101
|
Ewald DR, Sumner SCJ. Human microbiota, blood group antigens, and disease. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2018; 10:e1413. [PMID: 29316320 PMCID: PMC5902424 DOI: 10.1002/wsbm.1413] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/05/2017] [Accepted: 11/09/2017] [Indexed: 12/11/2022]
Abstract
Far from being just "bugs in our guts," the microbiota interacts with the body in previously unimagined ways. Research into the genome and the microbiome has revealed that the human body and the microbiota have a long-established but only recently recognized symbiotic relationship; homeostatic balance between them regulates body function. That balance is fragile, easily disturbed, and plays a fundamental role in human health-our very survival depends on the healthy functioning of these microorganisms. Increasing rates of cardiovascular, autoimmune, and inflammatory diseases, as well as epidemics in obesity and diabetes in recent decades are believed to be explained, in part, by unintended effects on the microbiota from vaccinations, poor diets, environmental chemicals, indiscriminate antibiotic use, and "germophobia." Discovery and exploration of the brain-gut-microbiota axis have provided new insights into functional diseases of the gut, autoimmune and stress-related disorders, and the role of probiotics in treating certain affective disorders; it may even explain some aspects of autism. Research into dietary effects on the human gut microbiota led to its classification into three proposed enterotypes, but also revealed the surprising role of blood group antigens in shaping those populations. Blood group antigens have previously been associated with disease risks; their subsequent association with the microbiota may reveal mechanisms that lead to development of nutritional interventions and improved treatment modalities. Further exploration of associations between specific enteric microbes and specific metabolites will foster new dietary interventions, treatment modalities, and genetic therapies, and inevitably, their application in personalized healthcare strategies. This article is categorized under: Laboratory Methods and Technologies > Metabolomics Translational, Genomic, and Systems Medicine > Translational Medicine Physiology > Mammalian Physiology in Health and Disease.
Collapse
Affiliation(s)
- D Rose Ewald
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, 28081
| | - Susan CJ Sumner
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, 28081
| |
Collapse
|
102
|
Ichinohe T, Miyama T, Kawase T, Honjo Y, Kitaura K, Sato H, Shin-I T, Suzuki R. Next-Generation Immune Repertoire Sequencing as a Clue to Elucidate the Landscape of Immune Modulation by Host-Gut Microbiome Interactions. Front Immunol 2018; 9:668. [PMID: 29666626 PMCID: PMC5891584 DOI: 10.3389/fimmu.2018.00668] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 03/19/2018] [Indexed: 12/23/2022] Open
Abstract
The human immune system is a fine network consisted of the innumerable numbers of functional cells that balance the immunity and tolerance against various endogenous and environmental challenges. Although advances in modern immunology have revealed a role of many unique immune cell subsets, technologies that enable us to capture the whole landscape of immune responses against specific antigens have been not available to date. Acquired immunity against various microorganisms including host microbiome is principally founded on T cell and B cell populations, each of which expresses antigen-specific receptors that define a unique clonotype. Over the past several years, high-throughput next-generation sequencing has been developed as a powerful tool to profile T- and B-cell receptor repertoires in a given individual at the single-cell level. Sophisticated immuno-bioinformatic analyses by use of this innovative methodology have been already implemented in clinical development of antibody engineering, vaccine design, and cellular immunotherapy. In this article, we aim to discuss the possible application of high-throughput immune receptor sequencing in the field of nutritional and intestinal immunology. Although there are still unsolved caveats, this emerging technology combined with single-cell transcriptomics/proteomics provides a critical tool to unveil the previously unrecognized principle of host–microbiome immune homeostasis. Accumulation of such knowledge will lead to the development of effective ways for personalized immune modulation through deeper understanding of the mechanisms by which the intestinal environment affects our immune ecosystem.
Collapse
Affiliation(s)
- Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima, Japan
| | - Takahiko Miyama
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima, Japan
| | - Takakazu Kawase
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima, Japan
| | - Yasuko Honjo
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima, Japan
| | | | | | | | - Ryuji Suzuki
- Repertoire Genesis Incorporation, Ibaraki, Japan.,Department of Rheumatology and Clinical Immunology, Clinical Research Center for Rheumatology and Allergy, National Hospital Organization Sagamihara Hospital, Sagamihara, Japan
| |
Collapse
|
103
|
Jahnsen FL, Bækkevold ES, Hov JR, Landsverk OJ. Do Long-Lived Plasma Cells Maintain a Healthy Microbiota in the Gut? Trends Immunol 2018; 39:196-208. [DOI: 10.1016/j.it.2017.10.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/25/2017] [Accepted: 10/25/2017] [Indexed: 02/07/2023]
|
104
|
Macpherson AJ, Yilmaz B, Limenitakis JP, Ganal-Vonarburg SC. IgA Function in Relation to the Intestinal Microbiota. Annu Rev Immunol 2018; 36:359-381. [PMID: 29400985 DOI: 10.1146/annurev-immunol-042617-053238] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
IgA is the dominant immunoglobulin isotype produced in mammals, largely secreted across the intestinal mucosal surface. Although induction of IgA has been a hallmark feature of microbiota colonization following colonization in germ-free animals, until recently appreciation of the function of IgA in host-microbial mutualism has depended mainly on indirect evidence of alterations in microbiota composition or penetration of microbes in the absence of somatic mutations in IgA (or compensatory IgM). Highly parallel sequencing techniques that enable high-resolution analysis of either microbial consortia or IgA sequence diversity are now giving us new perspectives on selective targeting of microbial taxa and the trajectory of IgA diversification according to induction mechanisms, between different individuals and over time. The prospects are to link the range of diversified IgA clonotypes to specific antigenic functions in modulating the microbiota composition, position and metabolism to ensure host mutualism.
Collapse
Affiliation(s)
- Andrew J Macpherson
- Maurice Müller Laboratories (Department of Biomedical Research), University of Bern, 3008 Bern, Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital, 3010 Bern, Switzerland;
| | - Bahtiyar Yilmaz
- Maurice Müller Laboratories (Department of Biomedical Research), University of Bern, 3008 Bern, Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital, 3010 Bern, Switzerland;
| | - Julien P Limenitakis
- Maurice Müller Laboratories (Department of Biomedical Research), University of Bern, 3008 Bern, Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital, 3010 Bern, Switzerland;
| | - Stephanie C Ganal-Vonarburg
- Maurice Müller Laboratories (Department of Biomedical Research), University of Bern, 3008 Bern, Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital, 3010 Bern, Switzerland;
| |
Collapse
|
105
|
Barteneva NS, Baiken Y, Fasler-Kan E, Alibek K, Wang S, Maltsev N, Ponomarev ED, Sautbayeva Z, Kauanova S, Moore A, Beglinger C, Vorobjev IA. Extracellular vesicles in gastrointestinal cancer in conjunction with microbiota: On the border of Kingdoms. Biochim Biophys Acta Rev Cancer 2017; 1868:372-393. [DOI: 10.1016/j.bbcan.2017.06.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/26/2017] [Accepted: 06/26/2017] [Indexed: 12/16/2022]
|
106
|
Lycke NY, Bemark M. The regulation of gut mucosal IgA B-cell responses: recent developments. Mucosal Immunol 2017; 10:1361-1374. [PMID: 28745325 DOI: 10.1038/mi.2017.62] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/20/2017] [Indexed: 02/04/2023]
Abstract
The majority of activated B cells differentiate into IgA plasma cells, with the gut being the largest producer of immunoglobulin in the body. Secretory IgA antibodies have numerous critical functions of which protection against infections and the role for establishing a healthy microbiota appear most important. Expanding our knowledge of the regulation of IgA B-cell responses and how effective mucosal vaccines can be designed are of critical importance. Here we discuss recent developments in the field that shed light on the uniqueness and complexity of mucosal IgA responses and the control of protective IgA responses in the gut, specifically.
