1
|
Ndeh DA, Nakjang S, Kwiatkowski KJ, Sawyers C, Koropatkin NM, Hirt RP, Bolam DN. A Bacteroides thetaiotaomicron genetic locus encodes activities consistent with mucin O-glycoprotein processing and N-acetylgalactosamine metabolism. Nat Commun 2025; 16:3485. [PMID: 40216766 PMCID: PMC11992087 DOI: 10.1038/s41467-025-58660-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
The gut microbiota is a key modulator of human health and the status of major diseases including cancer, diabetes and inflammatory bowel disease. Central to microbiota survival is the ability to metabolise complex dietary and host-derived glycans, including intestinal mucins. The prominent human gut microbe Bacteroides thetaiotaomicron (B. theta) is a versatile and highly efficient complex glycan degrader thanks to the expansion of gene clusters termed polysaccharide utilisation loci (PULs). While the mechanism of action for several singular dietary glycan-induced PULs have been elucidated, studies on the unusually high number of mucin-inducible PULs in B. theta significantly lag behind. Here we show that a mucin inducible PUL BT4240-50 encodes activities consistent with the processing and metabolism of mucin O-glycoproteins and their core sugar N-acetylgalactosamine (GalNAc). PUL BT4240-50 was also shown to be important for competitive growth on mucins in vitro, encoding a kinase (BT4240) critical for GalNAc metabolism. Additionally, BT4240-kinase was shown to be essential for glycosaminoglycan metabolism, extending the PULs function beyond mucins. These data advance our understanding of glycoprotein metabolism at mucosal surfaces, highlighting GalNAc as a key metabolite for competitive microbial survival in the human gut.
Collapse
Affiliation(s)
- Didier A Ndeh
- Division of Plant Sciences, School of Life Sciences, University of Dundee, Dundee, UK.
| | - Sirintra Nakjang
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, UK
| | - Kurt J Kwiatkowski
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Claire Sawyers
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Nicole M Koropatkin
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Robert P Hirt
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| | - David N Bolam
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
2
|
Yanagibashi T, Ikutani M, Nagai T, Arita M, Watanabe Y, Nagai Y, Takatsu K. IL-5-producing Group 2 innate lymphoid cells promote T cell-independent IgA production in cooperation with eosinophils. Int Immunol 2025; 37:273-285. [PMID: 39656643 DOI: 10.1093/intimm/dxae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/30/2024] [Indexed: 12/17/2024] Open
Abstract
Intestinal bacteria play a critical role in the regulation of the host immune system and an imbalance in the intestinal bacterial composition induces various host diseases. Therefore, maintaining a balance in the intestinal bacterial composition is crucial for health. Immunoglobulin A (IgA), produced through T cell-dependent and T cell-independent (TI) pathways, is essential for host defense against pathogen invasion and maintaining the balance of intestinal symbiotic bacteria. Interleukin (IL)-5 is constitutively produced by Group 2 innate lymphoid cells (ILC2s) and plays a critical role in the survival and proliferation of B cells and eosinophils. Here, we show the role of IL-5-producing ILC2s in intestinal TI IgA production at steady state using T cell receptor α deficient mice. In this mouse model, ILC2s increased fecal TI IgA levels in a non-inflammatory state in an IL-5-dependent manner. The administration of recombinant IL-33 (rIL-33) increased the amount of TI IgA production, accompanied by an increase in the number of IL-5-producing ILC2s in the large intestine. In addition, rIL-33 treatment increased IL-5-dependent IgA+ cells in isolated lymphoid follicles, the site of TI IgA production. Furthermore, eosinophils recruited by ILC2s were required for the maximal production of IgA in the TI pathway. Moreover, IL-5 increased the frequency of TI IgA-binding intestinal bacteria and was involved in the maintenance of intestinal bacterial composition. These findings indicate that IL-5-producing ILC2s together with eosinophils contribute to TI IgA production. In addition to their role in TI IgA production, IL-5-producing ILC2s may contribute to the homeostasis of intestinal commensal bacteria.
Collapse
Affiliation(s)
- Tsutomu Yanagibashi
- Toyama Prefectural Institute for Pharmaceutical Research, 17-1 Nakataikouyama, Imizu, Toyama 939-0363, Japan
| | - Masashi Ikutani
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | - Terumi Nagai
- WPI Nano Life Sciences Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-1 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Molecular and Cellular epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yasuharu Watanabe
- Toyama Prefectural Institute for Pharmaceutical Research, 17-1 Nakataikouyama, Imizu, Toyama 939-0363, Japan
| | - Yoshinori Nagai
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Kiyoshi Takatsu
- Toyama Prefectural Institute for Pharmaceutical Research, 17-1 Nakataikouyama, Imizu, Toyama 939-0363, Japan
| |
Collapse
|
3
|
Van Stigt AH, Nederend M, Smits G, Kuijer M, Schepp RM, Van Gageldonk PG, Hellinga AH, Van Binnendijk RS, Bont LJ, Van't Land B, Den Hartog G, Leusen JHW. Development and validation of a high throughput multiplex immunofluorescence assay to detect all human immunoglobulin isotypes and subclasses in human fluids. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:839-849. [PMID: 40073159 DOI: 10.1093/jimmun/vkae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 12/05/2024] [Indexed: 03/14/2025]
Abstract
Antibodies in human milk protect infants against infections, but currently no assay is described that is able to simultaneously measure all 9 antibody isotypes and subclasses immunoglobulins in human fluids, such as human milk. Our cohort "Protecting against Respiratory tract Infections through human Milk Analysis" (PRIMA) is focused on the relation between the occurrence of respiratory infections during the first year of life and concentration of maternal antibodies in breastfeeding. We developed and successfully validated a multiplex assay that is able to measure all nine antibody isotypes and subclasses in human plasma and milk (regardeless of the pathogen specificity), using a small sample volume. We used a multiplex immunofluorescence assay (MIA) requiring a minimal sample volume of 25 µl. Commercially available human isotype standards were used in spiking experiments to exclude the presence of cross reactivity. In addition, we prevented signal quenching by milk by determining the optimal dilution of human milk. In conclusion, we have developed a low-volume multiplex assay, that, for the first time, can reliably quantify functionally intact antibodies of all known human isotypes and subclasses and that is able to measure both kappa and lambda heavy chain antibodies. This assay can easily be implemented in other academic labs.
Collapse
Affiliation(s)
- Arthur H Van Stigt
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maaike Nederend
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gaby Smits
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Marjan Kuijer
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Rutger M Schepp
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Pieter G Van Gageldonk
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Anneke H Hellinga
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Robert S Van Binnendijk
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Louis J Bont
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital/University Medical Center Utrecht, Utrecht, The Netherlands
- ReSViNET Foundation, Zeist, The Netherlands
| | - Belinda Van't Land
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Danone Nutricia Research, Utrecht, The Netherlands
| | - Gerco Den Hartog
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Jeanette H W Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
4
|
Vruzhaj I, Gambirasi M, Busato D, Giacomin A, Toffoli G, Safa A. Gut Microbiota-Based Immunotherapy: Engineered Escherichia coli Nissle 1917 for Oral Delivery of Glypican-1 in Pancreatic Cancer. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:633. [PMID: 40282924 PMCID: PMC12028767 DOI: 10.3390/medicina61040633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: The administration of oral vaccines offers a potential strategy for cancer immunotherapy; yet, the development of effective platforms continues to pose a difficulty. This study examines Escherichia coli Nissle 1917 (EcN) as a microbial vector for the precise delivery of Glypican-1 (GPC1), a tumor-associated antigen significantly overexpressed in pancreatic ductal adenocarcinoma (PDAC).To evaluate the effectiveness of EcN as a vector for the delivery of GPC1 and assess its potential as an oral vaccination platform for cancer immunotherapy. Materials and Methods: EcN was genetically modified to produce a GPC1-flagellin fusion protein (GPC1-FL) to augment antigen immunogenicity. The expression and stability of GPC1 were confirmed in modified PANC02 cells using Western blot and flow cytometry, indicating that GPC1 expression did not influence tumor cell growth. A mouse model was employed to test immunogenicity post-oral delivery, measuring systemic IgG, IL-10, IL-2, and IFN-γ levels to indicate immune activation. Results: Oral immunization with EcN GPC1-FL elicited a robust systemic immune response, demonstrated by markedly increased levels of IgG and IL-10. IL-2 and IFN-γ concentrations were elevated in vaccinated mice relative to controls; however, the differences lacked statistical significance. Western blot examination of fecal samples verified consistent antigen expression in the gastrointestinal tract, indicating effective bacterial colonization and antigen retention. No detrimental impacts were noted, hence substantiating the safety of this methodology. Conclusions: These findings confirm EcN as a feasible and patient-friendly oral vaccination platform for cancer immunotherapy. The effective production of GPC1 in tumor cells, along with continuous antigen delivery and immune activation, underscores the promise of this approach for PDAC and other cancers. This study promotes microbial-based antigen delivery as a scalable, non-invasive substitute for traditional vaccine platforms.
Collapse
Affiliation(s)
- Idris Vruzhaj
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Marta Gambirasi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Davide Busato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Aurora Giacomin
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy (D.B.); (A.G.)
| | - Amin Safa
- Doctoral School in Pharmacological Sciences, University of Padua, 35122 Padova, Italy
- Institute of Research and Development, Duy Tan University, Da Nang 550000, Vietnam
| |
Collapse
|
5
|
Kliushnikova D, Otto F, Pilz G, Wipfler P, Harrer A. Intrathecal Immunoglobulin A Synthesis in Multiple Sclerosis: From Biological Aspects to Clinical Relevance. Biomolecules 2025; 15:108. [PMID: 39858502 PMCID: PMC11764441 DOI: 10.3390/biom15010108] [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: 12/09/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Intrathecal immunoglobulin A (IgA) synthesis in multiple sclerosis (MS) has long earned little attention, despite a potential significance in disease pathogenesis and prognosis. The presence of IgA-positive plasma cells in MS lesions and along damaged axons suggests a role in disease pathogenesis. Available clinical evidence about a potential positive or negative prognostic role is scarce and inconclusive. Recent observations, however, highlight the migration of immune regulatory IgA-producing plasma cells from the gut to the central nervous system (CNS) in experimental autoimmune encephalitis models. A connection between intrathecal IgA synthesis and the gut-brain axis in MS was further corroborated by the discovery of gut microbiota-specific IgA+ B cells in human CNS during relapse. In this review, we summarize current evidence on the occurrence and immunopathology of intrathecal IgA synthesis in MS, explore its biological implications, and address methodological challenges regarding the detection of IgA as a major limitation and possible source of inconsistencies in clinical studies. By synthesizing these diverse lines of evidence, we highlight the importance of further research and the need for standardized detection methods to clarify the role of IgA in MS pathogenesis, disease progression, and as potential biomarker.
Collapse
Affiliation(s)
- Dariia Kliushnikova
- Department of Neurology, Christian-Doppler University Hospital, Paracelsus Medical University, 5020 Salzburg, Austria; (F.O.); (G.P.); (P.W.)
| | - Ferdinand Otto
- Department of Neurology, Christian-Doppler University Hospital, Paracelsus Medical University, 5020 Salzburg, Austria; (F.O.); (G.P.); (P.W.)
- Department of Neurology, University Hospital Zurich, University of Zurich, 8006 Zürich, Switzerland
| | - Georg Pilz
- Department of Neurology, Christian-Doppler University Hospital, Paracelsus Medical University, 5020 Salzburg, Austria; (F.O.); (G.P.); (P.W.)
| | - Peter Wipfler
- Department of Neurology, Christian-Doppler University Hospital, Paracelsus Medical University, 5020 Salzburg, Austria; (F.O.); (G.P.); (P.W.)
| | - Andrea Harrer
- Department of Neurology, Christian-Doppler University Hospital, Paracelsus Medical University, 5020 Salzburg, Austria; (F.O.); (G.P.); (P.W.)
- Department of Dermatology and Allergology, Paracelsus Medical University, 5020 Salzburg, Austria
| |
Collapse
|
6
|
Colombo APV, Lourenço TGB, de Oliveira AM, da Costa ALA. Link Between Oral and Gut Microbiomes: The Oral-Gut Axis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1472:71-87. [PMID: 40111686 DOI: 10.1007/978-3-031-79146-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
In the last decades, groundbreaking research on the human microbiome has changed our reductionist conception of the etiology and pathogenesis of several chronic diseases. As a result, we have come to appreciate the significance of a balanced microbiome in maintaining human health. In this context, the upper and lower gastrointestinal tracts (GITs) comprise the most abundant and diverse microbiotas of the human body. In addition to its diversity, functional redundancy, and temporal stability, a healthy GIT microbiome is characterized by its body site specificity. In fact, current evidence has indicated that the translocation of oral species to the gut environment through the oral-gut axis is increased in an array of illnesses, including chronic inflammatory and metabolic diseases, neurological disorders, and cancer. Oral pathogens have also been shown to promote gut dysbiosis and systemic inflammation in animal models. Yet, some level of overlapping between oral and gut microbiomes may occur without disruption of these microbial communities and loss of site specificity. The uniqueness of each host-microbiome entity may hinder our ability to define a "universal" normal GIT microbiome. Despite that, this chapter summarizes the predominant health-related taxa along the human GIT, as well as their role in the physiology and immunity of the digestive system. Some mechanisms that may lead to disturbances and relevant shifts in the oral and gut microbiomes of major inflammatory chronic diseases are also pointed out. Lastly, oral-fecal microbial signatures are presented as potential biomarkers for several oral and systemic disorders. The recognition of such symbiotic/dysbiotic microbial profiles may provide insights into the development of more accurate early diagnosis and therapeutic ecological approaches to restore the balance of the GIT microbiome.
Collapse
Affiliation(s)
- Ana Paula Vieira Colombo
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, UFRJ, Rio de Janeiro, Brazil.
