells in the subepithelial dome (SED). Using in situ photoactivation analysis of antigen-specific SED B cells, we find migration of cells towards the GC. Following antigen injection into ligated intestinal loops containing PPs, 40% of antigen-specific SED B cells bind antigen within 2 h, whereas unspecifc cells do not, indicating B cell-receptor involvment. Antigen-loading is not observed in M cell-deficient mice, but is unperturbed in mice depleted of classical dendritic cells (DC). Thus, we report a M cell-B cell antigen-specific transporting pathway in PP that is independent of DC. We propose that this antigen transporting pathway has a critical role in gut IgA responses, and should be taken into account when developing mucosal vaccines.
Collapse
Affiliation(s)
- Rathan Joy Komban
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden
| | - Anneli Strömberg
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden
| | - Adi Biram
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Jakob Cervin
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden
| | - Cristina Lebrero-Fernández
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden
| | - Neil Mabbott
- The Roslin Institute, Edinburgh University, Edinburgh, EH25 9RG, Scotland
| | - Ulf Yrlid
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden
| | - Ziv Shulman
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Mats Bemark
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden.
| | - Nils Lycke
- Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, S405 30, Sweden.
| |
Collapse
|
38
|
Bagheri Y, Sanaei R, Yazdani R, Shekarabi M, Falak R, Mohammadi J, Abolhassani H, Aghamohammadi A. The Heterogeneous Pathogenesis of Selective Immunoglobulin A Deficiency. Int Arch Allergy Immunol 2019; 179:231-246. [DOI: 10.1159/000499044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/18/2019] [Indexed: 11/19/2022] Open
|
39
|
IgA Responses to Microbiota. Immunity 2019; 49:211-224. [PMID: 30134201 DOI: 10.1016/j.immuni.2018.08.011] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/03/2018] [Accepted: 08/06/2018] [Indexed: 12/12/2022]
Abstract
Various immune mechanisms are deployed in the mucosa to confront the immense diversity of resident bacteria. A substantial fraction of the commensal microbiota is coated with immunoglobulin A (IgA) antibodies, and recent findings have established the identities of these bacteria under homeostatic and disease conditions. Here we review the current understanding of IgA biology, and present a framework wherein two distinct types of humoral immunity coexist in the gastrointestinal mucosa. Homeostatic IgA responses employ a polyreactive repertoire to bind a broad but taxonomically distinct subset of microbiota. In contrast, mucosal pathogens and vaccines elicit high-affinity, T cell-dependent antibody responses. This model raises fundamental questions including how polyreactive IgA specificities are generated, how these antibodies exert effector functions, and how they exist together with other immune responses during homeostasis and disease.
Collapse
|
40
|
Stebegg M, Kumar SD, Silva-Cayetano A, Fonseca VR, Linterman MA, Graca L. Regulation of the Germinal Center Response. Front Immunol 2018; 9:2469. [PMID: 30410492 PMCID: PMC6209676 DOI: 10.3389/fimmu.2018.02469] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/05/2018] [Indexed: 12/24/2022] Open
Abstract
The germinal center (GC) is a specialized microstructure that forms in secondary lymphoid tissues, producing long-lived antibody secreting plasma cells and memory B cells, which can provide protection against reinfection. Within the GC, B cells undergo somatic mutation of the genes encoding their B cell receptors which, following successful selection, can lead to the emergence of B cell clones that bind antigen with high affinity. However, this mutation process can also be dangerous, as it can create autoreactive clones that can cause autoimmunity. Because of this, regulation of GC reactions is critical to ensure high affinity antibody production and to enforce self-tolerance by avoiding emergence of autoreactive B cell clones. A productive GC response requires the collaboration of multiple cell types. The stromal cell network orchestrates GC cell dynamics by controlling antigen delivery and cell trafficking. T follicular helper (Tfh) cells provide specialized help to GC B cells through cognate T-B cell interactions while Foxp3+ T follicular regulatory (Tfr) cells are key mediators of GC regulation. However, regulation of GC responses is not a simple outcome of Tfh/Tfr balance, but also involves the contribution of other cell types to modulate the GC microenvironment and to avoid autoimmunity. Thus, the regulation of the GC is complex, and occurs at multiple levels. In this review we outline recent developments in the biology of cell subsets involved in the regulation of GC reactions, in both secondary lymphoid tissues, and Peyer's patches (PPs). We discuss the mechanisms which enable the generation of potent protective humoral immunity whilst GC-derived autoimmunity is avoided.