Collapse
Affiliation(s)
- N Y Lycke
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - M Bemark
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
107
|
Bunker JJ, Erickson SA, Flynn TM, Henry C, Koval JC, Meisel M, Jabri B, Antonopoulos DA, Wilson PC, Bendelac A. Natural polyreactive IgA antibodies coat the intestinal microbiota. Science 2017; 358:science.aan6619. [PMID: 28971969 DOI: 10.1126/science.aan6619] [Citation(s) in RCA: 314] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/19/2017] [Indexed: 12/11/2022]
Abstract
Large quantities of immunoglobulin A (IgA) are constitutively secreted by intestinal plasma cells to coat and contain the commensal microbiota, yet the specificity of these antibodies remains elusive. Here we profiled the reactivities of single murine IgA plasma cells by cloning and characterizing large numbers of monoclonal antibodies. IgAs were not specific to individual bacterial taxa but rather polyreactive, with broad reactivity to a diverse, but defined, subset of microbiota. These antibodies arose at low frequencies among naïve B cells and were selected into the IgA repertoire upon recirculation in Peyer's patches. This selection process occurred independent of microbiota or dietary antigens. Furthermore, although some IgAs acquired somatic mutations, these did not substantially influence their reactivity. These findings reveal an endogenous mechanism driving homeostatic production of polyreactive IgAs with innate specificity to microbiota.
Collapse
Affiliation(s)
- Jeffrey J Bunker
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Steven A Erickson
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Theodore M Flynn
- Biosciences Division, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Carole Henry
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jason C Koval
- Biosciences Division, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Marlies Meisel
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Bana Jabri
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Dionysios A Antonopoulos
- Biosciences Division, Argonne National Laboratory, Argonne, IL 60439, USA.,Department of Medicine, University of Chicago, Chicago, IL 60637, USA.,Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Patrick C Wilson
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.,Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Albert Bendelac
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA. .,Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
108
|
Magri G, Comerma L, Pybus M, Sintes J, Lligé D, Segura-Garzón D, Bascones S, Yeste A, Grasset EK, Gutzeit C, Uzzan M, Ramanujam M, van Zelm MC, Albero-González R, Vazquez I, Iglesias M, Serrano S, Márquez L, Mercade E, Mehandru S, Cerutti A. Human Secretory IgM Emerges from Plasma Cells Clonally Related to Gut Memory B Cells and Targets Highly Diverse Commensals. Immunity 2017; 47:118-134.e8. [PMID: 28709802 PMCID: PMC5519504 DOI: 10.1016/j.immuni.2017.06.013] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 04/07/2017] [Accepted: 06/16/2017] [Indexed: 12/16/2022]
Abstract
Secretory immunoglobulin A (SIgA) enhances host-microbiota symbiosis, whereas SIgM remains poorly understood. We found that gut IgM+ plasma cells (PCs) were more abundant in humans than mice and clonally related to a large repertoire of memory IgM+ B cells disseminated throughout the intestine but rare in systemic lymphoid organs. In addition to sharing a gut-specific gene signature with memory IgA+ B cells, memory IgM+ B cells were related to some IgA+ clonotypes and switched to IgA in response to T cell-independent or T cell-dependent signals. These signals induced abundant IgM which, together with SIgM from clonally affiliated PCs, recognized mucus-embedded commensals. Bacteria recognized by human SIgM were dually coated by SIgA and showed increased richness and diversity compared to IgA-only-coated or uncoated bacteria. Thus, SIgM may emerge from pre-existing memory rather than newly activated naive IgM+ B cells and could help SIgA to anchor highly diverse commensal communities to mucus. IgM+ PCs generating SIgM are relatively abundant in human but not mouse gut IgM+ PCs clonally relate to a large gut repertoire of memory IgM+ B cells Gut memory IgM+ B cells express a tissue-specific signature and can switch to IgA Human but not mouse SIgM binds a highly diverse microbiota dually coated by SIgA
Collapse
Affiliation(s)
- Giuliana Magri
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain.
| | - Laura Comerma
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Marc Pybus
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Jordi Sintes
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - David Lligé
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Daniel Segura-Garzón
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Sabrina Bascones
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Ada Yeste
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain
| | - Emilie K Grasset
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Cindy Gutzeit
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mathieu Uzzan
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Meera Ramanujam
- Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Menno C van Zelm
- Department of Immunology and Pathology, Monash University and Alfred Hospital, Melbourne, VIC 3004, Australia
| | | | - Ivonne Vazquez
- Pathology Department, Hospital del Mar, Barcelona 08003, Spain
| | - Mar Iglesias
- Pathology Department, Hospital del Mar, Barcelona 08003, Spain; Universitat Autònoma de Barcelona, Barcelona 08003, Spain
| | - Sergi Serrano
- Pathology Department, Hospital del Mar, Barcelona 08003, Spain; Universitat Autònoma de Barcelona, Barcelona 08003, Spain
| | - Lucía Márquez
- Department of Gastroenterology, Hospital del Mar, Barcelona 08003, Spain
| | - Elena Mercade
- Department of Biology, Health and Environment, University of Barcelona, Barcelona 08028, Spain
| | - Saurabh Mehandru
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrea Cerutti
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona 08003, Spain; Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Catalan Institute for Research and Advanced Studies (ICREA), Barcelona 08003, Spain.
| |
Collapse
|
109
|
Nair N, Feng N, Blum LK, Sanyal M, Ding S, Jiang B, Sen A, Morton JM, He XS, Robinson WH, Greenberg HB. VP4- and VP7-specific antibodies mediate heterotypic immunity to rotavirus in humans. Sci Transl Med 2017; 9:eaam5434. [PMID: 28637924 PMCID: PMC6312383 DOI: 10.1126/scitranslmed.aam5434] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 05/14/2017] [Indexed: 12/18/2022]
Abstract
Human rotaviruses (RVs) are the leading cause of severe diarrhea in young children worldwide. The molecular mechanisms underlying the rapid induction of heterotypic protective immunity to RV, which provides the basis for the efficacy of licensed monovalent RV vaccines, have remained unknown for more than 30 years. We used RV-specific single cell-sorted intestinal B cells from human adults, barcode-based deep sequencing of antibody repertoires, monoclonal antibody expression, and serologic and functional characterization to demonstrate that infection-induced heterotypic immunoglobulins (Igs) primarily directed to VP5*, the stalk region of the RV attachment protein, VP4, are able to mediate heterotypic protective immunity. Heterotypic protective Igs against VP7, the capsid glycoprotein, and VP8*, the cell-binding region of VP4, are also generated after infection; however, our data suggest that homotypic anti-VP7 and non-neutralizing VP8* responses occur more commonly in people. These results indicate that humans can circumvent the extensive serotypic diversity of circulating RV strains by generating frequent heterotypic neutralizing antibody responses to VP7, VP8*, and most often, to VP5* after natural infection. These findings further suggest that recombinant VP5* may represent a useful target for the development of an improved, third-generation, broadly effective RV vaccine and warrants more direct examination.
Collapse
Affiliation(s)
- Nitya Nair
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ningguo Feng
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Lisa K Blum
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mrinmoy Sanyal
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Siyuan Ding
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Baoming Jiang
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Adrish Sen
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - John M Morton
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xiao-Song He
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - William H Robinson
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Harry B Greenberg
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| |
Collapse
|
110
|
Roy B, Neumann RS, Snir O, Iversen R, Sandve GK, Lundin KEA, Sollid LM. High-Throughput Single-Cell Analysis of B Cell Receptor Usage among Autoantigen-Specific Plasma Cells in Celiac Disease. THE JOURNAL OF IMMUNOLOGY 2017; 199:782-791. [PMID: 28600290 DOI: 10.4049/jimmunol.1700169] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/11/2017] [Indexed: 11/19/2022]
Abstract
Characterization of Ag-specific BCR repertoires is essential for understanding disease mechanisms involving humoral immunity. This is optimally done by interrogation of paired H chain V region (VH) and L chain V region (VL) sequences of individual and Ag-specific B cells. By applying single-cell high-throughput sequencing on gut lesion plasma cells (PCs), we have analyzed the transglutaminase 2 (TG2)-specific VH:VL autoantibody repertoire of celiac disease (CD) patients. Autoantibodies against TG2 are a hallmark of CD, and anti-TG2 IgA-producing gut PCs accumulate in patients upon gluten ingestion. Altogether, we analyzed paired VH and VL sequences of 1482 TG2-specific and 1421 non-TG2-specific gut PCs from 10 CD patients. Among TG2-specific PCs, we observed a striking bias in IGHV and IGKV/IGLV gene usage, as well as pairing preferences with a particular presence of the IGHV5-51:IGKV1-5 pair. Selective and biased VH:VL pairing was particularly evident among expanded clones. In general, TG2-specific PCs had lower numbers of mutations both in VH and VL genes than in non-TG2-specific PCs. TG2-specific PCs using IGHV5-51 had particularly few mutations. Importantly, VL segments paired with IGHV5-51 displayed proportionally low mutation numbers, suggesting that the low mutation rate among IGHV5-51 PCs is dictated by the BCR specificity. Finally, we observed selective amino acid changes in VH and VL and striking CDR3 length and J segment selection among TG2-specific IGHV5-51:IGKV1-5 pairs. Hence this study reveals features of a disease- and Ag-specific autoantibody repertoire with preferred VH:VL usage and pairings, limited mutations, clonal dominance, and selection of particular CDR3 sequences.