- School of Dentistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | | | - Adriana Miranda de Oliveira
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, UFRJ, Rio de Janeiro, Brazil
- School of Dentistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
7
|
Riaz S, Steinsland H, Andersen AZ, Boysen A, Hanevik K. Proportions of IgA antibodies targeting glycosylated epitopes of secreted Escherichia coli mucinase YghJ in initial plasmablast response differ from salivary and intestinally secreted IgA. Med Microbiol Immunol 2024; 214:2. [PMID: 39673573 DOI: 10.1007/s00430-024-00812-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/24/2024] [Indexed: 12/16/2024]
Abstract
Mucosal infections normally cause an immune response including activation of antigen-specific B cells in regional mucosa-associated lymphoid tissue. After recirculation of plasmablasts, and maturation at mucosal surfaces or bone marrow, plasma cells produce secretory or systemic IgA. It remains uncertain to what extent secretory and systemic IgA share the same target specificities. For vaccine candidate optimization, it is important to know whether IgA targeting of glycosylated epitopes of a protein antigen vary between mucosal and systemic sites. We evaluated glycosylated epitope specificity of systemic and mucosally secreted IgA against YghJ, a potential vaccine candidate antigen secreted by most pathogenic Escherichia coli. IgA from intestinal lavage, saliva, serum, and blood-derived antibody in lymphocyte supernatants (ALS) were collected from 21 volunteers following experimental infection with enterotoxigenic E. coli. Methods for preparing IgA from saliva and ALS were developed, and multiplex bead flow cytometric immunoassays were used to determine levels of IgA targeting natively glycosylated YghJ and estimating what proportion of these antibodies specifically targeted glycosylated epitopes. Following infection, anti-YghJ IgA levels increased substantially for most volunteers across all four specimen types. Target specificity of ALS IgA correlated well with serum IgA, but not with mucosally secreted IgA. Furthermore, glycosylation-specific proportion of salivary IgA was higher than, and did not correlate with, intestinally secreted IgA. These results indicate a new degree of complexity to our understanding of epitope-targeting and tissue specificity of mucosal antibody responses. Our findings also suggest that all features of an intestinal IgA response may not be well reflected in serum, saliva, or ALS, which are commonly used proxy specimens for evaluating intestinal immune responses.
Collapse
Affiliation(s)
- Saman Riaz
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Hans Steinsland
- Centre for Intervention Science in Maternal and Child Health (CISMAC), Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | | | - Kurt Hanevik
- Department of Clinical Science, University of Bergen, Bergen, Norway.
- National Centre for Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
8
|
Guo Y, Sheng X, Tang X, Xing J, Chi H, Zhan W. Immunoglobulin M-based local production in skin-associated lymphoid tissue of flounder (Paralichthys olivaceus) initiated by immersion with inactivated Edwardsiella tarda. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109982. [PMID: 39461396 DOI: 10.1016/j.fsi.2024.109982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
Fish skin, the mucosal site most exposed to external antigens, requires protection by an efficient local mucosal immune system. The mucosal reserve of IgM is recognized as an immune strategy that blocks pathogen invasion to maintain homeostasis, whereas the mechanism of skin-associated local IgM production induced by mucosal antigens is not well know. In this study, we found that the skin of flounder (Paralichthys olivaceus) was equipped with the immune cellular and molecular basis for processing mucosal antigens and triggering local specific responses, i.e., CD4+ Zap-70+ T cells, CD4- Zap-70+ T/NK cells, IgM+ MHCII+ B cells, PNA+ MHCII+ antigen-presenting cells, UEA-1+ WGA+ and UEA-1+ WGA- antigen-sampling cells, as well as secreted IgM and pIgR, as demonstrated by indirect immunofluorescence assay using different antibodies and lectins. After immersion immunization with inactivated Edwardsiella tarda, qPCR assay displayed up-regulation of immune-related genes in flounder skin. Flow cytometry analysis and EdU labeling demonstrated that the mucosal inactivated vaccine induced local proliferation and increased amounts of cutaneous IgM+ B cells. Skin explant culture proved the local production of specific IgM in the skin, which could bind to the surface of E. tarda. ELISA, laser scanning confocal microscopy, and Western blot revealed that, in addition to the elevated IgM levels, pIgR protein level was significantly up-regulated in skin tissue and surface mucus containing the pIgR (secretory component, SC)-tetrameric IgM complex, indicating that mucosal vaccine stimulated up-regulation of IgM and pIgR, which were secreted as a complex into skin mucus to exert the protective effects as secretory IgM. These findings deepen the understanding of IgM-based local responses in the mucosal immunity of teleosts, which will be critical for subsequent investigation into the protective mechanism of mucosal vaccines for fish health.
Collapse
Affiliation(s)
- Yuan Guo
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China.
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China
| |
Collapse
|
9
|
Liu J, Zhang K, Zhang X, Guan F, Zeng H, Kubo M, Lee P, Candotti F, James LK, Camara NOS, Benlagha K, Lei J, Forsman H, Yang L, Xiao W, Liu Z, Liu C. Immunoglobulin class-switch recombination: Mechanism, regulation, and related diseases. MedComm (Beijing) 2024; 5:e662. [PMID: 39144468 PMCID: PMC11322596 DOI: 10.1002/mco2.662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/30/2024] [Accepted: 06/30/2024] [Indexed: 08/16/2024] Open
Abstract
Maturation of the secondary antibody repertoire requires class-switch recombination (CSR), which switches IgM to other immunoglobulins (Igs), and somatic hypermutation, which promotes the production of high-affinity antibodies. Following immune response or infection within the body, activation of T cell-dependent and T cell-independent antigens triggers the activation of activation-induced cytidine deaminase, initiating the CSR process. CSR has the capacity to modify the functional properties of antibodies, thereby contributing to the adaptive immune response in the organism. Ig CSR defects, characterized by an abnormal relative frequency of Ig isotypes, represent a rare form of primary immunodeficiency. Elucidating the molecular basis of Ig diversification is essential for a better understanding of diseases related to Ig CSR defects and could provide clues for clinical diagnosis and therapeutic approaches. Here, we review the most recent insights on the diversification of five Ig isotypes and choose several classic diseases, including hyper-IgM syndrome, Waldenström macroglobulinemia, hyper-IgD syndrome, selective IgA deficiency, hyper-IgE syndrome, multiple myeloma, and Burkitt lymphoma, to illustrate the mechanism of Ig CSR deficiency. The investigation into the underlying mechanism of Ig CSR holds significant potential for the advancement of increasingly precise diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Jia‐Chen Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Ke Zhang
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Xu Zhang
- Department of RespiratoryThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
| | - Fei Guan
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Hu Zeng
- Department of ImmunologyMayo Clinic College of Medicine and ScienceRochesterUSA
| | - Masato Kubo
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama InstituteYokohamaJapan
| | - Pamela Lee
- Department of Paediatrics and Adolescent MedicineLKS Faculty of MedicineThe University of Hong KongHong KongChina
| | - Fabio Candotti
- Division of Immunology and AllergyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | | | | | - Kamel Benlagha
- Institut de Recherche Saint‐LouisUniversité de ParisParisFrance
| | - Jia‐Hui Lei
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Huamei Forsman
- Department of Rheumatology and Inflammation ResearchInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Lu Yang
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Wei Xiao
- Department of RespiratoryThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
| | - Zheng Liu
- Department of Otolaryngology‐Head and Neck SurgeryTongji Hospital, Tongji Medical College, HuazhongUniversity of Science and TechnologyWuhanChina
| | - Chao‐Hong Liu
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
10
|
Sarrigeorgiou I, Rouka E, Kotsiou OS, Perlepe G, Gerovasileiou ES, Gourgoulianis KI, Lymberi P, Zarogiannis SG. Natural antibodies targeting LPS in pleural effusions of various etiologies. Am J Physiol Lung Cell Mol Physiol 2024; 326:L727-L735. [PMID: 38591123 DOI: 10.1152/ajplung.00377.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/01/2024] [Accepted: 03/26/2024] [Indexed: 04/10/2024] Open
Abstract
Respiratory infection, cancer, and heart failure can cause abnormal accumulation of fluid in the pleural cavity. The immune responses within the cavity are orchestrated by leucocytes that reside in the serosal-associated lymphoid tissue. Natural antibodies (NAbs) are abundant in the serum (S) having a major role in systemic and mucosal immunity; however, their occurrence in pleural fluid (PF) remains an open question. Our aim herein was to detect and measure the levels of NAbs (IgM, IgG, IgA) targeting lipopolysaccharides (LPS) in both the pleural fluid and the serum of 78 patients with pleural effusions (PEs) of various etiologies. The values of anti-LPS NAb activity were extracted through a normalization step regarding the total IgM, IgG, and IgA levels, all determined by in-house ELISA. In addition, the ratios of PF/S values were analyzed further with other critical biochemical parameters from pleural fluids. Anti-LPS NAbs of all Ig classes were detected in most of the samples, while a significant increase of anti-LPS activity was observed in infectious and noninfectious compared with malignant PEs. Multivariate linear regression confirmed a negative correlation of IgM and IgA anti-LPS PF/S ratio with malignancy. Moreover, anti-LPS NAbs PF/S measurements led to increased positive and negative predictive power in ROC curves generated for the discrimination between benign and malignant PEs. Our results highlight the role of anti-LPS NAbs in the pleural cavity and demonstrate the potential translational impact that should be further explored.NEW & NOTEWORTHY Here we describe the detection and quantification of natural antibodies (NAbs) in the human pleural cavity. We show for the first time that IgM, IgG, and IgA anti-LPS natural antibodies are detected and measured in pleural effusions of infectious, noninfectious, and malignant etiologies and provide clinical correlates to demonstrate the translational impact of our findings.
Collapse
Affiliation(s)
- Ioannis Sarrigeorgiou
- Laboratory of Immunology, Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | - Erasmia Rouka
- Department of Nursing, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Ourania S Kotsiou
- Department of Nursing, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Garyfallia Perlepe
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Efrosini S Gerovasileiou
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Konstantinos I Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Peggy Lymberi
- Laboratory of Immunology, Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | - Sotirios G Zarogiannis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
11
|
May A, Gerhards H, Wollanke B. Effect of hospitalization on equine local intestinal immunoglobulin A (IgA) concentration measured in feces. J Equine Vet Sci 2024; 137:105078. [PMID: 38697372 DOI: 10.1016/j.jevs.2024.105078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/15/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
During hospitalization horses may develop gastrointestinal conditions triggered by a stress-associated weak local immune system. The prospective, clinical trial was conducted to find out whether fecal immunoglobulin A (IgA) concentrations could be determined in hospitalized horses and how they changed during hospitalization and in response to various stressors. Samples were obtained from 110 horses and a control group (n = 14). At arrival in the hospital, horses were categorized into pain grades (1-5), and elective versus strenuous surgery (> 2 hours, traumatic and emergency procedures). Feces were collected on day 1, day 2, day 3, and day 7 in all horses. Blood samples were obtained at the same intervals, but additionally after general anaesthesia in horses undergoing surgery (day 2). IgA concentration in feces was determined by ELISA and measured in optical density at 450nm. The control group showed constant IgA concentrations on all days (mean value 0.30 OD450 ±SD 0.11, 1.26 mg/g; n = 11). After general anaesthesia fecal IgA concentrations decreased considerably independent of duration and type of surgery (P < 0.001 for elective and P = 0.043 for traumatic surgeries). High plasma cortisol concentrations were weakly correlated with low fecal IgA on the day after surgery (P = 0.012, day 3, correlation coefficient r = 0.113). Equine fecal IgA concentrations showed a decline associated with transport, surgery, and hospitalization in general, indicating that stress has an impact on the local intestinal immune function and may predispose horses for developing gastrointestinal diseases such as enterocolitis.
Collapse
Affiliation(s)
- A May
- Equine Hospital, Ludwig-Maximilians-University Munich, Sonnenstrasse 14 85764 Oberschleissheim, Germany.
| | - H Gerhards
- retired, former head of Equine Hospital, Ludwig-Maximilians-University Munich, Germany
| | - B Wollanke
- Equine Hospital, Ludwig-Maximilians-University Munich, Sonnenstrasse 14 85764 Oberschleissheim, Germany
| |
Collapse
|
12
|
Ise W, Kurosaki T. Tissues of origin matter to plasma cell longevity. Nat Immunol 2024; 25:194-195. [PMID: 38191856 DOI: 10.1038/s41590-023-01731-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Affiliation(s)
- Wataru Ise
- Regulation of Host Defense Team, Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan.
| |
Collapse
|
13
|
Lin YL, Cheng PY, Chin CL, Chuang KT, Lin JY, Chang N, Pan CK, Lin CS, Pan SC, Chiang BL. A novel mucosal bivalent vaccine of EV-A71/EV-D68 adjuvanted with polysaccharides from Ganoderma lucidum protects mice against EV-A71 and EV-D68 lethal challenge. J Biomed Sci 2023; 30:96. [PMID: 38110940 PMCID: PMC10729491 DOI: 10.1186/s12929-023-00987-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Human enteroviruses A71 (EV-A71) and D68 (EV-D68) are the suspected causative agents of hand-foot-and-mouth disease, aseptic meningitis, encephalitis, acute flaccid myelitis, and acute flaccid paralysis in children. Until now, no cure nor mucosal vaccine existed for EV-A71 and EV-D68. Novel mucosal bivalent vaccines are highly important for preventing EV-A71 and EV-D68 infections. METHODS In this study, formalin-inactivated EV-A71 and EV-D68 were used as antigens, while PS-G, a polysaccharide from Ganoderma lucidum, was used as an adjuvant. Natural polysaccharides have the characteristics of intrinsic immunomodulation, biocompatibility, low toxicity, and safety. Mice were immunized intranasally with PBS, EV-A71, EV-D68, or EV-A71 + EV-D68, with or without PS-G as an adjuvant. RESULTS The EV-A71 + EV-D68 bivalent vaccine generated considerable EV-A71- and EV-D68-specific IgG and IgA titres in the sera, nasal washes, saliva, bronchoalveolar lavage fluid, and feces. These antibodies neutralized EV-D68 and EV-A71 infectivity. They also cross-neutralized infections by different EV-D68 and EV-A71 sub-genotypes. Furthermore, compared with the PBS group, EV-A71 + EV-D68 + PS-G-vaccinated mice exhibited an increased number of EV-D68- and EV-A71-specific IgA- and IgG-producing cells. In addition, T-cell proliferative responses, and IFN-γ and IL-17 secretion in the spleen were substantially induced when PS-G was used as an adjuvant with EV-A71 + EV-D68. Finally, in vivo challenge experiments demonstrated that the immune sera induced by EV-A71 + EV-D68 + PS-G conferred protection in neonate mice against lethal EV-A71 and EV-D68 challenges as indicated by the increased survival rate and decreased clinical score and viral RNA tissue expression. Taken together, all EV-A71/EV-D68 + PS-G-immunized mice developed potent specific humoral, mucosal, and cellular immune responses to EV-D68 and EV-A71 and were protected against them. CONCLUSIONS These findings demonstrated that PS-G can be used as a potential adjuvant for EV-A71 and EV-D68 bivalent mucosal vaccines. Our results provide useful information for the further preclinical and clinical development of a mucosal bivalent enterovirus vaccine against both EV-A71 and EV-D68 infections.