Collapse
Affiliation(s)
| | - Saumya D Kumar
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | - Valter R Fonseca
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Centro Hospitalar Lisboa Norte-Hospital de Santa Maria, Lisbon, Portugal
| | | | - Luis Graca
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto Gulbenkian de Ciência, Oeiras, Portugal
| |
Collapse
|
41
|
Iversen R, Snir O, Stensland M, Kroll JE, Steinsbø Ø, Korponay-Szabó IR, Lundin KEA, de Souza GA, Sollid LM. Strong Clonal Relatedness between Serum and Gut IgA despite Different Plasma Cell Origins. Cell Rep 2018; 20:2357-2367. [PMID: 28877470 PMCID: PMC5603730 DOI: 10.1016/j.celrep.2017.08.036] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/14/2017] [Accepted: 08/07/2017] [Indexed: 01/02/2023] Open
Abstract
Mucosal antigens induce generation of lamina propria plasma cells (PCs) that secrete dimeric immunoglobulin A (IgA) destined for transport across the epithelium. In addition, blood contains monomeric IgA. To study the relationship between mucosal and systemic antibody responses, we took advantage of celiac disease patient samples for isolation of gut PCs as well as serum IgA and IgG reactive with a gluten-derived peptide or the autoantigen transglutaminase 2. Proteomic analysis of serum IgA revealed antigen-specific V-gene preferences, which matched those found in gut PCs. Further, gut PC CDR-H3 sequences were abundant in serum IgA but also detectable in serum IgG. Our data indicate that the same B cell clones that give rise to gut PCs also contribute to the serum antibody pool. However, serum IgA antibodies had a molecular composition distinct from that of IgA antibodies secreted in the gut, suggesting that individual B cell clones give rise to different PC populations.
Collapse
Affiliation(s)
- Rasmus Iversen
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, NO-0372 Oslo, Norway
| | - Omri Snir
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, NO-0372 Oslo, Norway
| | - Maria Stensland
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, NO-0372 Oslo, Norway; Proteomics Core Facility, Oslo University Hospital-Rikshospitalet, NO-0372 Oslo, Norway
| | - José E Kroll
- Brain Institute, Federal University of Rio Grande do Norte, RN 59056-450 Natal, Brazil
| | - Øyvind Steinsbø
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, NO-0372 Oslo, Norway
| | | | - Knut E A Lundin
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, NO-0372 Oslo, Norway; Department of Gastroenterology, Oslo University Hospital-Rikshospitalet, NO-0372 Oslo, Norway; KG Jebsen Coeliac Disease Research Centre, University of Oslo, NO-0372 Oslo, Norway
| | - Gustavo A de Souza
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, NO-0372 Oslo, Norway; Proteomics Core Facility, Oslo University Hospital-Rikshospitalet, NO-0372 Oslo, Norway; Brain Institute, Federal University of Rio Grande do Norte, RN 59056-450 Natal, Brazil
| | - Ludvig M Sollid
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, NO-0372 Oslo, Norway; KG Jebsen Coeliac Disease Research Centre, University of Oslo, NO-0372 Oslo, Norway.