Collapse
Affiliation(s)
- Bishnudeo Roy
- Department of Immunology, Centre for Immune Regulation, University of Oslo, Oslo University Hospital, 0372 Oslo, Norway
| | - Ralf S Neumann
- Department of Immunology, KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo University Hospital, 0372 Oslo, Norway
| | - Omri Snir
- Department of Immunology, Centre for Immune Regulation, University of Oslo, Oslo University Hospital, 0372 Oslo, Norway
| | - Rasmus Iversen
- Department of Immunology, Centre for Immune Regulation, University of Oslo, Oslo University Hospital, 0372 Oslo, Norway
| | - Geir Kjetil Sandve
- Department of Informatics, KG Jebsen Coeliac Disease Research Centre, University of Oslo, 0373 Oslo, Norway; and
| | - Knut E A Lundin
- Department of Gastroenterology, KG Jebsen Coeliac Disease Research Centre, Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway
| | - Ludvig M Sollid
- Department of Immunology, Centre for Immune Regulation, University of Oslo, Oslo University Hospital, 0372 Oslo, Norway; .,Department of Immunology, KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo University Hospital, 0372 Oslo, Norway
| |
Collapse
|
111
|
Agace WW, McCoy KD. Regionalized Development and Maintenance of the Intestinal Adaptive Immune Landscape. Immunity 2017; 46:532-548. [PMID: 28423335 DOI: 10.1016/j.immuni.2017.04.004] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 12/14/2022]
Abstract
The intestinal immune system has the daunting task of protecting us from pathogenic insults while limiting inflammatory responses against the resident commensal microbiota and providing tolerance to food antigens. This role is particularly impressive when one considers the vast mucosal surface and changing landscape that the intestinal immune system must monitor. In this review, we highlight regional differences in the development and composition of the adaptive immune landscape of the intestine and the impact of local intrinsic and environmental factors that shape this process. To conclude, we review the evidence for a critical window of opportunity for early-life exposures that affect immune development and alter disease susceptibility later in life.
Collapse
Affiliation(s)
- William W Agace
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark (DTU), 2800 Kongens Lyngby, Denmark; Immunology Section, Department of Experimental Medical Science, Lund University, BMC D14, Sölvegatan 19, 221 84 Lund, Sweden.
| | - Kathy D McCoy
- Department of Physiology and Pharmacology and Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
112
|
Schickel JN, Glauzy S, Ng YS, Chamberlain N, Massad C, Isnardi I, Katz N, Uzel G, Holland SM, Picard C, Puel A, Casanova JL, Meffre E. Self-reactive VH4-34-expressing IgG B cells recognize commensal bacteria. J Exp Med 2017; 214:1991-2003. [PMID: 28500047 PMCID: PMC5502416 DOI: 10.1084/jem.20160201] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 11/08/2016] [Accepted: 04/10/2017] [Indexed: 12/14/2022] Open
Abstract
The human VH4-34 gene segment encodes intrinsically self-reactive antibodies that recognize I/i carbohydrates. Schickel et al. show that these self-reactive clones may represent an innate-like B cell population specialized in the containment of commensal bacteria when gut barriers are breached. The germline immunoglobulin (Ig) variable heavy chain 4–34 (VH4-34) gene segment encodes in humans intrinsically self-reactive antibodies that recognize I/i carbohydrates expressed by erythrocytes with a specific motif in their framework region 1 (FWR1). VH4-34–expressing clones are common in the naive B cell repertoire but are rarely found in IgG memory B cells from healthy individuals. In contrast, CD27+IgG+ B cells from patients genetically deficient for IRAK4 or MYD88, which mediate the function of Toll-like receptors (TLRs) except TLR3, contained VH4-34–expressing clones and showed decreased somatic hypermutation frequencies. In addition, VH4-34–encoded IgGs from IRAK4- and MYD88-deficient patients often displayed an unmutated FWR1 motif, revealing that these antibodies still recognize I/i antigens, whereas their healthy donor counterparts harbored FWR1 mutations abolishing self-reactivity. However, this paradoxical self-reactivity correlated with these VH4-34–encoded IgG clones binding commensal bacteria antigens. Hence, B cells expressing germline-encoded self-reactive VH4-34 antibodies may represent an innate-like B cell population specialized in the containment of commensal bacteria when gut barriers are breached.
Collapse
Affiliation(s)
- Jean-Nicolas Schickel
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| | - Salomé Glauzy
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| | - Yen-Shing Ng
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| | - Nicolas Chamberlain
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| | - Christopher Massad
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| | - Isabelle Isnardi
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| | - Nathan Katz
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| | - Gulbu Uzel
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Steven M Holland
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892.,Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Capucine Picard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Necker Hospital for Sick Children, 75015 Paris, France.,Paris Descartes University, Imagine Institute, 75015 Paris, France
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Necker Hospital for Sick Children, 75015 Paris, France.,Paris Descartes University, Imagine Institute, 75015 Paris, France
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Necker Hospital for Sick Children, 75015 Paris, France.,Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, 75015 Paris, France.,Paris Descartes University, Imagine Institute, 75015 Paris, France.,St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065.,Howard Hughes Medical Institute, New York, NY 10065
| | - Eric Meffre
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| |
Collapse
|
113
|
Zhang W, Feng Q, Wang C, Zeng X, Du Y, Lin L, Wu J, Fu L, Yang K, Xu X, Xu H, Zhao Y, Li X, Schoenauer UH, Stadlmayr A, Saksena NK, Tilg H, Datz C, Liu X. Characterization of the B Cell Receptor Repertoire in the Intestinal Mucosa and of Tumor-Infiltrating Lymphocytes in Colorectal Adenoma and Carcinoma. THE JOURNAL OF IMMUNOLOGY 2017; 198:3719-3728. [DOI: 10.4049/jimmunol.1602039] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/23/2017] [Indexed: 01/10/2023]
|
114
|
The Impact of the Gut Microbiota on Humoral Immunity to Pathogens and Vaccination in Early Infancy. PLoS Pathog 2016; 12:e1005997. [PMID: 28006021 PMCID: PMC5179050 DOI: 10.1371/journal.ppat.1005997] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
115
|
Bone Marrow Mesenchymal Stem Cells Enhance the Differentiation of Human Switched Memory B Lymphocytes into Plasma Cells in Serum-Free Medium. J Immunol Res 2016; 2016:7801781. [PMID: 27872867 PMCID: PMC5107863 DOI: 10.1155/2016/7801781] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/31/2016] [Accepted: 09/04/2016] [Indexed: 12/21/2022] Open
Abstract
The differentiation of human B lymphocytes into plasma cells is one of the most stirring questions with regard to adaptive immunity. However, the terminal differentiation and survival of plasma cells are still topics with much to be discovered, especially when targeting switched memory B lymphocytes. Plasma cells can migrate to the bone marrow in response to a CXCL12 gradient and survive for several years while secreting antibodies. In this study, we aimed to get closer to niches favoring plasma cell survival. We tested low oxygen concentrations and coculture with mesenchymal stem cells (MSC) from human bone marrow. Besides, all cultures were performed using an animal protein-free medium. Overall, our model enables the generation of high proportions of CD38+CD138+CD31+ plasma cells (≥50%) when CD40-activated switched memory B lymphocytes were cultured in direct contact with mesenchymal stem cells. In these cultures, the secretion of CXCL12 and TGF-β, usually found in the bone marrow, was linked to the presence of MSC. The level of oxygen appeared less impactful than the contact with MSC. This study shows for the first time that expanded switched memory B lymphocytes can be differentiated into plasma cells using exclusively a serum-free medium.