Collapse
Affiliation(s)
- Yu-Li Lin
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Pei-Yun Cheng
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chiao-Li Chin
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuan-Ting Chuang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Jing-Yi Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ning Chang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Kei Pan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Sheng Lin
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Siao-Cian Pan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Bor-Luen Chiang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan.
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
14
|
Planchais C, Molinos-Albert LM, Rosenbaum P, Hieu T, Kanyavuz A, Clermont D, Prazuck T, Lefrou L, Dimitrov JD, Hüe S, Hocqueloux L, Mouquet H. HIV-1 treatment timing shapes the human intestinal memory B-cell repertoire to commensal bacteria. Nat Commun 2023; 14:6326. [PMID: 37816704 PMCID: PMC10564866 DOI: 10.1038/s41467-023-42027-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
HIV-1 infection causes severe alterations of gut mucosa, microbiota and immune system, which can be curbed by early antiretroviral therapy. Here, we investigate how treatment timing affects intestinal memory B-cell and plasmablast repertoires of HIV-1-infected humans. We show that only class-switched memory B cells markedly differ between subjects treated during the acute and chronic phases of infection. Intestinal memory B-cell monoclonal antibodies show more prevalent polyreactive and commensal bacteria-reactive clones in late- compared to early-treated individuals. Mirroring this, serum IgA polyreactivity and commensal-reactivity are strongly increased in late-treated individuals and correlate with intestinal permeability and systemic inflammatory markers. Polyreactive blood IgA memory B cells, many of which egressed from the gut, are also substantially enriched in late-treated individuals. Our data establish gut and systemic B-cell polyreactivity to commensal bacteria as hallmarks of chronic HIV-1 infection and suggest that initiating treatment early may limit intestinal B-cell abnormalities compromising HIV-1 humoral response.
Collapse
Affiliation(s)
- Cyril Planchais
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cité, INSERM U1222, F-75015, Paris, France
| | - Luis M Molinos-Albert
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cité, INSERM U1222, F-75015, Paris, France
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036, Barcelona, Spain
| | - Pierre Rosenbaum
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cité, INSERM U1222, F-75015, Paris, France
| | - Thierry Hieu
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cité, INSERM U1222, F-75015, Paris, France
| | - Alexia Kanyavuz
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006, Paris, France
| | - Dominique Clermont
- Collection of the Institut Pasteur, Institut Pasteur, Université Paris Cité, 75015, Paris, France
| | - Thierry Prazuck
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, 45067, Orléans, France
| | - Laurent Lefrou
- Service d'Hépato-Gastro-Entérologie, CHR d'Orléans-La Source, 45067, Orléans, France
| | - Jordan D Dimitrov
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006, Paris, France
| | - Sophie Hüe
- INSERM U955-Équipe 16, Université Paris-Est Créteil, Faculté de Médecine, 94000, Créteil, France
| | - Laurent Hocqueloux
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, 45067, Orléans, France
| | - Hugo Mouquet
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cité, INSERM U1222, F-75015, Paris, France.
| |
Collapse
|
15
|
Das S, Stamnaes J, Kemppainen E, Hervonen K, Lundin KEA, Parmar N, Jahnsen FL, Jahnsen J, Lindfors K, Salmi T, Iversen R, Sollid LM. Separate Gut Plasma Cell Populations Produce Auto-Antibodies against Transglutaminase 2 and Transglutaminase 3 in Dermatitis Herpetiformis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300401. [PMID: 37424036 PMCID: PMC10477854 DOI: 10.1002/advs.202300401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/23/2023] [Indexed: 07/11/2023]
Abstract
Dermatitis herpetiformis (DH) is an inflammatory skin disorder often considered as an extra intestinal manifestation of celiac disease (CeD). Hallmarks of CeD and DH are auto-antibodies to transglutaminase 2 (TG2) and transglutaminase 3 (TG3), respectively. DH patients have auto-antibodies reactive with both transglutaminase enzymes. Here it is reported that in DH both gut plasma cells and serum auto-antibodies are specific for either TG2 or TG3 with no TG2-TG3 cross reactivity. By generating monoclonal antibodies from TG3-specific duodenal plasma cells of DH patients, three conformational epitope groups are defined. Both TG2-specific and TG3-specific gut plasma cells have few immunoglobulin (Ig) mutations, and the two transglutaminase-reactive populations show distinct selection of certain heavy and light chain V-genes. Mass spectrometry analysis of TG3-specific serum IgA corroborates preferential usage of IGHV2-5 in combination with IGKV4-1. Collectively, these results demonstrate parallel induction of anti-TG2 and anti-TG3 auto-antibody responses involving separate B-cell populations in DH patients.
Collapse
Affiliation(s)
- Saykat Das
- Department of ImmunologyOslo University Hospital‐RikshospitaletOslo0372Norway
- KG Jebsen Coeliac Disease Research CentreInstitute of Clinical MedicineUniversity of OsloOslo0372Norway
| | - Jorunn Stamnaes
- Department of ImmunologyOslo University Hospital‐RikshospitaletOslo0372Norway
- KG Jebsen Coeliac Disease Research CentreInstitute of Clinical MedicineUniversity of OsloOslo0372Norway
| | - Esko Kemppainen
- Celiac Disease Research CentreFaculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
| | - Kaisa Hervonen
- Celiac Disease Research CentreFaculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
- Department of DermatologyTampere University HospitalTampere33520Finland
| | - Knut E. A. Lundin
- KG Jebsen Coeliac Disease Research CentreInstitute of Clinical MedicineUniversity of OsloOslo0372Norway
- Department of GastroenterologyOslo University Hospital‐RikshospitaletOslo0372Norway
| | - Naveen Parmar
- Department of PathologyUniversity of Oslo and Institute of Clinical MedicineOslo University Hospital‐RikshospitaletOslo0372Norway
| | - Frode L. Jahnsen
- Department of PathologyUniversity of Oslo and Institute of Clinical MedicineOslo University Hospital‐RikshospitaletOslo0372Norway
| | - Jørgen Jahnsen
- Department of GastroenterologyAkershus University HospitalLørenskog1478Norway
| | - Katri Lindfors
- Celiac Disease Research CentreFaculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
| | - Teea Salmi
- Celiac Disease Research CentreFaculty of Medicine and Health TechnologyTampere UniversityTampere33520Finland
| | - Rasmus Iversen
- Department of ImmunologyOslo University Hospital‐RikshospitaletOslo0372Norway
- KG Jebsen Coeliac Disease Research CentreInstitute of Clinical MedicineUniversity of OsloOslo0372Norway
| | - Ludvig M. Sollid
- Department of ImmunologyOslo University Hospital‐RikshospitaletOslo0372Norway
- KG Jebsen Coeliac Disease Research CentreInstitute of Clinical MedicineUniversity of OsloOslo0372Norway
| |
Collapse
|
16
|
Azarias Da Silva M, Nioche P, Soudaramourty C, Bull-Maurer A, Tiouajni M, Kong D, Zghidi-Abouzid O, Picard M, Mendes-Frias A, Santa-Cruz A, Carvalho A, Capela C, Pedrosa J, Castro AG, Loubet P, Sotto A, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Tran TA, Tokunaga K, Silvestre R, Corbeau P, Mammano F, Estaquier J. Repetitive mRNA vaccination is required to improve the quality of broad-spectrum anti-SARS-CoV-2 antibodies in the absence of CXCL13. SCIENCE ADVANCES 2023; 9:eadg2122. [PMID: 37540749 PMCID: PMC10403221 DOI: 10.1126/sciadv.adg2122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/05/2023] [Indexed: 08/06/2023]
Abstract
Since the initial spread of severe acute respiratory syndrome coronavirus 2 infection, several viral variants have emerged and represent a major challenge for immune control, particularly in the context of vaccination. We evaluated the quantity, quality, and persistence of immunoglobulin G (IgG) and IgA in individuals who received two or three doses of messenger RNA (mRNA) vaccines, compared with previously infected vaccinated individuals. We show that three doses of mRNA vaccine were required to match the humoral responses of preinfected vaccinees. Given the importance of antibody-dependent cell-mediated immunity against viral infections, we also measured the capacity of IgG to recognize spike variants expressed on the cell surface and found that cross-reactivity was also strongly improved by repeated vaccination. Last, we report low levels of CXCL13, a surrogate marker of germinal center activation and formation, in vaccinees both after two and three doses compared with preinfected individuals, providing a potential explanation for the short duration and low quality of Ig induced.
Collapse
Affiliation(s)
| | - Pierre Nioche
- INSERM-U1124, Université Paris Cité, Paris, France
- Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | | | | | - Mounira Tiouajni
- INSERM-U1124, Université Paris Cité, Paris, France
- Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | - Dechuan Kong
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | - Ana Mendes-Frias
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - André Santa-Cruz
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal
| | - Alexandre Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal
| | - Carlos Capela
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal
| | - Jorge Pedrosa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António Gil Castro
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Paul Loubet
- Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Albert Sotto
- Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Laurent Muller
- Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | | | - Claire Roger
- Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | | | - Sandra Duvnjak
- Service de Gérontologie et Prévention du Vieillissement, CHU de Nîmes, Nîmes, France
| | - Tu-Anh Tran
- Service de Pédiatrie, CHU de Nîmes, Nîmes, France
| | - Kenzo Tokunaga
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Pierre Corbeau
- Institut de Génétique Humaine, UMR9002 CNRS-Université de Montpellier, Montpellier, France
- Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France
| | - Fabrizio Mammano
- INSERM-U1124, Université Paris Cité, Paris, France
- Université de Tours, INSERM, UMR1259 MAVIVH, Tours, France
| | - Jérôme Estaquier
- INSERM-U1124, Université Paris Cité, Paris, France
- CHU de Québec-Université Laval Research Center, Québec City, Québec, Canada
| |
Collapse
|
17
|
Tamura H. IgA nephropathy associated with Crohn's disease. World J Methodol 2023; 13:67-78. [PMID: 37456980 PMCID: PMC10348078 DOI: 10.5662/wjm.v13.i3.67] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/16/2023] [Accepted: 05/12/2023] [Indexed: 06/20/2023] Open
Abstract
The relationship between IgA nephropathy (IgAN) and Crohn’s disease was reported. IgAN is the most common primary glomerulonephritis and one of the leading causes of chronic kidney disease and end-stage renal failure, and up to 50% of cases progressed to end-stage renal disease within 25 years after IgAN diagnosis. However, specific and effective therapeutic strategies are still lacking. In this review, we discuss the possibility of the mechanism involved in IgAN associated with Crohn’s disease based on the findings of basic and clinical studies. Although the etiology of IgAN associated with Crohn’s disease is not permanent and various factors are thought to be involved, the stabilization of the disease condition of Crohn’s disease is believed to help treat IgAN.
Collapse
Affiliation(s)
- Hiroshi Tamura
- Department of Pediatrics, Kumamoto University, Kumamoto 8608556, Japan
| |
Collapse
|
18
|
Rathore APS, St John AL. Promises and challenges of mucosal COVID-19 vaccines. Vaccine 2023; 41:4042-4049. [PMID: 37045682 PMCID: PMC10083204 DOI: 10.1016/j.vaccine.2023.04.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
Coronavirus disease-2019 (COVID-19) is an ongoing pandemic caused by the newly emerged virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Currently, COVID-19 vaccines are given intramuscularly and they have been shown to evoke systemic immune responses that are highly efficacious towards preventing severe disease and death. However, vaccine-induced immunity wanes within a short time, and booster doses are currently recommended. Furthermore, current vaccine formulations do not adequately restrict virus infection at the mucosal sites, such as in the nasopharyngeal tract and, therefore, have limited capacity to block virus transmission. With these challenges in mind, several mucosal vaccines are currently being developed with the aim of inducing long-lasting protective immune responses at the mucosal sites where SARS-COV-2 infection begins. Past successes in mucosal vaccinations underscore the potential of these developmental stage SARS-CoV-2 vaccines to reduce disease burden, if not eliminate it altogether. Here, we discuss immune responses that are triggered at the mucosal sites and recent advances in our understanding of mucosal responses induced by SARS-CoV-2 infection and current COVID-19 vaccines. We also highlight several mucosal SARS-COV-2 vaccine formulations that are currently being developed or tested for human use and discuss potential challenges to mucosal vaccination.
Collapse
Affiliation(s)
- Abhay P S Rathore
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27705, USA
| | - Ashley L St John
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27705, USA; Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857 Singapore, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; SingHealth Duke-NUS Global Health Institute, Singapore.
| |
Collapse
|
19
|
Sun M, Ju J, Xu H, Wang Y. Intestinal fungi and antifungal secretory immunoglobulin A in Crohn's disease. Front Immunol 2023; 14:1177504. [PMID: 37359518 PMCID: PMC10285161 DOI: 10.3389/fimmu.2023.1177504] [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: 03/01/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
The human gastrointestinal tract harbors trillions of commensal microorganisms. Emerging evidence points to a possible link between intestinal fungal dysbiosis and antifungal mucosal immunity in inflammatory bowel disease, especially in Crohn's disease (CD). As a protective factor for the gut mucosa, secretory immunoglobulin A (SIgA) prevents bacteria from invading the intestinal epithelium and maintains a healthy microbiota community. In recent years, the roles of antifungal SIgA antibodies in mucosal immunity, including the regulation of intestinal immunity binding to hyphae-associated virulence factors, are becoming increasingly recognized. Here we review the current knowledge on intestinal fungal dysbiosis and antifungal mucosal immunity in healthy individuals and in patients with CD, discuss the factors governing antifungal SIgA responses in the intestinal mucosa in the latter group, and highlight potential antifungal vaccines targeting SIgA to prevent CD.