| |
Collapse
|
42
|
Liang CC, Li CS, Weng IC, Chen HY, Lu HH, Huang CC, Liu FT. Galectin-9 Is Critical for Mucosal Adaptive Immunity through the T Helper 17-IgA Axis. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1225-1235. [PMID: 29458010 DOI: 10.1016/j.ajpath.2018.01.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/17/2018] [Accepted: 01/26/2018] [Indexed: 01/20/2023]
Abstract
Impairment of the intestinal mucosal immunity significantly increases the risk of acute and chronic diseases. IgA plays a major role in humoral mucosal immunity to provide protection against pathogens and toxins in the gut. Here, we investigated the role of endogenous galectin-9, a tandem repeat-type β-galactoside-binding protein, in intestinal mucosal immunity. By mucosal immunization of Lgals9-/- and littermate control mice, it was found that lack of galectin-9 impaired mucosal antigen-specific IgA response in the gut. Moreover, Lgals9-/- mice were more susceptible to developing watery diarrhea and more prone to death in response to high-dose cholera toxin. The results indicate the importance of galectin-9 in modulating intestinal adaptive immunity. Furthermore, bone marrow chimera mice were established, and galectin-9 in hematopoietic cells was found to be critical for adaptive IgA response. In addition, immunized Lgals9-/- mice exhibited lower expression of Il17 and fewer T helper 17 (Th17) cells in the lamina propria, implying that the Th17-IgA axis is involved in this mechanism. Taken together, these findings suggest that galectin-9 plays a role in mucosal adaptive immunity through the Th17-IgA axis. By manipulating the expression or activity of galectin-9, intestinal mucosal immune response can be altered and may benefit the development of mucosal vaccination.
Collapse
Affiliation(s)
- Chih-Chia Liang
- Ph.D. Program in Translational Medicine, China Medical University and Academia Sinica, Taichung, Taiwan; Department of Medicine, College of Medicine, China Medical University, Taichung, Taiwan; Division of Nephrology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chi-Shan Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - I-Chun Weng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Huan-Yuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsueh-Han Lu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chiu-Ching Huang
- Ph.D. Program in Translational Medicine, China Medical University and Academia Sinica, Taichung, Taiwan; Department of Medicine, College of Medicine, China Medical University, Taichung, Taiwan; Division of Nephrology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Fu-Tong Liu
- Ph.D. Program in Translational Medicine, China Medical University and Academia Sinica, Taichung, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California.
| |
Collapse
|
43
|
Macpherson AJ, Yilmaz B, Limenitakis JP, Ganal-Vonarburg SC. IgA Function in Relation to the Intestinal Microbiota. Annu Rev Immunol 2018; 36:359-381. [PMID: 29400985 DOI: 10.1146/annurev-immunol-042617-053238] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
IgA is the dominant immunoglobulin isotype produced in mammals, largely secreted across the intestinal mucosal surface. Although induction of IgA has been a hallmark feature of microbiota colonization following colonization in germ-free animals, until recently appreciation of the function of IgA in host-microbial mutualism has depended mainly on indirect evidence of alterations in microbiota composition or penetration of microbes in the absence of somatic mutations in IgA (or compensatory IgM). Highly parallel sequencing techniques that enable high-resolution analysis of either microbial consortia or IgA sequence diversity are now giving us new perspectives on selective targeting of microbial taxa and the trajectory of IgA diversification according to induction mechanisms, between different individuals and over time. The prospects are to link the range of diversified IgA clonotypes to specific antigenic functions in modulating the microbiota composition, position and metabolism to ensure host mutualism.
Collapse
Affiliation(s)
- Andrew J Macpherson
- Maurice Müller Laboratories (Department of Biomedical Research), University of Bern, 3008 Bern, Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital, 3010 Bern, Switzerland;
| | - Bahtiyar Yilmaz
- Maurice Müller Laboratories (Department of Biomedical Research), University of Bern, 3008 Bern, Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital, 3010 Bern, Switzerland;
| | - Julien P Limenitakis
- Maurice Müller Laboratories (Department of Biomedical Research), University of Bern, 3008 Bern, Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital, 3010 Bern, Switzerland;
| | - Stephanie C Ganal-Vonarburg
- Maurice Müller Laboratories (Department of Biomedical Research), University of Bern, 3008 Bern, Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital, 3010 Bern, Switzerland;
| |
Collapse
|
44
|
Lycke NY, Bemark M. The regulation of gut mucosal IgA B-cell responses: recent developments. Mucosal Immunol 2017; 10:1361-1374. [PMID: 28745325 DOI: 10.1038/mi.2017.62] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/20/2017] [Indexed: 02/04/2023]
Abstract
The majority of activated B cells differentiate into IgA plasma cells, with the gut being the largest producer of immunoglobulin in the body. Secretory IgA antibodies have numerous critical functions of which protection against infections and the role for establishing a healthy microbiota appear most important. Expanding our knowledge of the regulation of IgA B-cell responses and how effective mucosal vaccines can be designed are of critical importance. Here we discuss recent developments in the field that shed light on the uniqueness and complexity of mucosal IgA responses and the control of protective IgA responses in the gut, specifically.