Collapse
|
116
|
Späth PJ, Schneider C, von Gunten S. Clinical Use and Therapeutic Potential of IVIG/SCIG, Plasma-Derived IgA or IgM, and Other Alternative Immunoglobulin Preparations. Arch Immunol Ther Exp (Warsz) 2016; 65:215-231. [DOI: 10.1007/s00005-016-0422-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 08/31/2016] [Indexed: 12/22/2022]
|
117
|
Spencer J, Sollid LM. The human intestinal B-cell response. Mucosal Immunol 2016; 9:1113-24. [PMID: 27461177 DOI: 10.1038/mi.2016.59] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/10/2016] [Indexed: 02/04/2023]
Abstract
The intestinal immune system is chronically challenged by a huge plethora of antigens derived from the lumen. B-cell responses in organized gut-associated lymphoid tissues and regional lymph nodes that are driven chronically by gut antigens generate the largest population of antibody-producing cells in the body: the gut lamina propria plasma cells. Although animal studies have provided insights into mechanisms that underpin this dynamic process, some very fundamental differences in this system appear to exist between species. Importantly, this prevents extrapolation from mice to humans to inform translational research questions. Therefore, in this review we will describe the structures and mechanisms involved in the propagation, dissemination, and regulation of this immense plasma cell population in man. Uniquely, we will seek our evidence exclusively from studies of human cells and tissues.
Collapse
Affiliation(s)
- J Spencer
- Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - L M Sollid
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway
| |
Collapse
|
118
|
Ulsemer P, Toutounian K, Kressel G, Goletz C, Schmidt J, Karsten U, Hahn A, Goletz S. Impact of oral consumption of heat-treated Bacteroides xylanisolvens DSM 23964 on the level of natural TFα-specific antibodies in human adults. Benef Microbes 2016; 7:485-500. [DOI: 10.3920/bm2015.0143] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
It is now generally accepted that the human body exists in close synergy with the gut microbiome and that this cross-talk plays an essential role in human health and disease. One facet from the many interactions between the microbiome and the immune system is the induction of natural antibodies to commensal bacterial glycans, such as blood group antigens, the alpha-Gal epitope or the Thomsen-Friedenreich (TFα) antigen. Since we have observed that certain species of the commensal genus Bacteroides express the TFα antigen, we examined whether the oral dietary supplementation of a pasteurised Bacteroides xylanisolvens strain might be able to enhance the level of natural anti-TFα antibodies in healthy adults. The data obtained from a double-blind, placebo-controlled study involving 140 healthy volunteers and lasting 8 weeks revealed that the oral uptake of this strain was indeed able to increase the level of TFα-specific immunoglobulin M serum antibodies. The effect was dose-dependent but remained – at any doses – within the physiological range determined before intervention. Furthermore, the effect reverted after stopping the intake. The results support the idea of the microbiome inducing the generation of systemic antigen-specific antibodies against sugar epitopes. They also demonstrate the possibility to modulate essential regulatory or defence processes through dietary supplementation of selected commensal bacteria with the aim to assist human health.
Collapse
Affiliation(s)
- P. Ulsemer
- Avitop GmbH, Robert-Roessler-Str. 10, 13125 Berlin, Germany
| | - K. Toutounian
- Avitop GmbH, Robert-Roessler-Str. 10, 13125 Berlin, Germany
| | - G. Kressel
- Leibniz University of Hannover, Institute of Food Science and Human Nutrition, Am Kleinen Felde 30, 30163 Hannover, Germany
| | - C. Goletz
- Glycotope GmbH, Robert-Roessle-Str. 10, 13125 Berlin, Germany
| | - J. Schmidt
- Avitop GmbH, Robert-Roessler-Str. 10, 13125 Berlin, Germany
| | - U. Karsten
- Glycotope GmbH, Robert-Roessle-Str. 10, 13125 Berlin, Germany
| | - A. Hahn
- Leibniz University of Hannover, Institute of Food Science and Human Nutrition, Am Kleinen Felde 30, 30163 Hannover, Germany
| | - S. Goletz
- Glycotope GmbH, Robert-Roessle-Str. 10, 13125 Berlin, Germany
| |
Collapse
|
119
|
Abstract
Hypomorphic Rag mutations in humans cause Omenn Syndrome (OS) a severe immunodeficiency associated with autoimmune-like manifestations mediated by oligoclonal activated T and B cells. The clinical and immunological spectrum of OS presentation is extremely broad. However, the role played by environmental triggers in the disease pathogenesis remains largely unknown. We have recently shown in a murine model that gut microbiota has a substantial role in determining the distinctive immune dysregulation of OS. Here, we describe how dysbiosis and loss of T cell tolerance to commensals influence the expression of autoimmunity at the barrier site and beyond, and the disease hallmark hyper-IgE. We discuss how commensal antigens and gut-derived pathogenic T cells could potentially modulate skin immunity to determine cutaneous degenerations in OS. These mechanisms may have broader implications for a deeper understanding of the role of gut microbes in influencing barriers integrity and host physiology.
Collapse
Affiliation(s)
- Rosita Rigoni
- Milan Unit, Istituto di Ricerca Genetica e
Biomedica, Consiglio Nazionale delle Ricerche, Milan,
Italy,Humanitas Clinical and Research
Center, Rozzano, Milan, Italy
| | - Fabio Grassi
- Istituto Nazionale Genetica Molecolare,
Department of Medical Biotechnology and Translational Medicine, University of
Milan, Milan, Italy,Institute for Research in
Biomedicine, Bellinzona, Switzerland
| | - Anna Villa
- Milan Unit, Istituto di Ricerca Genetica e
Biomedica, Consiglio Nazionale delle Ricerche, Milan,
Italy,Telethon Institute for Gene Therapy, Division
of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a
Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan,
Italy
| | - Barbara Cassani
- Milan Unit, Istituto di Ricerca Genetica e
Biomedica, Consiglio Nazionale delle Ricerche, Milan,
Italy,Humanitas Clinical and Research
Center, Rozzano, Milan, Italy,CONTACT Barbara Cassani Humanitas Clinical and Research Center, via Manzoni
56, 20089 Rozzano (Mi), Italy
| |
Collapse
|
120
|
Prigent J, Lorin V, Kök A, Hieu T, Bourgeau S, Mouquet H. Scarcity of autoreactive human blood IgA + memory B cells. Eur J Immunol 2016; 46:2340-2351. [PMID: 27469325 PMCID: PMC5113776 DOI: 10.1002/eji.201646446] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/17/2016] [Accepted: 07/26/2016] [Indexed: 12/17/2022]
Abstract
Class‐switched memory B cells are key components of the “reactive” humoral immunity, which ensures a fast and massive secretion of high‐affinity antigen‐specific antibodies upon antigenic challenge. In humans, IgA class‐switched (IgA+) memory B cells and IgA antibodies are abundant in the blood. Although circulating IgA+ memory B cells and their corresponding secreted immunoglobulins likely possess major protective and/or regulatory immune roles, little is known about their specificity and function. Here, we show that IgA+ and IgG+ memory B‐cell antibodies cloned from the same healthy humans share common immunoglobulin gene features. IgA and IgG memory antibodies have comparable lack of reactivity to vaccines, common mucosa‐tropic viruses and commensal bacteria. However, the IgA+ memory B‐cell compartment contains fewer polyreactive clones and importantly, only rare self‐reactive clones compared to IgG+ memory B cells. Self‐reactivity of IgAs is acquired following B‐cell affinity maturation but not antibody class switching. Together, our data suggest the existence of different regulatory mechanisms for removing autoreactive clones from the IgG+ and IgA+ memory B‐cell repertoires, and/or different maturation pathways potentially reflecting the distinct nature and localization of the cognate antigens recognized by individual B‐cell populations.