Collapse
|
20
|
El-Demerdash AS, Mohamady SN, Megahed HM, Ali NM. Evaluation of gene expression related to immunity, apoptosis, and gut integrity that underlies Artemisia's therapeutic effects in necrotic enteritis-challenged broilers. 3 Biotech 2023; 13:181. [PMID: 37193331 PMCID: PMC10182211 DOI: 10.1007/s13205-023-03560-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/15/2023] [Indexed: 05/18/2023] Open
Abstract
The experiment was designed to validate the effect of Artemisia annua and its novel commercial product (Navy Cox) on the control of necrotic enteritis (NE). A total of one hundred forty broiler chicks were randomly distributed into seven equal groups: G1, control negative; G2, infected with Eimeria (day 15) and C. perfringens (day 19); G3, treated with Navy Cox before challenge; G4, treated with Artemisia before challenge; G5, infected and then treated with Navy Cox; G6, infected and then treated with Artemisia; and G7, infected and treated with amoxicillin. Chicken response and immune organ indicants were recorded during the observation period (4 weeks). Whole blood and serum samples were collected for immunological evaluation, and tissue samples were collected for bacterial counts and estimation of mRNA expression of genes encoding apoptosis, tight junctions, and immunity. Chickens in the infected group revealed a significant decrease in RBCS, HB, PCV% total protein, Lysozyme, and nitric oxide activity in addition to leukocytosis, heterophilia, monocytosis, increase in cortisol, interleukins, and malondialdehyde. Treated groups displayed lower lesions, colony-forming units, and no mortality. Concurrently, a complete blood profile, antioxidants, and immune markers showed significant improvements. The mRNA expression levels of CASP, CLDN-1, OCLN, TJPI, MUC2, and cell-mediated immune response genes (p < 0.0001) were significantly alleviated in the treated groups compared with the challenged counterpart. This is the first-ever report on the efficacy valuation of Navy Cox compared to standard antibiotic treatment of clostridial NE. Navy Cox proved remarkable capability to minimize C. perfringens colonization in broiler intestines, modulation of mucus production, gut health integrity, immune organs, and immune response when used as a prophylactic agent in this form or naturally as Artemisia.
Collapse
Affiliation(s)
- Azza S. El-Demerdash
- Microbiology Department, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Zagazig, Egypt
| | - Sahar N. Mohamady
- Clinical Pathology Department, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Zagazig, Egypt
| | - Hend M. Megahed
- Biochemistry Department, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Zagazig, Egypt
| | - Naglaa M. Ali
- Poultry Disease Department, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Assuit, Egypt
| |
Collapse
|
21
|
Jemelkova J, Stuchlova Horynova M, Kosztyu P, Zachova K, Zadrazil J, Galuszkova D, Takahashi K, Novak J, Raska M. GalNAc-T14 may Contribute to Production of Galactose-Deficient Immunoglobulin A1, the Main Autoantigen in IgA Nephropathy. Kidney Int Rep 2023; 8:1068-1075. [PMID: 37180502 PMCID: PMC10166743 DOI: 10.1016/j.ekir.2023.02.1072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/13/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction Immunoglobulin A1 (IgA1) with galactose-deficient O-glycans (Gd-IgA1) play a key role in the pathogenesis of IgA nephropathy (IgAN). Mucosal-tissue infections increase IL-6 production and, in patients with IgAN, are often associated with macroscopic hematuria. IgA1-secreting cell lines derived from the circulation of patients with IgAN, compared to those of healthy controls (HCs), produce more IgA1 that has O-glycans with terminal or sialylated N-acetylgalactosamine (GalNAc). GalNAc residues are added to IgA1 hinge region by some of the 20 GalNAc transferases, the O-glycosylation-initiating enzymes. Expression of GALNT2, encoding GalNAc-T2, the main enzyme initiating IgA1 O-glycosylation, is similar in cells derived from patients with IgAN and HCs. In this report, we extend our observations of GALNT14 overexpression in IgA1-producing cell lines from patients with IgAN. Methods GALNT14 expression was analyzed in peripheral blood mononuclear cells (PBMCs) from patients with IgAN and from HCs. Moreover, the effect of GALNT14 overexpression or knock-down on Gd-IgA1 production in Dakiki cells was assessed. Results GALNT14 was overexpressed in PBMCs from patients with IgAN. IL-6 increased GALNT14 expression in PBMCs from patients with IgAN and HCs. We used IgA1-producing cell line Dakiki, a previously reported model of Gd-IgA1-producing cells, and showed that overexpression of GalNAc-T14 enhanced galactose deficiency of IgA1, whereas siRNA-mediated GalNAc-T14 knock-down reduced it. GalNAc-T14 was localized in trans-Golgi network, as expected. Conclusions Overexpression of GALNT14 due to inflammatory signals during mucosal infections may contribute to overproduction of Gd-IgA1 in patients with IgAN.
Collapse
Affiliation(s)
- Jana Jemelkova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Milada Stuchlova Horynova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Petr Kosztyu
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Katerina Zachova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Josef Zadrazil
- Department of Internal Medicine III Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Dana Galuszkova
- Department of Transfusion Medicine, University Hospital Olomouc, Olomouc, Czech Republic
| | - Kazuo Takahashi
- Department of Biomedical Molecular Sciences, School of Medicine, Fujita Health University, Nagoya, Aichi, Japan
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Milan Raska
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
- Department of Immunology, University Hospital Olomouc, Olomouc, Czech Republic
| |
Collapse
|
22
|
Liu RX, Wen C, Ye W, Li Y, Chen J, Zhang Q, Li W, Liang W, Wei L, Zhang J, Chan KW, Wang X, Yang X, Liu H. Altered B cell immunoglobulin signature exhibits potential diagnostic values in human colorectal cancer. iScience 2023; 26:106140. [PMID: 36879799 PMCID: PMC9984553 DOI: 10.1016/j.isci.2023.106140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 12/27/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Antibody-secreting B cells have long been considered the central element of gut homeostasis; however, tumor-associated B cells in human colorectal cancer (CRC) have not been well characterized. Here, we show that the clonotype, phenotype, and immunoglobulin subclasses of tumor-infiltrating B cells have changed compared to adjacent normal tissue B cells. Remarkably, the tumor-associated B cell immunoglobulin signature alteration can also be detected in the plasma of patients with CRC, suggesting that a distinct B cell response was also evoked in CRC. We compared the altered plasma immunoglobulin signature with the existing method of CRC diagnosis. Our diagnostic model exhibits improved sensitivity compared to the traditional biomarkers, CEA and CA19-9. These findings disclose the altered B cell immunoglobulin signature in human CRC and highlight the potential of using the plasma immunoglobulin signature as a non-invasive method for the assessment of CRC.
Collapse
Affiliation(s)
- Rui-Xian Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Chuangyu Wen
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523059, China
| | - Weibiao Ye
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523059, China
| | - Yewei Li
- Department of Statistical Science, School of Mathematics, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Junxiong Chen
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Qian Zhang
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Weiqian Li
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Wanfei Liang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Lili Wei
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Jingdan Zhang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Ka-Wo Chan
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xueqin Wang
- International Institute of Finance, School of Management, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiangling Yang
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Huanliang Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| |
Collapse
|
23
|
Mes L, Steffen U, Chen HJ, Veth J, Hoepel W, Griffith GR, Schett G, den Dunnen J. IgA2 immune complexes selectively promote inflammation by human CD103+ dendritic cells. Front Immunol 2023; 14:1116435. [PMID: 37006318 PMCID: PMC10061090 DOI: 10.3389/fimmu.2023.1116435] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
While immunoglobulin A (IgA) is well known for its neutralizing and anti-inflammatory function, it is becoming increasingly clear that IgA can also induce human inflammatory responses by various different immune cells. Yet, little is known about the relative role of induction of inflammation by the two IgA subclasses i.e. IgA1, most prominent subclass in circulation, and IgA2, most prominent subclass in the lower intestine. Here, we set out to study the inflammatory function of IgA subclasses on different human myeloid immune cell subsets, including monocytes, and in vitro differentiated macrophages and intestinal CD103+ dendritic cells (DCs). While individual stimulation with IgA immune complexes only induced limited inflammatory responses by human immune cells, both IgA subclasses strongly amplified pro-inflammatory cytokine production upon co-stimulation with Toll-like receptor (TLR) ligands such as Pam3CSK4, PGN, and LPS. Strikingly, while IgA1 induced slightly higher or similar levels of pro-inflammatory cytokines by monocytes and macrophages, respectively, IgA2 induced substantially more inflammation than IgA1 by CD103+ DCs. In addition to pro-inflammatory cytokine proteins, IgA2 also induced higher mRNA expression levels, indicating that amplification of pro-inflammatory cytokine production is at least partially regulated at the level of gene transcription. Interestingly, cytokine amplification by IgA1 was almost completely dependent on Fc alpha receptor I (FcαRI), whilst blocking this receptor only partially reduced cytokine induction by IgA2. In addition, IgA2-induced amplification of pro-inflammatory cytokines was less dependent on signaling through the kinases Syk, PI3K, and TBK1/IKKϵ. Combined, these findings indicate that IgA2 immune complexes, which are most abundantly expressed in the lower intestine, particularly promote inflammation by human CD103+ intestinal DCs. This may serve an important physiological function upon infection, by enabling inflammatory responses by this otherwise tolerogenic DC subset. Since various inflammatory disorders are characterized by disturbances in IgA subclass balance, this may also play a role in the induction or exacerbation of chronic intestinal inflammation.
Collapse
Affiliation(s)
- Lynn Mes
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
- Department of Medical Microbiology, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Ulrike Steffen
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Hung-Jen Chen
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Jennifer Veth
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Willianne Hoepel
- Department of Experimental Immunology, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Centers (UMC), Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Guillermo Romeo Griffith
- Department of Medical Biochemistry, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jeroen den Dunnen
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
- *Correspondence: Jeroen den Dunnen,
| |
Collapse
|
24
|
Canelli E, Ferrari L, Borghetti P, Candela F, Abiakam NS, Bianchera A, Buttini F, Magi GE, Sonvico F, Martelli P, Bettini R. Nano-adjuvanted dry powder vaccine for the mucosal immunization against airways pathogens. Front Vet Sci 2023; 10:1116722. [PMID: 36998637 PMCID: PMC10043307 DOI: 10.3389/fvets.2023.1116722] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/15/2023] [Indexed: 03/15/2023] Open
Abstract
Nasal vaccination has been shown to provide optimal protection against respiratory pathogens. However, mucosal vaccination requires the implementation of specific immunization strategies to improve its effectiveness. Nanotechnology appears a key approach to improve the effectiveness of mucosal vaccines, since several nanomaterials provide mucoadhesion, enhance mucosal permeability, control antigen release and possess adjuvant properties. Mycoplasma hyopneumoniae is the main causative agent of enzootic pneumonia in pigs, a respiratory disease responsible for considerable economic losses in the pig farming worldwide. The present work developed, characterized, and tested in vivo an innovative dry powder nasal vaccine, obtained from the deposition on a solid carrier of an inactivated antigen and a chitosan-coated nanoemulsion, as an adjuvant. The nanoemulsion was obtained through a low-energy emulsification technique, a method that allowed to achieve nano droplets in the order of 200 nm. The oil phase selected was alpha-tocopherol, sunflower oil, and poly(ethylene glycol) hydroxystearate used as non-ionic tensioactive. The aqueous phase contained chitosan, which provides a positive charge to the emulsion, conferring mucoadhesive properties and favoring interactions with inactivated M. hyopneumoniae. Finally, the nanoemulsion was layered with a mild and scalable process onto a suitable solid carrier (i.e., lactose, mannitol, or calcium carbonate) to be transformed into a solid dosage form for administration as dry powder. In the experimental study, the nasal vaccine formulation with calcium carbonate was administered to piglets and compared to intramuscular administration of a commercial vaccine and of the dry powder without antigen, aimed at evaluating the ability of IN vaccination to elicit an in vivo local immune response and a systemic immune response. Intranasal vaccination was characterized by a significantly higher immune response in the nasal mucosa at 7 days post-vaccination, elicited comparable levels of Mycoplasma-specific IFN-γ secreting cells and comparable, if not higher, responsiveness of B cells expressing IgA and IgG in peripheral blood mononuclear cells, with those detected upon a conventional intramuscular immunization. In conclusion, this study illustrates a simple and effective strategy for the development of a dry powder vaccine formulation for nasal administration which could be used as alternative to current parenteral commercial vaccines.
Collapse
Affiliation(s)
- Elena Canelli
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Luca Ferrari
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Paolo Borghetti
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Francesco Candela
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug, University of Parma, Parma, Italy
| | - Nkemjika Sopuru Abiakam
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug, University of Parma, Parma, Italy
| | - Annalisa Bianchera
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug, University of Parma, Parma, Italy
- Interdepartmental Centre Biopharmanet-Tec, University of Parma, Parma, Italy
| | - Francesca Buttini
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug, University of Parma, Parma, Italy
- Interdepartmental Centre Biopharmanet-Tec, University of Parma, Parma, Italy
| | - Gian Enrico Magi
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Fabio Sonvico
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug, University of Parma, Parma, Italy
- Interdepartmental Centre Biopharmanet-Tec, University of Parma, Parma, Italy
- *Correspondence: Fabio Sonvico
| | - Paolo Martelli
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Ruggero Bettini
- Advanced Drug Delivery Research Laboratory, Department of Food and Drug, University of Parma, Parma, Italy
- Interdepartmental Centre Biopharmanet-Tec, University of Parma, Parma, Italy
| |
Collapse
|
25
|
Wang X, Hu Y, Zhu X, Cai L, Farooq MZ, Yan X. Bacteroides-derived isovaleric acid enhances mucosal immunity by facilitating intestinal IgA response in broilers. J Anim Sci Biotechnol 2023; 14:4. [PMID: 36604758 PMCID: PMC9817248 DOI: 10.1186/s40104-022-00807-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 11/22/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The interaction between nutrition and immunity plays a vital role in nutrient digestion, absorption, and metabolism during poultry production. Recent studies showed that the gut microbiota contributes to the development of intestinal mucosal immunity. However, the mechanisms by which gut microbes regulate this process remain unclear. METHODS We compared the intestinal mucosal immunity and gut microbiota of Arbor Acre broilers (AA (lower mucosal immunity) and Chinese native Wuliang Mountain Black-bone chickens (WLMB) (higher mucosal immunity) using 16S rDNA sequencing, transcriptomic analysis, and immunoglobulin A (IgA) antibody repertoire sequencing. We then combined 16S rDNA sequencing with transcriptomics to identify the key microbes and found that they were positively correlated with IgA production. Next, we transplanted candidate microbes into 1-day-old broiler to explore their role in intestinal mucosal immunity. Finally, we verified the function of candidate microbial metabolites in regulating the immune function of macrophages and the intestinal-epithelial cells (IECs) using in vitro experiments. RESULTS WLMB performs stronger mucosal immunity than AA, including higher IgA levels, more diverse IgA antibody repertoire, and higher bacterial affinity. Bacteroides was identified as the key microbes related to the intestinal IgA response. Bacteroides transplantation could increase IgA concentration in the duodenal contents by enhancing the expression of IgA, polymeric immunoglobin receptor (PIgR), B cell-activating factor of the TNF family (BAFF), and activation-induced cytidine deaminase (AID) in the duodenum. Additionally, Bacteroides-derived isovaleric acid promoted M2 macrophage polarization of macrophage via mTOR/PPAR-γ/STAT3 signaling pathways and regulated the immunologic function of IECs to produce cytokines, including interleukin (IL)-10, IL-4, BAFF, and transforming growth factor-beta (TGF-β), thus promoting IgA production in B cells by facilitating AID expression. CONCLUSION Our study revealed that Bacteroides modulate the intestinal IgA response and maintain gut health in broilers. Bacteroides may be a promising alternative as an immunomodulatory microbial agent for developing next-generation probiotics for broiler production.