Collapse
Affiliation(s)
- N Y Lycke
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - M Bemark
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
45
|
Gotoh Y, Suzuki S, Amako M, Kitamura S, Toda T. Effect of orally administered exopolysaccharides produced by Lactococcus lactis subsp. cremoris FC on a mouse model of dermatitis induced by repeated exposure to 2,4,6-trinitro-1-chlorobenzene. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.04.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
46
|
Wu W, Sun M, Chen F, Cao AT, Liu H, Zhao Y, Huang X, Xiao Y, Yao S, Zhao Q, Liu Z, Cong Y. Microbiota metabolite short-chain fatty acid acetate promotes intestinal IgA response to microbiota which is mediated by GPR43. Mucosal Immunol 2017; 10:946-956. [PMID: 27966553 PMCID: PMC5471141 DOI: 10.1038/mi.2016.114] [Citation(s) in RCA: 335] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 10/28/2016] [Indexed: 02/04/2023]
Abstract
Intestinal IgA, which is regulated by gut microbiota, has a crucial role in maintenance of intestinal homeostasis and in protecting the intestines from inflammation. However, the means by which microbiota promotes intestinal IgA responses remain unclear. Emerging evidence suggests that the host can sense gut bacterial metabolites in addition to pathogen-associated molecular patterns and that recognition of these small molecules influences host immune response in the intestines and beyond. We reported here that microbiota metabolite short-chain fatty acid acetate promoted intestinal IgA responses, which was mediated by "metabolite-sensing" GPR43. GPR43-/- mice demonstrated lower levels of intestinal IgA and IgA+ gut bacteria compared with those in wild type (WT) mice. Feeding WT but not GPR43-/- mice acetate but not butyrate promoted intestinal IgA response independent of T cells. Acetate promoted B-cell IgA class switching and IgA production in vitro in the presence of WT but not GPR43-/- dendritic cells (DCs). Mechanistically, acetate-induced DC expression of Aldh1a2, which converts Vitamin A into its metabolite retinoic acid (RA). Moreover, blockade of RA signaling inhibited the acetate induction of B-cell IgA production. Our studies thus identified a new pathway by which microbiota promotes intestinal IgA response through its metabolites.