Collapse
Affiliation(s)
- Julie Prigent
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris, France.,CNRS-URA 1961, Paris, France
| | - Valérie Lorin
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris, France.,CNRS-URA 1961, Paris, France
| | - Ayrin Kök
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris, France.,CNRS-URA 1961, Paris, France
| | - Thierry Hieu
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris, France.,CNRS-URA 1961, Paris, France
| | - Salomé Bourgeau
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris, France.,CNRS-URA 1961, Paris, France
| | - Hugo Mouquet
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris, France. .,CNRS-URA 1961, Paris, France.
| |
Collapse
|
121
|
Jones L, Ho WQ, Ying S, Ramakrishna L, Srinivasan KG, Yurieva M, Ng WP, Subramaniam S, Hamadee NH, Joseph S, Dolpady J, Atarashi K, Honda K, Zolezzi F, Poidinger M, Lafaille JJ, Curotto de Lafaille MA. A subpopulation of high IL-21-producing CD4(+) T cells in Peyer's Patches is induced by the microbiota and regulates germinal centers. Sci Rep 2016; 6:30784. [PMID: 27499025 PMCID: PMC4976330 DOI: 10.1038/srep30784] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 07/11/2016] [Indexed: 01/02/2023] Open
Abstract
The production of IL-21 by T follicular helper (Tfh) cells is vital in driving the germinal centre reaction and high affinity antibody formation. However, the degree of Tfh cell heterogeneity and function is not fully understood. We used a novel IL-21eGFP reporter mouse strain to analyze the diversity and role of Tfh cells. Through the analysis of GFP expression in lymphoid organs of IL-21eGFP mice, we identified a subpopulation of GFP+, high IL-21 producing Tfh cells present only in Peyer’s Patches. GFP+Tfh cells were found to be polyclonal and related to GFP−Tfh cells of Peyer’s Patches in TCR repertoire composition and overall gene expression. Studies on the mechanisms of induction of GFP+Tfh cells demonstrated that they required the intestinal microbiota and a diverse repertoire of CD4+ T cells and B cells. Importantly, ablation of GFP+ cells resulted in a reduced frequency of Peyer’s Patches IgG1 and germinal center B cells in addition to small but significant shifts in gut microbiome composition. Our work highlights the diversity among IL-21 producing CD4+ Tfh cells, and the interrelationship between the intestinal bacteria and Tfh cell responses in the gut.
Collapse
Affiliation(s)
- Leigh Jones
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Wen Qi Ho
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sze Ying
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore.,Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lakshmi Ramakrishna
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kandhadayar G Srinivasan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Marina Yurieva
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Wan Pei Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore
| | - Sharrada Subramaniam
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Nur H Hamadee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sabrina Joseph
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jayashree Dolpady
- Skirball Institute and Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Koji Atarashi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, and RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Kenya Honda
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, and RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Francesca Zolezzi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Juan J Lafaille
- Skirball Institute and Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Maria A Curotto de Lafaille
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine and Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
122
|
Salerno-Goncalves R, Safavie F, Fasano A, Sztein MB. Free and complexed-secretory immunoglobulin A triggers distinct intestinal epithelial cell responses. Clin Exp Immunol 2016; 185:338-47. [PMID: 27084834 PMCID: PMC4991520 DOI: 10.1111/cei.12801] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/31/2016] [Accepted: 04/13/2016] [Indexed: 12/12/2022] Open
Abstract
Secretory immunoglobulin A (SIgA) antibodies play an important role in protecting the mucosal surfaces against pathogens and maintaining homeostasis with the commensal microbiota. Because a substantial portion of the gut microbiota is coated with SIgA, we hypothesized that microbiota–SIgA complexes are important for the maintenance of gut homeostasis. Here we investigated the relationship between microbiota–SIgA complexes and inflammatory epithelial cell responses. We used a multi‐cellular three‐dimensional (3D) organotypical model of the human intestinal mucosa composed of an intestinal epithelial cell line and primary human lymphocytes/monocytes, endothelial cells and fibroblasts. We also used human SIgA from human colostrum, and a prominent bacterial member of the first colonizers, Escherichia coli, as a surrogate commensal. We found that free and microbiota‐complexed SIgA triggered different epithelial responses. While free SIgA up‐regulated mucus production, expression of polymeric immunoglobulin receptor (pIgR) and secretion of interleukin‐8 and tumoir necrosis factor‐α, microbiota‐complexed SIgA mitigated these responses. These results suggest that free and complexed SIgA have different functions as immunoregulatory agents in the gut and that an imbalance between the two may affect gut homeostasis.
Collapse
Affiliation(s)
- R Salerno-Goncalves
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| | - F Safavie
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| | - A Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | - M B Sztein
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
123
|
Rios D, Wood MB, Li J, Chassaing B, Gewirtz AT, Williams IR. Antigen sampling by intestinal M cells is the principal pathway initiating mucosal IgA production to commensal enteric bacteria. Mucosal Immunol 2016; 9:907-16. [PMID: 26601902 PMCID: PMC4917673 DOI: 10.1038/mi.2015.121] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 10/08/2015] [Indexed: 02/04/2023]
Abstract
Secretory IgA (SIgA) directed against gut resident bacteria enables the mammalian mucosal immune system to establish homeostasis with the commensal gut microbiota after weaning. Germinal centers (GCs) in Peyer's patches (PPs) are the principal inductive sites where naive B cells specific for bacterial antigens encounter their cognate antigens and receive T-cell help driving their differentiation into IgA-producing plasma cells. We investigated the role of antigen sampling by intestinal M cells in initiating the SIgA response to gut bacteria by developing mice in which receptor activator of nuclear factor-κB ligand (RANKL)-dependent M-cell differentiation was abrogated by conditional deletion of Tnfrsf11a in the intestinal epithelium. Mice without intestinal M cells had profound delays in PP GC maturation and emergence of lamina propria IgA plasma cells, resulting in diminished levels of fecal SIgA that persisted into adulthood. We conclude that M-cell-mediated sampling of commensal bacteria is a required initial step for the efficient induction of intestinal SIgA.
Collapse
Affiliation(s)
- D Rios
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - M B Wood
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - J Li
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - B Chassaing
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - A T Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - I R Williams
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
124
|
Bessman NJ, Sonnenberg GF. Emerging roles for antigen presentation in establishing host-microbiome symbiosis. Immunol Rev 2016; 272:139-50. [PMID: 27319348 PMCID: PMC4916850 DOI: 10.1111/imr.12425] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Trillions of beneficial bacteria inhabit the intestinal tract of healthy mammals from birth. Accordingly, mammalian hosts have evolved a series of complementary and redundant pathways to limit pathologic immune responses against these bacteria, while simultaneously protecting against enteric pathogen invasion. These pathways can be generically responsive to the presence of any commensal bacteria and innate in nature, as for IL-22-related pathways. Alternatively, specific bacterial antigens can drive a distinct set of adaptive immune cell responses, including IgA affinity maturation and secretion, and a recently described pathway of intestinal selection whereby MHCII(+) ILC3 deletes commensal bacteria-reactive CD4 T cells. These pathways can either promote or inhibit colonization by specific subsets of commensal bacteria, and cooperatively maintain intestinal homeostasis. In this review, we will highlight recent developments in understanding how these diverse pathways complement each other to cooperatively shape the symbiotic relationship between commensal bacteria and mammalian hosts.
Collapse
Affiliation(s)
- Nicholas J Bessman
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
| | - Gregory F Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
125
|
Soto R, Round J. Immunology: You Remind Me of a Microbe I Know. Curr Biol 2016; 26:R373-6. [PMID: 27166699 DOI: 10.1016/j.cub.2016.03.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The majority of bacteria found within the gut are commensals, although it is unclear whether these organisms can elicit systemic immunity. New research indicates that gut-microbiota-specific serum antibodies targeting an epitope conserved among Gram-negative bacteria can protect the host from systemic pathogenic infection.