Collapse
Affiliation(s)
- Xinkai Wang
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| | - Yifan Hu
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| | - Xiaoyan Zhu
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| | - Liyuan Cai
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| | - Muhammad Zahid Farooq
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei China ,grid.412967.f0000 0004 0609 0799Department of Animal Sciences, University of Veterinary and Animal Sciences (Jhang Campus), Lahore, 54000 Pakistan
| | - Xianghua Yan
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| |
Collapse
|
26
|
Bertrand Y, Sánchez-Montalvo A, Hox V, Froidure A, Pilette C. IgA-producing B cells in lung homeostasis and disease. Front Immunol 2023; 14:1117749. [PMID: 36936934 PMCID: PMC10014553 DOI: 10.3389/fimmu.2023.1117749] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
Immunoglobulin A (IgA) is the most abundant Ig in mucosae where it plays key roles in host defense against pathogens and in mucosal immunoregulation. Whereas intense research has established the different roles of secretory IgA in the gut, its function has been much less studied in the lung. This review will first summarize the state-of-the-art knowledge on the distribution and phenotype of IgA+ B cells in the human lung in both homeostasis and disease. Second, it will analyze the studies looking at cellular and molecular mechanisms of homing and priming of IgA+ B cells in the lung, notably following immunization. Lastly, published data on observations related to IgA and IgA+ B cells in lung and airway disease such as asthma, cystic fibrosis, idiopathic pulmonary fibrosis, or chronic rhinosinusitis, will be discussed. Collectively it provides the state-of-the-art of our current understanding of the biology of IgA-producing cells in the airways and identifies gaps that future research should address in order to improve mucosal protection against lung infections and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Youri Bertrand
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
| | - Alba Sánchez-Montalvo
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Allergy and Clinical Immunology Research Group, Department of Microbiology, Immunology and Transplantation, Katholieke universiteit (KU) Leuven, Leuven, Belgium
| | - Valérie Hox
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Department of Otorhinolaryngology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Antoine Froidure
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Service de Pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Charles Pilette
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Service de Pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- *Correspondence: Charles Pilette,
| |
Collapse
|
27
|
Romero-Ramírez S, Sosa-Hernández VA, Cervantes-Díaz R, Carrillo-Vázquez DA, Meza-Sánchez DE, Núñez-Álvarez C, Torres-Ruiz J, Gómez-Martín D, Maravillas-Montero JL. Salivary IgA subtypes as novel disease biomarkers in systemic lupus erythematosus. Front Immunol 2023; 14:1080154. [PMID: 36911711 PMCID: PMC9992540 DOI: 10.3389/fimmu.2023.1080154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Introduction Immunoglobulin A (IgA) is the main antibody isotype in body fluids such as tears, intestinal mucous, colostrum, and saliva. There are two subtypes of IgA in humans: IgA1, mainly present in blood and mucosal sites, and IgA2, preferentially expressed in mucosal sites like the colon. In clinical practice, immunoglobulins are typically measured in venous or capillary blood; however, alternative samples, including saliva, are now being considered, given their non-invasive and easy collection nature. Several autoimmune diseases have been related to diverse abnormalities in oral mucosal immunity, such as rheumatoid arthritis, Sjogren's syndrome, and systemic lupus erythematosus (SLE). Methods We decided to evaluate the levels of both IgA subtypes in the saliva of SLE patients. A light chain capture-based ELISA measured specific IgA1 and IgA2 levels in a cohort of SLE patients compared with age and gender-matched healthy volunteers. Results Surprisingly, our results indicated that in the saliva of SLE patients, total IgA and IgA1 subtype were significantly elevated; we also found that salivary IgA levels, particularly IgA2, positively correlate with anti-dsDNA IgG antibody titers. Strikingly, we also detected the presence of salivary anti-nucleosome IgA antibodies in SLE patients, a feature not previously reported elsewhere. Conclusions According to our results and upon necessary validation, IgA characterization in saliva could represent a potentially helpful tool in the clinical care of SLE patients with the advantage of being a more straightforward, faster, and safer method than manipulating blood samples.
Collapse
Affiliation(s)
- Sandra Romero-Ramírez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Víctor A Sosa-Hernández
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Rodrigo Cervantes-Díaz
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Daniel A Carrillo-Vázquez
- Departamento de Medicina Interna, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - David E Meza-Sánchez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Carlos Núñez-Álvarez
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jiram Torres-Ruiz
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diana Gómez-Martín
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José L Maravillas-Montero
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
28
|
Garcia B, Dong F, Casadei E, Rességuier J, Ma J, Cain KD, Castrillo PA, Xu Z, Salinas I. A Novel Organized Nasopharynx-Associated Lymphoid Tissue in Teleosts That Expresses Molecular Markers Characteristic of Mammalian Germinal Centers. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2215-2226. [PMID: 36426979 DOI: 10.4049/jimmunol.2200396] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/16/2022] [Indexed: 01/04/2023]
Abstract
Nasal immunity is an ancient and conserved arm of the mucosal immune system in vertebrates. In teleost fish, we previously reported the presence of a nasopharynx-associated lymphoid tissue (NALT) characterized by scattered immune cells located in the trout olfactory lamellae. This diffuse NALT mounts innate and adaptive immune responses to nasal infection or vaccination. In mammals, lymphoid structures such as adenoids and tonsils support affinity maturation of the adaptive immune response in the nasopharyngeal cavity. These structures, known as organized NALT (O-NALT), have not been identified in teleost fish to date, but their evolutionary forerunners exist in sarcopterygian fish. In this study, we report that the rainbow trout nasal cavity is lined with a lymphoepithelium that extends from the most dorsal opening of the nares to the ventral nasal cavity. Within the nasal lymphoepithelium we found lymphocyte aggregates called O-NALT in this study that are composed of ∼ 56% CD4+, 24% IgM+, 16% CD8α+, and 4% IgT+ lymphocytes and that have high constitutive aicda mRNA expression. Intranasal (i.n.) vaccination with live attenuated infectious hematopoietic necrosis virus triggers expansions of B and T cells and aicda expression in response to primary i.n. vaccination. IgM+ B cells undergo proliferation and apoptosis within O-NALT upon prime but not boost i.n. vaccination. Our results suggest that novel mucosal microenvironments such as O-NALT may be involved in the affinity maturation of the adaptive immune response in early vertebrates.
Collapse
Affiliation(s)
- Benjamin Garcia
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM
| | - Fen Dong
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM.,Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, China
| | - Elisa Casadei
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM
| | - Julien Rességuier
- Section for Physiology and Cell Biology, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jie Ma
- Department of Fish and Wildlife Sciences, University of Idaho, Moscow, ID; and
| | - Kenneth D Cain
- Department of Fish and Wildlife Sciences, University of Idaho, Moscow, ID; and
| | - Pedro A Castrillo
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM.,Departamento de Anatomía, Producción Animal y Ciencias Clínicas Veterinarias, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Zhen Xu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, China
| | - Irene Salinas
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM
| |
Collapse
|
29
|
Mucosal Immune System Dysregulation in the Pathogenesis of IgA Nephropathy. Biomedicines 2022; 10:biomedicines10123027. [PMID: 36551783 PMCID: PMC9775168 DOI: 10.3390/biomedicines10123027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
The mucosal immune system, via a dynamic immune network, serves as the first line of defense against exogenous antigens. Mucosal immune system dysregulation is closely associated with the pathogenesis of immunoglobulin A nephropathy (IgAN), as illustrated by IgAN having the clinical feature of gross hematuria, often concurrent with mucosal infections. Notably, previous studies have demonstrated the efficacy of tonsillectomy and found that a targeted-release formulation of budesonide reduced proteinuria in patients with IgAN. However, it remains unclear how exogenous antigens interact with the mucosal immune system to induce or exacerbate IgAN. Thus, in this review, we focus on the dysregulation of mucosal immune response in the pathogenesis of IgAN.
Collapse
|
30
|
Ng KW, Hobbs A, Wichmann C, Victora GD, Donaldson GP. B cell responses to the gut microbiota. Adv Immunol 2022; 155:95-131. [PMID: 36357013 DOI: 10.1016/bs.ai.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Most antibody produced by humans originates from mucosal B cell responses. The rules, mechanisms, and outcomes of this process are distinct from B cell responses to infection. Within the context of the intestine, we discuss the induction of follicular B cell responses by microbiota, the development and maintenance of mucosal antibody-secreting cells, and the unusual impacts of mucosal antibody on commensal bacteria. Much remains to be learned about the interplay between B cells and the microbiota, but past and present work hints at a complex, nuanced relationship that may be critical to the way the mammalian gut fosters a beneficial microbial ecosystem.
Collapse
Affiliation(s)
- Kevin W Ng
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, United States
| | - Alvaro Hobbs
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, United States
| | - Christopher Wichmann
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, United States; Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, United States; Immune Regulation Group, Department of Pediatrics, University Medical Center Rostock, Rostock, Germany
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, United States.
| | - Gregory P Donaldson
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, United States.
| |
Collapse
|
31
|
Ruiz MJ, Siracusano G, Cottignies-Calamarte A, Tudor D, Real F, Zhu A, Pastori C, Capron C, Rosenberg AR, Temperton N, Cantoni D, Liao H, Ternette N, Moine P, Godement M, Geri G, Chiche JD, Annane D, Cramer Bordé E, Lopalco L, Bomsel M. Persistent but dysfunctional mucosal SARS-CoV-2-specific IgA and low lung IL-1β associate with COVID-19 fatal outcome: A cross-sectional analysis. Front Immunol 2022; 13:842468. [PMID: 36248831 PMCID: PMC9560774 DOI: 10.3389/fimmu.2022.842468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
The role of the mucosal pulmonary antibody response in coronavirus disease 2019 (COVID-19) outcome remains unclear. Here, we found that in bronchoalveolar lavage (BAL) samples from 48 patients with severe COVID-19-infected with the ancestral Wuhan virus, mucosal IgG and IgA specific for S1, receptor-binding domain (RBD), S2, and nucleocapsid protein (NP) emerged in BAL containing viruses early in infection and persist after virus elimination, with more IgA than IgG for all antigens tested. Furthermore, spike-IgA and spike-IgG immune complexes were detected in BAL, especially when the lung virus has been cleared. BAL IgG and IgA recognized the four main RBD variants. BAL neutralizing titers were higher early in COVID-19 when virus replicates in the lung than later in infection after viral clearance. Patients with fatal COVID-19, in contrast to survivors, developed higher levels of mucosal spike-specific IgA than IgG but lost neutralizing activities over time and had reduced IL-1β in the lung. Altogether, mucosal spike and NP-specific IgG and S1-specific IgA persisting after lung severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) clearance and low pulmonary IL-1β correlate with COVID-19 fatal outcome. Thus, mucosal SARS-CoV-2-specific antibodies may have adverse functions in addition to protective neutralization.
Collapse
Affiliation(s)
- Maria Julia Ruiz
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Gabriel Siracusano
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Andréa Cottignies-Calamarte
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Daniela Tudor
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Fernando Real
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Aiwei Zhu
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Claudia Pastori
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Claude Capron
- AP-HP, Hôpital Ambroise Paré, Service d'Hématologie, Boulogne-Billancourt, France
| | - Arielle R. Rosenberg
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
- AP-HP, Hôpital Cochin, Service de Virologie, Paris, France
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham, United Kingdom
| | - Diego Cantoni
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham, United Kingdom
| | - Hanqing Liao
- Centre for Cellular and Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicola Ternette
- Centre for Cellular and Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Pierre Moine
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - Mathieu Godement
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - Guillaume Geri
- AP-HP, Hôpital Ambroise Paré, Service de Réanimation, Boulogne-Billancourt, France
- Université de Versailles-St Quentin en Yvelines, Versailles, France
| | | | - Djillali Annane
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | | | - Lucia Lopalco
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Morgane Bomsel
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
- *Correspondence: Morgane Bomsel,
| |
Collapse
|
32
|
André S, Azarias da Silva M, Picard M, Alleaume-Buteau A, Kundura L, Cezar R, Soudaramourty C, André SC, Mendes-Frias A, Carvalho A, Capela C, Pedrosa J, Gil Castro A, Loubet P, Sotto A, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Tran TA, Zghidi-Abouzid O, Nioche P, Silvestre R, Corbeau P, Mammano F, Estaquier J. Low quantity and quality of anti-spike humoral response is linked to CD4 T-cell apoptosis in COVID-19 patients. Cell Death Dis 2022; 13:741. [PMID: 36030261 PMCID: PMC9419645 DOI: 10.1038/s41419-022-05190-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 01/21/2023]
Abstract
In addition to an inflammatory reaction, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)-infected patients present lymphopenia, which we recently reported as being related to abnormal programmed cell death. As an efficient humoral response requires CD4 T-cell help, we hypothesized that the propensity of CD4 T cells to die may impact the quantity and quality of the humoral response in acutely infected individuals. In addition to specific immunoglobulins (Ig)A, IgM, and IgG against SARS-CoV-2 nucleocapsid (N), membrane (M), and spike (S1) proteins, we assessed the quality of IgG response by measuring the avidity index. Because the S protein represents the main target for neutralization and antibody-dependent cellular cytotoxicity responses, we also analyzed anti-S-specific IgG using S-transfected cells (S-Flow). Our results demonstrated that most COVID-19 patients have a predominant IgA anti-N humoral response during the early phase of infection. This specific humoral response preceded the anti-S1 in time and magnitude. The avidity index of anti-S1 IgG was low in acutely infected individuals compared to convalescent patients. We showed that the percentage of apoptotic CD4 T cells is inversely correlated with the levels of specific IgG antibodies. These lower levels were also correlated positively with plasma levels of CXCL10, a marker of disease severity, and soluble Fas ligand that contributes to T-cell death. Finally, we found lower S-Flow responses in patients with higher CD4 T-cell apoptosis. Altogether, these results demonstrate that individuals with high levels of CD4 T-cell apoptosis and CXCL10 have a poor ability to build an efficient anti-S response. Consequently, preventing CD4 T-cell death might be a strategy for improving humoral response during the acute phase, thereby reducing COVID-19 pathogenicity.