Collapse
Affiliation(s)
- Wei Wu
- Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Mingming Sun
- Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Feidi Chen
- Department of Pathology, University of Texas Medical Branch, Galveston, TX
| | - Anthony T Cao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Han Liu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Ye Zhao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Xiangsheng Huang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Yi Xiao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | | | - Zhanju Liu
- Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China,Corresponding authors: Yingzi Cong, Ph.D., Department of Microbiology and Immunology, University of Texas Medical Branch, 4.142C Medical Research Building, 301 University Blvd, Galveston, TX 77555-1019. Phone: (409) 772-4902. Fax: (409) 772-5065. or Dr. Zhanju Liu, Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China; ; fax: (86) 21-6630-3983
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX,Department of Pathology, University of Texas Medical Branch, Galveston, TX,Corresponding authors: Yingzi Cong, Ph.D., Department of Microbiology and Immunology, University of Texas Medical Branch, 4.142C Medical Research Building, 301 University Blvd, Galveston, TX 77555-1019. Phone: (409) 772-4902. Fax: (409) 772-5065. or Dr. Zhanju Liu, Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China; ; fax: (86) 21-6630-3983
| |
Collapse
|
47
|
Karaffová V, Csank T, Mudroňová D, Király J, Revajová V, Gancarčíková S, Nemcová R, Pistl J, Vilček Š, Levkut M. Influence of Lactobacillus reuteri L26 Biocenol™ on immune response against porcine circovirus type 2 infection in germ-free mice. Benef Microbes 2017; 8:367-378. [PMID: 28504566 DOI: 10.3920/bm2016.0114] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Probiotic bacteria are frequently used for prevention of bacterial infections of the gastrointestinal tract, but there are only limited studies on their efficacy against viral gut infections in animals. The aim of this study was to investigate the effect of probiotic Lactobacillus reuteri L26 BiocenolTM on the innate and adaptive immune responses in germ-free Balb/c mice, experimentally infected by porcine circovirus type 2 (PCV2), which confers immunosuppressive effect. A total of 30 six-week-old female mice were divided into 3 groups and animals in experimental group LPCV (n=10) were inoculated with L. reuteri L26, animals in the control group (C; n=10) and experimental group PCV (n=10) received sterile De Man-Rogosa-Sharpe broth for 7 days. Subsequently, mice from both experimental groups were infected with PCV2; however, mice in the control group received virus cultivation medium (mock). Virus load in faeces, ileum and mesenteric lymph nodes (MLN); as well as gene expression of selected cytokines, immunoglobulin A (IgA) and polymeric Ig receptor (PIgR) in the ileum, and percentage of CD8+, CD19+ and CD49b+CD8- cells in the MLN were evaluated. Our results showed that L. reuteri significantly decreased the amount of PCV2 in faeces and in the ileum, and up-regulated the gene expression of chemokines, interferon (IFN)-γ, IgA and PIgR in the ileum. Increased IFN-γ mRNA level was accompanied by higher proportion of natural killer cells and up-regulated IgA and PIgR gene expressions were in accordance with significantly higher percentage of CD19+ lymphocytes in the MLN. These findings indicate that probiotic L. reuteri has an antiviral effect on PCV2 in the intestine which is mediated by stimulation of local gut immune response.
Collapse
Affiliation(s)
- V Karaffová
- 1 Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - T Csank
- 2 Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - D Mudroňová
- 2 Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - J Király
- 2 Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - V Revajová
- 1 Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - S Gancarčíková
- 2 Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - R Nemcová
- 2 Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - J Pistl
- 2 Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - Š Vilček
- 3 Department of Epizootiology and Parasitology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - M Levkut
- 1 Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| |
Collapse
|
48
|
Faria AMC, Reis BS, Mucida D. Tissue adaptation: Implications for gut immunity and tolerance. J Exp Med 2017; 214:1211-1226. [PMID: 28432200 PMCID: PMC5413340 DOI: 10.1084/jem.20162014] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/31/2017] [Accepted: 04/04/2017] [Indexed: 12/22/2022] Open
Abstract
Faria et al. discuss the concept that immune cells undergo specialized adaptation to tissue-specific conditions and its potential implications for tolerance and immunity. Tissue adaptation is an intrinsic component of immune cell development, influencing both resistance to pathogens and tolerance. Chronically stimulated surfaces of the body, in particular the gut mucosa, are the major sites where immune cells traffic and reside. Their adaptation to these environments requires constant discrimination between natural stimulation coming from harmless microbiota and food, and pathogens that need to be cleared. This review will focus on the adaptation of lymphocytes to the gut mucosa, a highly specialized environment that can help us understand the plasticity of leukocytes arriving at various tissue sites and how tissue-related factors operate to shape immune cell fate and function.