Collapse
Affiliation(s)
- Ray Soto
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - June Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
126
|
Abstract
Secondary lymphoid tissues share the important function of bringing together antigens and rare antigen-specific lymphocytes to foster induction of adaptive immune responses. Peyer's patches (PPs) are unique compared to other secondary lymphoid tissues in their continual exposure to an enormous diversity of microbiome- and food-derived antigens and in the types of pathogens they encounter. Antigens are delivered to PPs by specialized microfold (M) epithelial cells and they may be captured and presented by resident dendritic cells (DCs). In accord with their state of chronic microbial antigen exposure, PPs exhibit continual germinal center (GC) activity. These GCs not only contribute to the generation of B cells and plasma cells producing somatically mutated gut antigen-specific IgA antibodies but have also been suggested to support non-specific antigen diversification of the B-cell repertoire. Here, we review current understanding of how PPs foster B-cell encounters with antigen, how they favor isotype switching to the secretory IgA isotype, and how their GC responses may uniquely contribute to mucosal immunity.
Collapse
Affiliation(s)
- Andrea Reboldi
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
127
|
van der Meulen TA, Harmsen HJM, Bootsma H, Spijkervet FKL, Kroese FGM, Vissink A. The microbiome-systemic diseases connection. Oral Dis 2016; 22:719-734. [DOI: 10.1111/odi.12472] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/01/2016] [Accepted: 03/02/2016] [Indexed: 12/28/2022]
Affiliation(s)
- TA van der Meulen
- Department of Oral and Maxillofacial Surgery; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - HJM Harmsen
- Department of Medical Microbiology; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - H Bootsma
- Department of Rheumatology and Clinical Immunology; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - FKL Spijkervet
- Department of Oral and Maxillofacial Surgery; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - FGM Kroese
- Department of Rheumatology and Clinical Immunology; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - A Vissink
- Department of Oral and Maxillofacial Surgery; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| |
Collapse
|
128
|
Meng W, Li L, Xiong W, Fan X, Deng H, Bett AJ, Chen Z, Tang A, Cox KS, Joyce JG, Freed DC, Thoryk E, Fu TM, Casimiro DR, Zhang N, A Vora K, An Z. Efficient generation of monoclonal antibodies from single rhesus macaque antibody secreting cells. MAbs 2016; 7:707-18. [PMID: 25996084 PMCID: PMC4622687 DOI: 10.1080/19420862.2015.1051440] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Nonhuman primates (NHPs) are used as a preclinical model for vaccine development, and the antibody profiles to experimental vaccines in NHPs can provide critical information for both vaccine design and translation to clinical efficacy. However, an efficient protocol for generating monoclonal antibodies from single antibody secreting cells of NHPs is currently lacking. In this study we established a robust protocol for cloning immunoglobulin (IG) variable domain genes from single rhesus macaque (Macaca mulatta) antibody secreting cells. A sorting strategy was developed using a panel of molecular markers (CD3, CD19, CD20, surface IgG, intracellular IgG, CD27, Ki67 and CD38) to identify the kinetics of B cell response after vaccination. Specific primers for the rhesus macaque IG genes were designed and validated using cDNA isolated from macaque peripheral blood mononuclear cells. Cloning efficiency was averaged at 90% for variable heavy (VH) and light (VL) domains, and 78.5% of the clones (n = 335) were matched VH and VL pairs. Sequence analysis revealed that diverse IGHV subgroups (for VH) and IGKV and IGLV subgroups (for VL) were represented in the cloned antibodies. The protocol was tested in a study using an experimental dengue vaccine candidate. About 26.6% of the monoclonal antibodies cloned from the vaccinated rhesus macaques react with the dengue vaccine antigens. These results validate the protocol for cloning monoclonal antibodies in response to vaccination from single macaque antibody secreting cells, which have general applicability for determining monoclonal antibody profiles in response to other immunogens or vaccine studies of interest in NHPs.
Collapse
Affiliation(s)
- Weixu Meng
- a Texas Therapeutics Institute; Brown Foundation Institute of Molecular Medicine; University of Texas Health Science Center at Houston ; Houston , TX , USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Zeng MY, Cisalpino D, Varadarajan S, Hellman J, Warren HS, Cascalho M, Inohara N, Núñez G. Gut Microbiota-Induced Immunoglobulin G Controls Systemic Infection by Symbiotic Bacteria and Pathogens. Immunity 2016; 44:647-658. [PMID: 26944199 DOI: 10.1016/j.immuni.2016.02.006] [Citation(s) in RCA: 272] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 11/10/2015] [Accepted: 12/07/2015] [Indexed: 12/21/2022]
Abstract
The gut microbiota is compartmentalized in the intestinal lumen and induces local immune responses, but it remains unknown whether the gut microbiota can induce systemic response and contribute to systemic immunity. We report that selective gut symbiotic gram-negative bacteria were able to disseminate systemically to induce immunoglobulin G (IgG) response, which primarily targeted gram-negative bacterial antigens and conferred protection against systemic infections by E. coli and Salmonella by directly coating bacteria to promote killing by phagocytes. T cells and Toll-like receptor 4 on B cells were important in the generation of microbiota-specific IgG. We identified murein lipoprotein (MLP), a highly conserved gram-negative outer membrane protein, as a major antigen that induced systemic IgG homeostatically in both mice and humans. Administration of anti-MLP IgG conferred crucial protection against systemic Salmonella infection. Thus, our findings reveal an important function for the gut microbiota in combating systemic infection through the induction of protective IgG.
Collapse
Affiliation(s)
- Melody Y Zeng
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Daniel Cisalpino
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Saranyaraajan Varadarajan
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, Division of Critical Care Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - H Shaw Warren
- Infectious Disease Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Marilia Cascalho
- Transplantation Biology, Department of Surgery and Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Naohiro Inohara
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Gabriel Núñez
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
130
|
High-dimensional immune profiling of total and rotavirus VP6-specific intestinal and circulating B cells by mass cytometry. Mucosal Immunol 2016; 9:68-82. [PMID: 25899688 PMCID: PMC4618273 DOI: 10.1038/mi.2015.36] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 03/07/2015] [Indexed: 02/04/2023]
Abstract
In-depth phenotyping of human intestinal antibody secreting cells (ASCs) and their precursors is important for developing improved mucosal vaccines. We used single-cell mass cytometry to simultaneously analyze 34 differentiation and trafficking markers on intestinal and circulating B cells. In addition, we labeled rotavirus (RV) double-layered particles with a metal isotope and characterized B cells specific to the RV VP6 major structural protein. We describe the heterogeneity of the intestinal B-cell compartment, dominated by ASCs with some phenotypic and transcriptional characteristics of long-lived plasma cells. Using principal component analysis, we visualized the phenotypic relationships between major B-cell subsets in the intestine and blood, and revealed that IgM(+) memory B cells (MBCs) and naive B cells were phenotypically related as were CD27(-) MBCs and switched MBCs. ASCs in the intestine and blood were highly clonally related, but associated with distinct trajectories of phenotypic development. VP6-specific B cells were present among diverse B-cell subsets in immune donors, including naive B cells, with phenotypes representative of the overall B-cell pool. These data provide a high dimensional view of intestinal B cells and the determinants regulating humoral memory to a ubiquitous, mucosal pathogen at steady-state.
Collapse
|
131
|
Weise C, Worm M. MALT (»mucosa-associated lymphoid tissue«). ALLERGOLOGIE 2016. [DOI: 10.1007/978-3-642-37203-2_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
132
|
Di Niro R, Snir O, Kaukinen K, Yaari G, Lundin KEA, Gupta NT, Kleinstein SH, Cols M, Cerutti A, Mäki M, Shlomchik MJ, Sollid LM. Responsive population dynamics and wide seeding into the duodenal lamina propria of transglutaminase-2-specific plasma cells in celiac disease. Mucosal Immunol 2016; 9:254-64. [PMID: 26153762 PMCID: PMC4703456 DOI: 10.1038/mi.2015.57] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 05/12/2015] [Indexed: 02/04/2023]
Abstract
A hallmark of celiac disease is autoantibodies to transglutaminase 2 (TG2). By visualizing TG2-specific antibodies by antigen staining of affected gut tissue, we identified TG2-specific plasma cells in the lamina propria as well as antibodies in the subepithelial layer, inside the epithelium, and at the brush border. The frequency of TG2-specific plasma cells were found not to correlate with serum antibody titers, suggesting that antibody production at other sites may contribute to serum antibody levels. Upon commencement of a gluten-free diet, the frequency of TG2-specific plasma cells in the lesion dropped dramatically within 6 months, yet some cells remained. The frequency of TG2-specific plasma cells in the celiac lesion is thus dynamically regulated in response to gluten exposure. Laser microdissection of plasma cell patches, followed by antibody gene sequencing, demonstrated that clonal cells were seeded in distinct areas of the mucosa. This was confirmed by immunoglobulin heavy chain repertoire analysis of plasma cells isolated from individual biopsies of two untreated patients, both for TG2-specific and non-TG2-specific cells. Our results shed new light on the processes underlying the B-cell response in celiac disease, and the approach of staining for antigen-specific antibodies should be applicable to other antibody-mediated diseases.