Collapse
Affiliation(s)
- Sonia André
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France
| | - Marne Azarias da Silva
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France
| | - Morgane Picard
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France
| | - Aurélie Alleaume-Buteau
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,grid.508487.60000 0004 7885 7602Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | - Lucy Kundura
- grid.411165.60000 0004 0593 8241Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France
| | - Renaud Cezar
- grid.411165.60000 0004 0593 8241Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France
| | | | - Santa Cruz André
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal ,Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Ana Mendes-Frias
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Alexandre Carvalho
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal ,Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Carlos Capela
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal ,Department of Internal Medicine, Hospital of Braga, Braga, Portugal ,grid.512329.eClinical Academic Center-Braga, Braga, Portugal
| | - Jorge Pedrosa
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António Gil Castro
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Paul Loubet
- grid.411165.60000 0004 0593 8241Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Albert Sotto
- grid.411165.60000 0004 0593 8241Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Laurent Muller
- grid.411165.60000 0004 0593 8241Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | - Jean-Yves Lefrant
- grid.411165.60000 0004 0593 8241Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | - Claire Roger
- grid.411165.60000 0004 0593 8241Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | - Pierre-Géraud Claret
- grid.411165.60000 0004 0593 8241Urgences Médico-Chirugicales Hospitalisation, CHU de Nîmes, Nîmes, France
| | - Sandra Duvnjak
- grid.411165.60000 0004 0593 8241Service de Gérontologie et Prévention du Vieillissement, CHU de Nîmes, Nîmes, France
| | - Tu-Anh Tran
- grid.411165.60000 0004 0593 8241Service de Pédiatrie, CHU de Nîmes, Nîmes, France
| | | | - Pierre Nioche
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,grid.508487.60000 0004 7885 7602Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | - Ricardo Silvestre
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Pierre Corbeau
- grid.411165.60000 0004 0593 8241Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France ,grid.121334.60000 0001 2097 0141Institut de Génétique Humaine UMR9002 CNRS-Université de Montpellier, Montpellier, France
| | - Fabrizio Mammano
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,INSERM U1259 MAVIVH, Université de Tours, Tours, France
| | - Jérôme Estaquier
- grid.508487.60000 0004 7885 7602Université Paris Cité, INSERM U1124, F-75006 Paris, France ,CHU de Québec—Université Laval Research Center, Québec City, QC Canada
| |
Collapse
|
33
|
Tsukinoki K, Yamamoto T, Saito J, Sakaguchi W, Iguchi K, Inoue Y, Ishii S, Sato C, Yokoyama M, Shiraishi Y, Kato N, Shimada H, Makabe A, Saito A, Tanji M, Nagaoka I, Saruta J, Yamaguchi T, Kimoto S, Yamaguchi H. Prevalence of saliva immunoglobulin A antibodies reactive with severe acute respiratory syndrome coronavirus 2 among Japanese people unexposed to the virus. Microbiol Immunol 2022; 66:403-410. [PMID: 35607844 PMCID: PMC9347685 DOI: 10.1111/1348-0421.13011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/11/2022] [Accepted: 05/21/2022] [Indexed: 11/29/2022]
Abstract
While the COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a threat to public health as the number of cases and COVID-19-related deaths are increasing worldwide, the incidence of the virus infection is extremely low in Japan compared with many other countries. To explain this uncommon phenomenon, we investigated the prevalence of naturally occurring ("natural") antibodies, focusing on those of the secretory immunoglobulin A (sIgA) form, reactive with SARS-CoV-2 among Japanese people. One hundred and eighty healthy Japanese volunteers of a wide range of age who had been considered to be unexposed to SARS-CoV-2 participated in this study. Saliva samples and blood samples were collected from all of the 180 participants and 139 adults (aged ≥ 20 years) included therein, respectively. The determination of saliva IgA antibodies, mostly comprising sIgA antibodies, as well as serum IgA and immunoglobulin G antibodies, reactive with the receptor binding domain of the SARS-CoV-2 spike-1 subunit proteins was conducted using an enzyme-linked immunosorbent assay. The major findings were that 52.78% (95% confidence interval, 45.21%-60.25%) of the individuals who had not been exposed to SARS-CoV-2 were positive for saliva IgA antibodies with a wide range of levels between 0.002 and 3.272 ng/mL, and that there may be a negative trend in positivity for the antibodies according to age. As we had expected, a frequent occurrence of assumable "natural" sIgA antibodies reactive with SARS-CoV-2 among the studied Japanese participant population was observed.
Collapse
Affiliation(s)
- Keiichi Tsukinoki
- Department of Environmental Pathology, Graduate School of DentistryKanagawa Dental UniversityKanagawaJapan
| | | | | | - Wakako Sakaguchi
- Department of Environmental Pathology, Graduate School of DentistryKanagawa Dental UniversityKanagawaJapan
| | - Keiichiro Iguchi
- Department of OrthodonticsKanagawa Dental UniversityKanagawaJapan
| | - Yoshinori Inoue
- Department of Pediatric DentistryKanagawa Dental UniversityKanagawaJapan
| | - Shigeru Ishii
- Department of Advanced Oral SurgeryKDU Yokohama ClinicKanagawaJapan
| | | | - Mina Yokoyama
- Department of Pediatric DentistryKanagawa Dental UniversityKanagawaJapan
| | | | - Noriaki Kato
- EPS Research Center, EPS Holdings, Inc.TokyoJapan
| | | | - Akio Makabe
- Sites Support Section, Foods DepartmentEP Mediate Co., Ltd.TokyoJapan
| | - Akihiro Saito
- Sites Support Section, Foods DepartmentEP Mediate Co., Ltd.TokyoJapan
| | | | - Isao Nagaoka
- Department of Host Defense and Biochemical Research, Faculty of Health ScienceJuntendo UniversityTokyoJapan
| | - Juri Saruta
- Department of Education PlanningKanagawa Dental UniversityKanagawaJapan
| | | | - Shigenari Kimoto
- Department of Pediatric DentistryKanagawa Dental UniversityKanagawaJapan
| | - Hideyo Yamaguchi
- EPS Research Center, EPS Holdings, Inc.TokyoJapan
- Department of Diagnostics and Disease Control, Institute of Medical MycologyTeikyo UniversityTokyoJapan
| |
Collapse
|
34
|
Mucosal vaccine delivery: A focus on the breakthrough of specific barriers. Acta Pharm Sin B 2022; 12:3456-3474. [PMID: 35818435 PMCID: PMC9259023 DOI: 10.1016/j.apsb.2022.07.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/03/2022] [Accepted: 06/30/2022] [Indexed: 12/30/2022] Open
Abstract
Mucosal vaccines can effectively induce an immune response at the mucosal site and form the first line of defense against microbial invasion. The induced mucosal immunity includes the proliferation of effector T cells and the production of IgG and IgA antibodies, thereby effectively blocking microbial infection and transmission. However, after a long period of development, the transformation of mucosal vaccines into clinical use is still relatively slow. To date, fewer than ten mucosal vaccines have been approved. Only seven mucosal vaccines against coronavirus disease 2019 (COVID-19) are under investigation in clinical trials. A representative vaccine is the adenovirus type-5 vectored COVID-19 vaccine (Ad5-nCoV) developed by Chen and coworkers, which is currently in phase III clinical trials. The reason for the limited progress of mucosal vaccines may be the complicated mucosal barriers. Therefore, this review summarizes the characteristics of mucosal barriers and highlights strategies to overcome these barriers for effective mucosal vaccine delivery.
Collapse
|
35
|
Fukao Y, Suzuki H, Kim JS, Jeong KH, Makita Y, Kano T, Nihei Y, Nakayama M, Lee M, Kato R, Chang JM, Lee SH, Suzuki Y. Galactose-Deficient IgA1 as a Candidate Urinary Marker of IgA Nephropathy. J Clin Med 2022; 11:jcm11113173. [PMID: 35683557 PMCID: PMC9181435 DOI: 10.3390/jcm11113173] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 01/28/2023] Open
Abstract
In patients with IgA nephropathy (IgAN), circulatory IgA1 and IgA1 in the mesangial deposits contain galactose-deficient IgA1 (Gd-IgA1). Some of the Gd-IgA1 from the glomerular deposits is excreted in the urine and thus urinary Gd-IgA1 may represent a disease-specific marker. We recruited 338 Japanese biopsy-proven IgAN patients and 120 patients with other renal diseases (disease controls). Urine samples collected at the time of renal biopsy were used to measure Gd-IgA1 levels using a specific monoclonal antibody (KM55 mAb). Urinary Gd-IgA1 levels were significantly higher in patients with IgAN than in disease controls. Moreover, urinary Gd-IgA1 was significantly correlated with the severity of the histopathological parameters in IgAN patients. Next, we validated the use of urinary Gd-IgA1 levels in the other Asian cohorts. In the Korean cohort, urinary Gd-IgA1 levels were also higher in patients with IgAN than in disease controls. Even in Japanese patients with IgAN and trace proteinuria (less than 0.3 g/gCr), urinary Gd-IgA1 was detected. Thus, urinary Gd-IgA1 may be an early disease-specific biomarker useful for determining the disease activity of IgAN.
Collapse
Affiliation(s)
- Yusuke Fukao
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo 113-8421, Japan; (Y.F.); (Y.M.); (T.K.); (Y.N.); (M.N.); (M.L.); (R.K.)
| | - Hitoshi Suzuki
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo 113-8421, Japan; (Y.F.); (Y.M.); (T.K.); (Y.N.); (M.N.); (M.L.); (R.K.)
- Department of Nephrology, Juntendo University Urayasu Hospital, Chiba 279-0021, Japan
- Correspondence: (H.S.); (Y.S.)
| | - Jin Sug Kim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 130-701, Korea; (J.S.K.); (K.H.J.); (S.H.L.)
| | - Kyung Hwan Jeong
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 130-701, Korea; (J.S.K.); (K.H.J.); (S.H.L.)
| | - Yuko Makita
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo 113-8421, Japan; (Y.F.); (Y.M.); (T.K.); (Y.N.); (M.N.); (M.L.); (R.K.)
| | - Toshiki Kano
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo 113-8421, Japan; (Y.F.); (Y.M.); (T.K.); (Y.N.); (M.N.); (M.L.); (R.K.)
| | - Yoshihito Nihei
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo 113-8421, Japan; (Y.F.); (Y.M.); (T.K.); (Y.N.); (M.N.); (M.L.); (R.K.)
| | - Maiko Nakayama
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo 113-8421, Japan; (Y.F.); (Y.M.); (T.K.); (Y.N.); (M.N.); (M.L.); (R.K.)
| | - Mingfeng Lee
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo 113-8421, Japan; (Y.F.); (Y.M.); (T.K.); (Y.N.); (M.N.); (M.L.); (R.K.)
| | - Rina Kato
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo 113-8421, Japan; (Y.F.); (Y.M.); (T.K.); (Y.N.); (M.N.); (M.L.); (R.K.)
| | - Jer-Ming Chang
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
| | - Sang Ho Lee
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 130-701, Korea; (J.S.K.); (K.H.J.); (S.H.L.)
| | - Yusuke Suzuki
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo 113-8421, Japan; (Y.F.); (Y.M.); (T.K.); (Y.N.); (M.N.); (M.L.); (R.K.)
- Correspondence: (H.S.); (Y.S.)
| |
Collapse
|
36
|
Richards A, Baranova D, Mantis NJ. The prospect of orally administered monoclonal secretory IgA (SIgA) antibodies to prevent enteric bacterial infections. Hum Vaccin Immunother 2022; 18:1964317. [PMID: 34491878 PMCID: PMC9103515 DOI: 10.1080/21645515.2021.1964317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/08/2021] [Accepted: 07/30/2021] [Indexed: 12/20/2022] Open
Abstract
Eliminating diarrheal diseases as a leading cause of childhood morbidity and mortality in low- and middle-income countries (LMICs) will require multiple intervention strategies. In this review, we spotlight a series of preclinical studies investigating the potential of orally administered monoclonal secretory IgA (SIgA) antibodies (MAbs) to reduce disease associated with three enteric bacterial pathogens: Campylobacter jejuni, enterotoxigenic Escherichia coli (ETEC), and invasive Salmonella enterica serovar Typhimurium. IgA MAbs targeting bacterial surface antigens (flagella, adhesins, and lipopolysaccharide) were generated from mice, humanized mice, and human tonsillar B cells. Recombinant SIgA1 and/or SIgA2 derivates of those MAbs were purified from supernatants following transient transfection of 293 cells with plasmids encoding antibody heavy and light chains, J-chain, and secretory component (SC). When administered to mice by gavage immediately prior to (or admixed with) the bacterial challenge, SIgA MAbs reduced infection C. jejuni, ETEC, and S. Typhimurium infections. Fv-matched IgG1 MAbs by comparison were largely ineffective against C. jejuni and S. Typhimurium under the same conditions, although they were partially effective against ETEC. While these findings highlight future applications of orally administered SIgA, the studies also underscored the fundamental challenges associated with using MAbs as prophylactic tools against enteric bacterial diseases.