Collapse
Affiliation(s)
- Ana M C Faria
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065 .,Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270901, Brazil
| | - Bernardo S Reis
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065
| |
Collapse
|
49
|
Almejún MB, Campos BC, Patiño V, Galicchio M, Zelazko M, Oleastro M, Oppezzo P, Danielian S. Noninfectious complications in patients with pediatric-onset common variable immunodeficiency correlated with defects in somatic hypermutation but not in class-switch recombination. J Allergy Clin Immunol 2016; 139:913-922. [PMID: 27713077 DOI: 10.1016/j.jaci.2016.08.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 08/03/2016] [Accepted: 08/10/2016] [Indexed: 11/17/2022]
Abstract
BACKGROUND Common variable immunodeficiency (CVID) is a heterogeneous syndrome characterized by impaired immunoglobulin production and usually presents with a normal quantity of peripheral B cells. Most attempts aiming to classify these patients have mainly been focused on T- or B-cell phenotypes and their ability to produce protective antibodies, but it is still a major challenge to find a suitable classification that includes the clinical and immunologic heterogeneity of these patients. OBJECTIVE In this study we evaluated the late stages of B-cell differentiation in a heterogeneous population of patients with pediatric-onset CVID to clinically correlate and assess their ability to perform somatic hypermutation (SHM), class-switch recombination (CSR), or both. METHODS We performed a previously reported assay, the restriction enzyme hotspot mutation assay (IgκREHMA), to evaluate in vivo SHM status. We amplified switch regions from genomic DNA to investigate the quality of the double-strand break repairs in the class-switch recombination process in vivo. We also tested the ability to generate immunoglobulin germline and circle transcripts and to upregulate the activation-induced cytidine deaminase gene through in vitro T-dependent and T-independent stimuli. RESULTS Our results showed that patients could be classified into 2 groups according to their degree of SHM alteration. This stratification showed a significant association between patients of group A, severe alteration, and the presence of noninfectious complications. Additionally, 60% of patients presented with increased microhomology use at switched regions. In vitro activation revealed that patients with CVID behaved heterogeneously in terms of responsiveness to T-dependent stimuli. CONCLUSIONS The correlation between noninfectious complications and SHM could be an important tool for physicians to further characterize patients with CVID. This categorization would help to improve elucidation of the complex mechanisms involved in B-cell differentiation pathways.
Collapse
Affiliation(s)
- María Belén Almejún
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr Juan P. Garrahan, Buenos Aires, Argentina.
| | - Bárbara Carolina Campos
- Coordinación de Laboratorio, Hospital Nacional de Pediatría Prof. Dr Juan P. Garrahan, Buenos Aires, Argentina
| | - Virginia Patiño
- Unidad de Proteínas Recombinantes, Instituto Pasteur, Montevideo, Uruguay
| | - Miguel Galicchio
- Hospital de Niños Víctor J. Vilela, Rosario, Santa Fé, Argentina
| | - Marta Zelazko
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr Juan P. Garrahan, Buenos Aires, Argentina
| | - Matías Oleastro
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr Juan P. Garrahan, Buenos Aires, Argentina
| | - Pablo Oppezzo
- Unidad de Proteínas Recombinantes, Instituto Pasteur, Montevideo, Uruguay
| | - Silvia Danielian
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof. Dr Juan P. Garrahan, Buenos Aires, Argentina
| |
Collapse
|
50
|
Abstract
Secondary lymphoid tissues share the important function of bringing together antigens and rare antigen-specific lymphocytes to foster induction of adaptive immune responses. Peyer's patches (PPs) are unique compared to other secondary lymphoid tissues in their continual exposure to an enormous diversity of microbiome- and food-derived antigens and in the types of pathogens they encounter. Antigens are delivered to PPs by specialized microfold (M) epithelial cells and they may be captured and presented by resident dendritic cells (DCs). In accord with their state of chronic microbial antigen exposure, PPs exhibit continual germinal center (GC) activity. These GCs not only contribute to the generation of B cells and plasma cells producing somatically mutated gut antigen-specific IgA antibodies but have also been suggested to support non-specific antigen diversification of the B-cell repertoire. Here, we review current understanding of how PPs foster B-cell encounters with antigen, how they favor isotype switching to the secretory IgA isotype, and how their GC responses may uniquely contribute to mucosal immunity.
Collapse
Affiliation(s)
- Andrea Reboldi
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|