Collapse
Affiliation(s)
- R Di Niro
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA,
| | - O Snir
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - K Kaukinen
- Departments of Internal Medicine and Gastroenterology, Tampere University Hospital and School of Medicine, University of Tampere, Tampere, Finland
| | - G Yaari
- Bioengineering Program, Faculty of Engineering, Bar-Ilan University, Ramat Gan, Israel
| | - K E A Lundin
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway,Centre for Immune Regulation and Department of Gastroenterology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - N T Gupta
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, USA
| | - S H Kleinstein
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, USA,Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - M Cols
- Immunology Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - A Cerutti
- Immunology Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - M Mäki
- Tampere Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - M J Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - L M Sollid
- Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway
| |
Collapse
|
133
|
Maurer MA, Tuller F, Gredler V, Berger T, Lutterotti A, Lünemann JD, Reindl M. Rituximab induces clonal expansion of IgG memory B-cells in patients with inflammatory central nervous system demyelination. J Neuroimmunol 2016; 290:49-53. [DOI: 10.1016/j.jneuroim.2015.11.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/09/2015] [Accepted: 11/12/2015] [Indexed: 11/24/2022]
|
134
|
Tsuda R, Ozawa T, Kobayashi E, Hamana H, Taki H, Tobe K, Sugiyama E, Iwamoto M, Imura J, Kishi H, Muraguchi A. Monoclonal antibody against citrullinated peptides obtained from rheumatoid arthritis patients reacts with numerous citrullinated microbial and food proteins. Arthritis Rheumatol 2015; 67:2020-31. [PMID: 25892475 DOI: 10.1002/art.39161] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 04/14/2015] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To investigate the reactivity of monoclonal anti-citrullinated protein antibody (ACPA) obtained from peripheral blood B cells of rheumatoid arthritis (RA) patients with human autoantigens as well as environmental proteins by determining the essential epitope for the ACPA. METHODS A human monoclonal ACPA (cyclic citrullinated peptide antibody 1 [CCP-Ab1]) was obtained by screening peripheral blood lymphocytes from 31 patients with RA using a novel monoclonal antibody-secreting cell (ASC) screening system, the immunospot-array assay on a chip. The essential epitope for CCP-Ab1 was determined using epitope mapping. Then, human, microbial, and plant proteins that share the essential epitope identified were searched using BLAST. Finally, representative proteins identified by the search were produced in vitro, and their reactivity with CCP-Ab1 was examined. RESULTS CCP-Ab1 bound CCP in a citrulline-indispensable manner. In CCP, the 6 amino acid residues required for CCP-Ab1 binding were identified. In the BLAST search, 38 human, 56 viral, 1,383 fungal, 547 bacterial, and 1,072 plant proteins were found to share the essential epitope, and CCP-Ab1 reacted with all of the recombinant citrullinated proteins tested, which included the various environmental factors, such as various plant proteins that are part of the daily diet. CONCLUSION Our findings demonstrate, for the first time, that a monoclonal ACPA (CCP-Ab1) derived from RA patients cross-reacts not only with various autoantigens but also with numerous plant and microbial proteins. We propose that countless environmental factors, including microbes and diet, may trigger the generation of ACPAs that then cross-react with various citrullinated human autoantigens through molecular mimicry to induce RA.
Collapse
Affiliation(s)
- Reina Tsuda
- Dana-Farber Cancer Institute, Boston, Massachusetts), Hiroshi Hamana, PhD, Hirofumi Taki, MD, PhD, Kazuyuki Tobe, MD, PhD, Eiji Sugiyama, MD, PhD (current address: Hiroshima University Hospital, Hiroshima, Japan), Johji Imura, MD, PhD, Hiroyuki Kishi, PhD, Atsushi Muraguchi, MD, PhD: University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Sugitani Campus, Toyama, Japan
| | - Tatsuhiko Ozawa
- Dana-Farber Cancer Institute, Boston, Massachusetts), Hiroshi Hamana, PhD, Hirofumi Taki, MD, PhD, Kazuyuki Tobe, MD, PhD, Eiji Sugiyama, MD, PhD (current address: Hiroshima University Hospital, Hiroshima, Japan), Johji Imura, MD, PhD, Hiroyuki Kishi, PhD, Atsushi Muraguchi, MD, PhD: University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Sugitani Campus, Toyama, Japan
| | - Eiji Kobayashi
- Dana-Farber Cancer Institute, Boston, Massachusetts), Hiroshi Hamana, PhD, Hirofumi Taki, MD, PhD, Kazuyuki Tobe, MD, PhD, Eiji Sugiyama, MD, PhD (current address: Hiroshima University Hospital, Hiroshima, Japan), Johji Imura, MD, PhD, Hiroyuki Kishi, PhD, Atsushi Muraguchi, MD, PhD: University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Sugitani Campus, Toyama, Japan
| | - Hiroshi Hamana
- Dana-Farber Cancer Institute, Boston, Massachusetts), Hiroshi Hamana, PhD, Hirofumi Taki, MD, PhD, Kazuyuki Tobe, MD, PhD, Eiji Sugiyama, MD, PhD (current address: Hiroshima University Hospital, Hiroshima, Japan), Johji Imura, MD, PhD, Hiroyuki Kishi, PhD, Atsushi Muraguchi, MD, PhD: University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Sugitani Campus, Toyama, Japan
| | - Hirofumi Taki
- Dana-Farber Cancer Institute, Boston, Massachusetts), Hiroshi Hamana, PhD, Hirofumi Taki, MD, PhD, Kazuyuki Tobe, MD, PhD, Eiji Sugiyama, MD, PhD (current address: Hiroshima University Hospital, Hiroshima, Japan), Johji Imura, MD, PhD, Hiroyuki Kishi, PhD, Atsushi Muraguchi, MD, PhD: University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Sugitani Campus, Toyama, Japan
| | - Kazuyuki Tobe
- Dana-Farber Cancer Institute, Boston, Massachusetts), Hiroshi Hamana, PhD, Hirofumi Taki, MD, PhD, Kazuyuki Tobe, MD, PhD, Eiji Sugiyama, MD, PhD (current address: Hiroshima University Hospital, Hiroshima, Japan), Johji Imura, MD, PhD, Hiroyuki Kishi, PhD, Atsushi Muraguchi, MD, PhD: University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Sugitani Campus, Toyama, Japan
| | - Eiji Sugiyama
- Dana-Farber Cancer Institute, Boston, Massachusetts), Hiroshi Hamana, PhD, Hirofumi Taki, MD, PhD, Kazuyuki Tobe, MD, PhD, Eiji Sugiyama, MD, PhD (current address: Hiroshima University Hospital, Hiroshima, Japan), Johji Imura, MD, PhD, Hiroyuki Kishi, PhD, Atsushi Muraguchi, MD, PhD: University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Sugitani Campus, Toyama, Japan
| | | | - Johji Imura
- Dana-Farber Cancer Institute, Boston, Massachusetts), Hiroshi Hamana, PhD, Hirofumi Taki, MD, PhD, Kazuyuki Tobe, MD, PhD, Eiji Sugiyama, MD, PhD (current address: Hiroshima University Hospital, Hiroshima, Japan), Johji Imura, MD, PhD, Hiroyuki Kishi, PhD, Atsushi Muraguchi, MD, PhD: University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Sugitani Campus, Toyama, Japan
| | - Hiroyuki Kishi
- Dana-Farber Cancer Institute, Boston, Massachusetts), Hiroshi Hamana, PhD, Hirofumi Taki, MD, PhD, Kazuyuki Tobe, MD, PhD, Eiji Sugiyama, MD, PhD (current address: Hiroshima University Hospital, Hiroshima, Japan), Johji Imura, MD, PhD, Hiroyuki Kishi, PhD, Atsushi Muraguchi, MD, PhD: University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Sugitani Campus, Toyama, Japan
| | - Atsushi Muraguchi
- Dana-Farber Cancer Institute, Boston, Massachusetts), Hiroshi Hamana, PhD, Hirofumi Taki, MD, PhD, Kazuyuki Tobe, MD, PhD, Eiji Sugiyama, MD, PhD (current address: Hiroshima University Hospital, Hiroshima, Japan), Johji Imura, MD, PhD, Hiroyuki Kishi, PhD, Atsushi Muraguchi, MD, PhD: University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Sugitani Campus, Toyama, Japan