Collapse
Affiliation(s)
- Angelene Richards
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
| | - Danielle Baranova
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
| | - Nicholas J. Mantis
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| |
Collapse
|
37
|
Gender dimorphism in IgA subclasses in T2-high asthma. Clin Exp Med 2022:10.1007/s10238-022-00828-x. [PMID: 35467314 DOI: 10.1007/s10238-022-00828-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
Immunoglobulin A (Chan in J Allergy Clin Immunol 134:1394-14014e4, 2014), the second most abundant immunoglobulin in serum, plays an important role in mucosal homeostasis. In human serum, there are two subclasses of IgA, IgA1 (≅ 90%) and IgA2 (≅ 10%), transcribed from two distinct heavy chain constant regions. This study evaluated the serum concentrations of total IgA, IgA1, and IgA2, and total IgG, IgG1, IgG2, IgG3, and IgG4 in T2-high asthmatics compared to healthy controls and the presence of gender-related variations of immunoglobulins. Total IgA levels were increased in asthmatics compared to controls. Even more marked was the increase in total IgA in male asthmatics compared to healthy male donors. IgA1 were increased only in male, but not in female asthmatics, compared to controls. Concentrations of IgG2, but not IgG1, IgG3, and IgG4, were reduced in asthmatics compared to controls. IgG4 levels were reduced in female compared to male asthmatics. In female asthmatics, IgA and IgA1 levels were increased in postmenopause compared to premenopause. IgA concentrations were augmented in mild, but not severe asthmatics. A positive correlation was found between IgA levels and the age of patients and an inverse correlation between serum concentrations of IgA2 and IgE in asthmatics. A positive correlation between total IgA or IgA2 and IgG2 was found in asthmatics. These results highlight a gender dimorphism in IgA subclasses in male and female T2-high asthmatics. More adequate consideration of immunological gender disparity in asthma may open new opportunities in personalized medicine by optimizing diagnosis and targeted therapy.
Collapse
|
38
|
Derksen VFAM, Allaart CF, Van der Helm-Van Mil AHM, Huizinga TWJ, Toes REM, van der Woude D. In rheumatoid arthritis patients, total IgA1 and IgA2 levels are elevated: implications for the mucosal origin hypothesis. Rheumatology (Oxford) 2022; 62:407-416. [PMID: 35416963 PMCID: PMC9788813 DOI: 10.1093/rheumatology/keac237] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/03/2022] [Accepted: 04/03/2022] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Mucosal initiated immune responses may be involved in the pathophysiology of RA. The most abundant immunoglobulin at mucosal surfaces is IgA, of which two subclasses exist: IgA1 and IgA2. IgA2 is mainly present at mucosal sites and has been ascribed pro-inflammatory properties. As IgA subclasses might provide insights into mucosal involvement and pro-inflammatory mechanisms, we investigated IgA responses in sera of RA patients. METHODS In two cohorts of RA patients, the EAC and IMPROVED, total IgA1 and IgA2 were measured by ELISA. Furthermore, IgA subclass levels of RF and anti-citrullinated protein antibodies (anti-CCP2) were determined. The association of these IgA subclass levels with CRP and smoking was investigated. RESULTS Total IgA1 and IgA2 were increased in RA patients compared with healthy donors in both cohorts. This increase was more pronounced in seropositive RA vs seronegative RA. For RF and anti-CCP2, both IgA1 and IgA2 could be detected. No strong associations were found between IgA subclasses (total, RF and anti-CCP2) and CRP. In smoking RA patients, a trend towards a selective increase in total IgA2 and RF IgA1 and IgA2 was observed. CONCLUSION RA patients have raised IgA1 and IgA2 levels. No shift towards IgA2 was observed, indicating that the increase in total IgA is not due to translocation of mucosal IgA into the bloodstream. However, mucosal inflammation might play a role, given the association between smoking and total IgA2 levels. Despite its pro-inflammatory properties, IgA2 does not associate strongly with pro-inflammatory markers in RA patients.
Collapse
Affiliation(s)
- Veerle F A M Derksen
- Correspondence to: Veerle Derksen, Department of Rheumatology, Leiden University Medical Center, C1-R-041, Albinusdreef 2, PO Box 9600, 2300 RC Leiden, The Netherlands. E-mail:
| | | | - Annette H M Van der Helm-Van Mil
- Department of Rheumatology, Leiden University Medical Center, Leiden,Department of Rheumatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tom W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden
| | - René E M Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden
| | | |
Collapse
|
39
|
Abstract
Hypertension is a worldwide problem with major impacts on health including morbidity and mortality, as well as consumption of health care resources. Nearly 50% of American adults have high blood pressure, and this rate is rising. Even with multiple antihypertensive drugs and aggressive lifestyle modifications, blood pressure is inadequately controlled in about 1 of 5 hypertensive individuals. This review highlights a hypothesis for hypertension that suggests alternative mechanisms for blood pressure elevation and maintenance. A better understanding of these mechanisms could open avenues for more successful treatments. The hypothesis accounts for recent understandings of the involvement of gut physiology, gut microbiota, and neuroinflammation in hypertension. It includes bidirectional communication between gut microbiota and gut epithelium in the gut-brain axis that is involved in regulation of autonomic nervous system activity and blood pressure control. Dysfunction of this gut-brain axis, including dysbiosis of gut microbiota, gut epithelial dysfunction, and deranged input to the brain, contributes to hypertension via inflammatory mediators, metabolites, bacteria in the circulation, afferent information alterations, etc resulting in neuroinflammation and unbalanced autonomic nervous system activity that elevates blood pressure. This in turn negatively affects gut function and its microbiota exacerbating the problem. We focus this review on the gut-brain axis hypothesis for hypertension and possible contribution to racial disparities in hypertension. A novel idea, that immunoglobulin A-coated bacteria originating in the gut with access to the brain could be involved in hypertension, is raised. Finally, minocycline, with its anti-inflammatory and antimicrobial properties, is evaluated as a potential antihypertensive drug acting on this axis.
Collapse
Affiliation(s)
- Elaine M Richards
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Jing Li
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Bruce R Stevens
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Carl J Pepine
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
40
|
Zachova K, Jemelkova J, Kosztyu P, Ohyama Y, Takahashi K, Zadrazil J, Orsag J, Matousovic K, Galuszkova D, Petejova N, Mestecky J, Raska M. Galactose-Deficient IgA1 B cells in the Circulation of IgA Nephropathy Patients Carry Preferentially Lambda Light Chains and Mucosal Homing Receptors. J Am Soc Nephrol 2022; 33:908-917. [PMID: 35115327 PMCID: PMC9063893 DOI: 10.1681/asn.2021081086] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Background: Immunoglobulin A nephropathy (IgAN) primary glomerulonephritis is characterized by the deposition of circulating immune complexes (CIC) composed of polymeric (p)IgA1 molecules with altered O-glycans (Gd-IgA1) and anti-glycan antibodies in the kidney mesangium. The mesangial IgA deposits and serum IgA1 contain predominantly lambda (λ) light (L) chain, but the nature and origin of such IgA remains enigmatic Methods: We analyzed λ L chain expression in peripheral blood B cells of 30 IgAN patients, 30 healthy controls (HC), and 18 membranous nephropathy patients selected as disease controls (Non-IgAN). Results: In comparison to HC and Non-IgAN, in peripheral blood surface/membrane bound (mb)-Gd-IgA1+ cells from IgAN patients express predominantly λ L chain. In contrast, total mb-IgA+, mb-IgG+, and mb-IgM+ cells were preferentially positive for kappa (κ) L chain, in all analyzed groups. Although minor in comparison to κ L chain, λ L chain subsets of mb-IgG+, mb-IgM,+ and mb-IgA+ cells were significantly enriched in IgAN in comparison to Non-IgAN and/or HC. In contrast to HC, the peripheral blood of IgAN patients was enriched for λ+ mb-Gd-IgA1,+ CCR10,+ and CCR9+ cells, which preferentially home to the upper respiratory and digestive tract, respectively. Furthermore, we observed that mb-Gd-IgA1+ cell populations comprise more CD138+ cells and plasmablasts (CD38+) in comparison to total mb-IgA+ cells. Conclusions: Peripheral blood of IgAN patients is enriched for migratory λ+ mb-GdIgA1+ B cells, with the potential to home to mucosal sites where Gd-IgA1 could be produced during local respiratory or digestive tract infections.
Collapse
Affiliation(s)
- Katerina Zachova
- K Zachova, Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Jana Jemelkova
- J Jemelkova, Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Petr Kosztyu
- P Kosztyu, Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Yukako Ohyama
- Y Ohyama, Department of Biomedical Molecular Sciences, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Kazuo Takahashi
- K Takahashi, Department of Biomedical Molecular Sciences, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Josef Zadrazil
- J Zadrazil, Department of Internal Medicine III Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Jiri Orsag
- J Orsag, Department of Internal Medicine III Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Karel Matousovic
- K Matousovic, Department of Medicine, Charles University Second Faculty of Medicine, Praha, Czech Republic
| | - Dana Galuszkova
- D Galuszkova, Department of Transfusion Medicine, University Hospital Olomouc, Olomouc, Czech Republic
| | - Nadezda Petejova
- N Petejova, Department of Internal Medicine III Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Jiri Mestecky
- J Mestecky, Departments of Microbiology and Medicine, The University of Alabama at Birmingham, Birmingham, United States
| | - Milan Raska
- M Raska, Department of Immunology, Palacky University Olomouc, Olomouc, Czech Republic
| |
Collapse
|
41
|
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
|
42
|
Yamamoto Y, Carreras J, Shimizu T, Kakizaki M, Kikuti YY, Roncador G, Nakamura N, Kotani A. Anti-HBV drug entecavir ameliorates DSS-induced colitis through PD-L1 induction. Pharmacol Res 2022; 179:105918. [PMID: 35031477 DOI: 10.1016/j.phrs.2021.105918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 12/30/2022]
Abstract
PD-L1-mediated signaling is one of the major processes that regulate local inflammatory responses in the gut. To date, protective effects against colitis through direct Fc-fused PD-L1 administration or indirect PD-L1 induction by probiotics have been reported. We have previously shown that the anti-HBV drug entecavir (ETV) induces PD-L1 expression in human hepatocytes. In the present study, we investigated whether ETV induces PD-L1 expression in intestinal cells and provides a protective effect against DSS-induced colitis. ETV induced PD-L1 expression in epithelial cells, rather than T and B cells, improving the symptoms of colitis. In the mechanistic analysis, Th17 cell differentiation was inhibited and B cell infiltration into the lamina propria was reduced. In addition, PD-L1 expression was positively correlated with Foxp3 or CSF1-R. In conclusion, ETV upregulated PD-L1 expression in epithelial cells and ameliorated inflammation in DSS-induced colitis. These results suggest that ETV may be a potential therapeutic agent as a PD-L1 enhancer for the treatment of human IBD.
Collapse
Affiliation(s)
- Yuichiro Yamamoto
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Japan. 259-1193; Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan. 259-1193
| | - Joaquim Carreras
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan. 259-1193
| | - Takanobu Shimizu
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan. 259-1193
| | - Masatoshi Kakizaki
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Japan. 259-1193; Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan. 259-1193
| | - Yara Yukie Kikuti
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan. 259-1193
| | - Giovanna Roncador
- Monoclonal Antibodies Unit. Spanish National Cancer Research Institute (CNIO). Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Naoya Nakamura
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan. 259-1193
| | - Ai Kotani
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Japan. 259-1193; Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan. 259-1193.
| |
Collapse
|
43
|
Nagasawa Y, Misaki T, Ito S, Naka S, Wato K, Nomura R, Matsumoto-Nakano M, Nakano K. Title IgA Nephropathy and Oral Bacterial Species Related to Dental Caries and Periodontitis. Int J Mol Sci 2022; 23:725. [PMID: 35054910 PMCID: PMC8775524 DOI: 10.3390/ijms23020725] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
A relationship between IgA nephropathy (IgAN) and bacterial infection has been suspected. As IgAN is a chronic disease, bacteria that could cause chronic infection in oral areas might be pathogenetic bacteria candidates. Oral bacterial species related to dental caries and periodontitis should be candidates because these bacteria are well known to be pathogenic in chronic dental disease. Recently, several reports have indicated that collagen-binding protein (cnm)-(+) Streptococcs mutans is relate to the incidence of IgAN and the progression of IgAN. Among periodontal bacteria, Treponema denticola, Porphyromonas gingivalis and Campylobacte rectus were found to be related to the incidence of IgAN. These bacteria can cause IgAN-like histological findings in animal models. While the connection between oral bacterial infection, such as infection with S. mutans and periodontal bacteria, and the incidence of IgAN remains unclear, these bacterial infections might cause aberrantly glycosylated IgA1 in nasopharynx-associated lymphoid tissue, which has been reported to cause IgA deposition in mesangial areas in glomeruli, probably through the alteration of microRNAs related to the expression of glycosylation enzymes. The roles of other factors related to the incidence and progression of IgA, such as genes and cigarette smoking, can also be explained from the perspective of the relationship between these factors and oral bacteria. This review summarizes the relationship between IgAN and oral bacteria, such as cnm-(+) S. mutans and periodontal bacteria.
Collapse
Affiliation(s)
- Yasuyuki Nagasawa
- Department of General Internal Medicine, Hyogo College of Medicine, Nishinomiya 663-8501, Hyogo, Japan
| | - Taro Misaki
- Division of Nephrology, Seirei Hamamatsu General Hospital, Hamamatsu 430-8558, Shizuoka, Japan;
- Department of Nursing, Faculty of Nursing, Seirei Christopher University, Hamamatsu 433-8558, Shizuoka, Japan
| | - Seigo Ito
- Department of Internal Medicine, Japan Self-Defense Gifu Hospital, Kakamigahara 502-0817, Gifu, Japan;
| | - Shuhei Naka
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Okayama, Japan; (S.N.); (M.M.-N.)
| | - Kaoruko Wato
- Department of Pediatric Dentistry, Division of Oral Infection and Disease Control, Osaka University Graduate School of Dentistry, Suita 565-0871, Osaka, Japan; (K.W.); (R.N.); (K.N.)
| | - Ryota Nomura
- Department of Pediatric Dentistry, Division of Oral Infection and Disease Control, Osaka University Graduate School of Dentistry, Suita 565-0871, Osaka, Japan; (K.W.); (R.N.); (K.N.)
| | - Michiyo Matsumoto-Nakano
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Okayama, Japan; (S.N.); (M.M.-N.)
| | - Kazuhiko Nakano
- Department of Pediatric Dentistry, Division of Oral Infection and Disease Control, Osaka University Graduate School of Dentistry, Suita 565-0871, Osaka, Japan; (K.W.); (R.N.); (K.N.)
| |
Collapse
|
44
|
James KR, Elmentaite R, Teichmann SA, Hold GL. Redefining intestinal immunity with single-cell transcriptomics. Mucosal Immunol 2022; 15:531-541. [PMID: 34848830 PMCID: PMC8630196 DOI: 10.1038/s41385-021-00470-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 02/04/2023]
Abstract
The intestinal immune system represents the largest collection of immune cells in the body and is continually exposed to antigens from food and the microbiota. Here we discuss the contribution of single-cell transcriptomics in shaping our understanding of this complex system. We consider the impact on resolving early intestine development, engagement with the neighbouring microbiota, diversity of intestinal immune cells, compartmentalisation within the intestines and interactions with non-immune cells. Finally, we offer a perspective on open questions about gut immunity that evolving single-cell technologies are well placed to address.
Collapse
Affiliation(s)
- Kylie Renee James
- grid.415306.50000 0000 9983 6924Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW 2010 Australia ,grid.1005.40000 0004 4902 0432School of Medical Sciences, University of New South Wales, Sydney, NSW 2006 Australia
| | - Rasa Elmentaite
- grid.10306.340000 0004 0606 5382Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA UK
| | - Sarah Amalia Teichmann
- grid.10306.340000 0004 0606 5382Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA UK ,grid.5335.00000000121885934Theory of Condensed Matter Group, Cavendish Laboratory/Department of Physics, University of Cambridge, Cambridge, NSW CB3 0HE UK
| | - Georgina Louise Hold
- grid.1005.40000 0004 4902 0432University of New South Wales Microbiome Research Centre, Sydney, NSW 2217 Australia
| |
Collapse
|
45
|
McIlwain DR, Chen H, Rahil Z, Bidoki NH, Jiang S, Bjornson Z, Kolhatkar NS, Martinez CJ, Gaudillière B, Hedou J, Mukherjee N, Schürch CM, Trejo A, Affrime M, Bock B, Kim K, Liebowitz D, Aghaeepour N, Tucker SN, Nolan GP. Human influenza virus challenge identifies cellular correlates of protection for oral vaccination. Cell Host Microbe 2021; 29:1828-1837.e5. [PMID: 34784508 PMCID: PMC8665113 DOI: 10.1016/j.chom.2021.10.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/16/2021] [Accepted: 10/21/2021] [Indexed: 01/23/2023]
Abstract
Developing new influenza vaccines with improved performance and easier administration routes hinges on defining correlates of protection. Vaccine-elicited cellular correlates of protection for influenza in humans have not yet been demonstrated. A phase-2 double-blind randomized placebo and active (inactivated influenza vaccine) controlled study provides evidence that a human-adenovirus-5-based oral influenza vaccine tablet (VXA-A1.1) can protect from H1N1 virus challenge in humans. Mass cytometry characterization of vaccine-elicited cellular immune responses identified shared and vaccine-type-specific responses across B and T cells. For VXA-A1.1, the abundance of hemagglutinin-specific plasmablasts and plasmablasts positive for integrin α4β7, phosphorylated STAT5, or lacking expression of CD62L at day 8 were significantly correlated with protection from developing viral shedding following virus challenge at day 90 and contributed to an effective machine learning model of protection. These findings reveal the characteristics of vaccine-elicited cellular correlates of protection for an oral influenza vaccine.
Collapse
Affiliation(s)
- David R McIlwain
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; WCCT Global, Cypress, CA, USA.
| | - Han Chen
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zainab Rahil
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Neda Hajiakhoond Bidoki
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Biomedical Informatics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sizun Jiang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Zach Bjornson
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Julien Hedou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nilanjan Mukherjee
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian M Schürch
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA; Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Angelica Trejo
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Kenneth Kim
- Ark Clinical Research, LLC, Long Beach, CA, USA
| | | | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Biomedical Informatics, Stanford University School of Medicine, Stanford, CA, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Garry P Nolan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
46
|
Zhong Z, Nan K, Weng M, Yue Y, Zhou W, Wang Z, Chu Y, Liu R, Miao C. Pro- and Anti- Effects of Immunoglobulin A- Producing B Cell in Tumors and Its Triggers. Front Immunol 2021; 12:765044. [PMID: 34868013 PMCID: PMC8640120 DOI: 10.3389/fimmu.2021.765044] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/01/2021] [Indexed: 12/18/2022] Open
Abstract
B cells are well known as key mediators of humoral immune responses via the production of antibodies. Immunoglobulin A (IgA) is the most abundantly produced antibody isotype and provides the first line of immune protection at mucosal surfaces. However, IgA has long been a divisive molecule with respect to tumor progression. IgA exerts anti- or pro-tumor effect in different tumor types. In this review, we summarize emerging evidence regarding the production and effects of IgA and IgA+ cells in the tumor microenvironment (TME). Moreover, we discuss that the TME cytokines, host diet, microbiome, and metabolites play a pivotal role in controlling the class-switch recombination (CSR) of IgA. The analysis of intratumoral Ig repertoires and determination of metabolites that influence CSR may help establish novel therapeutic targets for the treatment of cancers.
Collapse
Affiliation(s)
- Ziwen Zhong
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ke Nan
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meilin Weng
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Yue
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenchang Zhou
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiqiang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Shanghai Fifth People's Hospital and Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ronghua Liu
- Shanghai Fifth People's Hospital and Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Martelli P, Saleri R, Andrani M, Cavalli V, De Angelis E, Ferrari L, Borghetti P. Immune B cell responsiveness to single-dose intradermal vaccination against Mycoplasma hyopneumoniae. Res Vet Sci 2021; 141:66-75. [PMID: 34688042 DOI: 10.1016/j.rvsc.2021.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/21/2021] [Accepted: 10/05/2021] [Indexed: 10/20/2022]
Abstract
Mycoplasma hyopneumoniae is a major pathogen affecting pig herds and vaccination is the most utilized approach, despite providing partial protection. Age at vaccination, the delivery route, and vaccination protocol can influence vaccine efficacy. The influence of age and the presence of maternally-derived antibodies at vaccination on single-dose needle-less intradermal (ID) administration of an inactivated bacterin-based vaccine (Porcilis® M Hyo ID Once) were assessed in conventional pigs under field conditions. The induction of IgA+ and IgG+ B cell responses and the expression of the activation markers TLR2, TLR7, CCR9, and CCR10 were determined in PBMC. Vaccination at 4 weeks efficiently elicited an anamnestic antibody response associated with TLR2 and TLR7 upregulation. Although animals vaccinated at 1 week did not show seroconversion and a recall response upon infection, the responsiveness of Mycoplasma-recalled IgA+ B cells suggests the activation of mucosal immune cells after vaccination and infection. Vaccination at 1 week induced TLR2, TLR7, and CCR9 upregulation, suggesting the potential for systemic and local activation of immune cell trafficking between blood and target tissues. Vaccination at 4 weeks induced a CCR10 increase, suggesting that recalled IgA+ and IgG+ B cells can display an activated status upon infection. The antibody response after Mycoplasma infection in 4-week-old ID-vaccinated pigs was associated with TLR2 and CCR10 increases, confirming the potential use of this vaccination schedule for the safe and efficient delivery of single-dose M. hyopneumoniae vaccines. ID vaccination, especially at 4 weeks, was associated with a great degree of protection against enzootic pneumonia (EP)-like lung lesions.
Collapse
Affiliation(s)
- Paolo Martelli
- Department of Veterinary Science, University of Parma, Strada del Taglio, 10, Parma 43126, Italy.
| | - Roberta Saleri
- Department of Veterinary Science, University of Parma, Strada del Taglio, 10, Parma 43126, Italy.
| | - Melania Andrani
- Department of Veterinary Science, University of Parma, Strada del Taglio, 10, Parma 43126, Italy.
| | - Valeria Cavalli
- Department of Veterinary Science, University of Parma, Strada del Taglio, 10, Parma 43126, Italy.
| | - Elena De Angelis
- Department of Veterinary Science, University of Parma, Strada del Taglio, 10, Parma 43126, Italy.
| | - Luca Ferrari
- Department of Veterinary Science, University of Parma, Strada del Taglio, 10, Parma 43126, Italy.
| | - Paolo Borghetti
- Department of Veterinary Science, University of Parma, Strada del Taglio, 10, Parma 43126, Italy.
| |
Collapse
|
48
|
Popov J, Caputi V, Nandeesha N, Rodriguez DA, Pai N. Microbiota-Immune Interactions in Ulcerative Colitis and Colitis Associated Cancer and Emerging Microbiota-Based Therapies. Int J Mol Sci 2021; 22:11365. [PMID: 34768795 PMCID: PMC8584103 DOI: 10.3390/ijms222111365] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/02/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic autoimmune disorder affecting the colonic mucosa. UC is a subtype of inflammatory bowel disease along with Crohn's disease and presents with varying extraintestinal manifestations. No single etiology for UC has been found, but a combination of genetic and environmental factors is suspected. Research has focused on the role of intestinal dysbiosis in the pathogenesis of UC, including the effects of dysbiosis on the integrity of the colonic mucosal barrier, priming and regulation of the host immune system, chronic inflammation, and progression to tumorigenesis. Characterization of key microbial taxa and their implications in the pathogenesis of UC and colitis-associated cancer (CAC) may present opportunities for modulating intestinal inflammation through microbial-targeted therapies. In this review, we discuss the microbiota-immune crosstalk in UC and CAC, as well as the evolution of microbiota-based therapies.
Collapse
Affiliation(s)
- Jelena Popov
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada;
- College of Medicine and Health, University College Cork, T12 XF62 Cork, Ireland
| | - Valentina Caputi
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Nandini Nandeesha
- School of Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland;
| | | | - Nikhil Pai
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada;
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
49
|
Mahallawi WH, Aljeraisi TM. Infection with SARS-CoV-2 primes immunological memory in human nasal-associated lymphoid tissue. Clin Immunol 2021; 231:108850. [PMID: 34506944 PMCID: PMC8423672 DOI: 10.1016/j.clim.2021.108850] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/24/2021] [Accepted: 09/04/2021] [Indexed: 12/03/2022]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, has resulted in considerable morbidity and mortality in humans. Little is known regarding the development of immunological memory following SARS-CoV-2 infection or whether immunological memory can provide long-lasting protection against reinfection. Urgent need for vaccines is a considerable issue for all governments worldwide. METHODS A total of 39 patients were recruited in this study. Tonsillar mononuclear cells (MNCs) were co-cultured in RPMI medium and stimulated with the full-length SARS-CoV-2 spike protein in the presence and absence of a CpG-DNA adjuvant. An enzyme-linked immunosorbent assay (ELISA) was utilised to measure the specific antibody response to the spike protein in the cell culture supernatants. RESULTS The SARS-CoV-2 spike protein primed a potent memory B cell-mediated immune response in nasal-associated lymphoid tissue (NALT) from patients previously infected with the virus. Additionally, spike protein combined with the CpG-DNA adjuvant induced a significantly increased level of specific anti-spike protein IgG antibody compared with the spike protein alone (p < 0.0001, n = 24). We also showed a strong positive correlation between the specific anti-spike protein IgG antibody level in a serum samples and that produced by MNCs derived from the same COVID-19-recovered patients following stimulation (r = 0.76, p = 0.0002, n = 24). CONCLUSION Individuals with serological evidence of previous SARS-CoV-2 exposure showed a significant anti-spike protein-specific memory humoral immune response to the viral spike protein upon stimulation. Additionally, our results demonstrated the functional response of NALT-derived MNCs to the viral spike protein. CpG-DNA adjuvant combined with spike protein induced significantly stronger humoral immune responses than the spike protein alone. These data indicate that the S protein antigen combined with CpG-DNA adjuvant could be used as a future vaccine candidate.
Collapse
Affiliation(s)
- Waleed H Mahallawi
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia.
| | - Talal M Aljeraisi
- Otorhinolaryngology, Head& Neck Surgery Department, Faculty of Medicine, Taibah University, Madinah, Saudi Arabia.
| |
Collapse
|
50
|
Pathogenesis of IgA Nephropathy: Current Understanding and Implications for Development of Disease-Specific Treatment. J Clin Med 2021; 10:jcm10194501. [PMID: 34640530 PMCID: PMC8509647 DOI: 10.3390/jcm10194501] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022] Open
Abstract
IgA nephropathy, initially described in 1968 as a kidney disease with glomerular “intercapillary deposits of IgA-IgG”, has no disease-specific treatment and is a common cause of kidney failure. Clinical observations and laboratory analyses suggest that IgA nephropathy is an autoimmune disease wherein the kidneys are damaged as innocent bystanders due to deposition of IgA1-IgG immune complexes from the circulation. A multi-hit hypothesis for the pathogenesis of IgA nephropathy describes four sequential steps in disease development. Specifically, patients with IgA nephropathy have elevated circulating levels of IgA1 with some O-glycans deficient in galactose (galactose-deficient IgA1) and these IgA1 glycoforms are recognized as autoantigens by unique IgG autoantibodies, resulting in formation of circulating immune complexes, some of which deposit in glomeruli and activate mesangial cells to induce kidney injury. This proposed mechanism is supported by observations that (i) glomerular immunodeposits in patients with IgA nephropathy are enriched for galactose-deficient IgA1 glycoforms and the corresponding IgG autoantibodies; (ii) circulatory levels of galactose-deficient IgA1 and IgG autoantibodies predict disease progression; and (iii) pathogenic potential of galactose-deficient IgA1 and IgG autoantibodies was demonstrated in vivo. Thus, a better understanding of the structure–function of these immunoglobulins as autoantibodies and autoantigens will enable development of disease-specific treatments.
Collapse
|