| |
Collapse
|
135
|
Bosqui LR, Gonçalves ALR, Gonçalves-Pires MDRF, Custodio LA, de Menezes MCND, Murad VA, de Paula FM, Pavanelli WR, Conchon-Costa I, Costa-Cruz JM, Costa IN. Detection of parasite-specific IgG and IgA in paired serum and saliva samples for diagnosis of human strongyloidiasis in northern Paraná state, Brazil. Acta Trop 2015; 150:190-5. [PMID: 26238982 DOI: 10.1016/j.actatropica.2015.07.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 01/04/2023]
Abstract
Human strongyloidiasis is an infection caused by the helminth Strongyloides stercoralis that can be fatal, especially in immunosuppressed patients. The aim of this study is to evaluate parasite-specific IgG and IgA levels using S. venezuelensis third-stage (L3) infective larvae alkaline extract as a heterologous antigen by ELISA in paired serum and saliva samples with improved sensitivity and specificity. Individuals from northern Paraná state, Brazil were divided into three groups: 30 patients copropositive for S. stercoralis (Group I); 30 clinically healthy individuals (Group II); and 30 patients copropositive for other parasites (Group III). The area under ROC curve (AUC), an overall index of diagnostic accuracy, and Kappa index were calculated. Data were analyzed using analysis of variance (ANOVA) followed by a Kruskal-Wallis test. Probability (p) values of <0.05 were regarded as significant. In Group I, IgG was detected in 96.7% serum and in 6.7% saliva samples. IgG was not detected in Group II. In Group III, cross-reactivity was observed for serum IgG in 26.7% and in 6.7% for saliva samples. In Group I, IgA was detected in 76.7% serum and 56.7% saliva samples. In Group II, 3.3% were positive for IgA in serum, whereas IgA was not detected in any saliva samples. Group III showed 6.7% serum and 26.7% saliva-positive samples. The sensitivity values for detection of IgG and IgA in serum samples were 96.7% and 76.7%, respectively. In saliva samples, the sensitivity values for detection of IgG and IgA were 6.7% and 56.7%, respectively. The specificity value was 100% for the detection of IgG in serum and for detection of IgG and IgA in saliva, and 96.7% for detection of IgA in serum samples. The proper choice of immunological diagnosis to supplement parasitological methods is essential to estimate the true prevalence of the parasite, and will permit analysis of population immune response profiles, particularly in northern Paraná state, where there are no previous reports.
Collapse
Affiliation(s)
- Larissa R Bosqui
- Departamento de Ciências Patológicas, Laboratório de Parasitologia, Universidade Estadual de Londrina, PR, Brazil
| | - Ana Lúcia R Gonçalves
- Departamento de Imunologia, Microbiologia e Parasitologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brazil
| | - Maria do Rosário F Gonçalves-Pires
- Departamento de Imunologia, Microbiologia e Parasitologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brazil
| | | | - Maria Cláudia N D de Menezes
- Departamento de Ciências Patológicas, Laboratório de Parasitologia, Universidade Estadual de Londrina, PR, Brazil
| | - Valter A Murad
- Ambulatório de Especialidades do Hospital Universitário de Londrina, PR, Brazil
| | - Fabiana M de Paula
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
| | - Wander R Pavanelli
- Departamento de Ciências Patológicas, Laboratório de Parasitologia, Universidade Estadual de Londrina, PR, Brazil
| | - Ivete Conchon-Costa
- Departamento de Ciências Patológicas, Laboratório de Parasitologia, Universidade Estadual de Londrina, PR, Brazil
| | - Julia Maria Costa-Cruz
- Departamento de Imunologia, Microbiologia e Parasitologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brazil
| | - Idessania N Costa
- Departamento de Ciências Patológicas, Laboratório de Parasitologia, Universidade Estadual de Londrina, PR, Brazil.
| |
Collapse
|
136
|
Sundin J, Rangel I, Repsilber D, Brummer RJ. Cytokine Response after Stimulation with Key Commensal Bacteria Differ in Post-Infectious Irritable Bowel Syndrome (PI-IBS) Patients Compared to Healthy Controls. PLoS One 2015; 10:e0134836. [PMID: 26366730 PMCID: PMC4569289 DOI: 10.1371/journal.pone.0134836] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/15/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Microbial dysbiosis and prolonged immune activation resulting in low-grade inflammation and intestinal barrier dysfunction have been suggested to be underlying causes of post-infectious irritable bowel syndrome (PI-IBS). The aim of this study was to evaluate the difference in cytokine response between mucosal specimens of PI-IBS patients and healthy controls (HC) after ex vivo stimulation with key anaerobic bacteria. METHODS Colonic biopsies from 11 PI-IBS patients and 10 HC were stimulated ex vivo with the commensal bacteria Bacteroides ovatus, Ruminococcus gnavus, Akkermansia muciniphila, Subdoligranulum variabile and Eubacterium limosum, respectively. The cytokine release (IL-1β, IL-2, IL-8, IL-10, IL-13, IL-17, TNF-α and IFN-γ) in stimulation supernatants was analyzed using the LUMINEX assay. Comparison of cytokine release between PI-IBS patients and healthy controls was performed taking both unstimulated and bacterially stimulated mucosal specimens into account. KEY RESULTS IL-13 release from mucosal specimens without bacterial stimulation was significantly lower in PI-IBS patients compared to HC (p < 0.05). After stimulation with Subdoligranulum variabile, IL-1β release from PI-IBS patients was significantly increased compared to HC (p < 0.05). Stimulation with Eubacterium limosum resulted in a significantly decreased IL-10 release in HC compared to PI-IBS patients (p < 0.05) and a tendency to decreased IL-13 release in HC compared to PI-IBS patients (p = 0.07). CONCLUSIONS & INFERENCES PI-IBS patients differ from HC with regard to cytokine release ex vivo after stimulation with selected commensal bacteria. Hence, our results support that the pathogenesis of PI-IBS comprises an altered immune response against commensal gut microbes.
Collapse
Affiliation(s)
- Johanna Sundin
- School of Health and Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ignacio Rangel
- School of Health and Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Dirk Repsilber
- School of Health and Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Robert-Jan Brummer
- School of Health and Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
137
|
Cao AT, Yao S, Gong B, Nurieva RI, Elson CO, Cong Y. Interleukin (IL)-21 promotes intestinal IgA response to microbiota. Mucosal Immunol 2015; 8:1072-82. [PMID: 25586558 PMCID: PMC4501922 DOI: 10.1038/mi.2014.134] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 12/13/2014] [Indexed: 02/04/2023]
Abstract
Commensal microbiota-specific T helper type 17 (Th17) cells are enriched in the intestines, which can convert into T follicular helper (Tfh) in Peyer's patches, and are crucial for production of intestinal immunoglobulin A (IgA) against microbiota; however, the role of Th17 and Tfh cytokines in regulating the mucosal IgA response to enteric microbiota is still not completely known. In this study, we found that intestinal IgA was impaired in mice deficient in interleukin (IL)-17 or IL-21 signaling. IL-21, but not IL-17, is able to augment B-cell differentiation to IgA(+) cells as mediated by transforming growth factor β1 (TGFβ1) and accelerate IgA class switch recombination (CSR). IL-21 and retinoic acid (RA) induce IgA(+) B-cell development and IgA production and drives autocrine TGFβ1 production to initiate IgA CSR. Repletion of T-cell-deficient TCRβxδ(-/-) mice with Th17 cells specific for commensal bacterial antigen increased the levels of IgA(+) B cells and IgA production in the intestine, which was blocked by neutralizing IL-21. Thus IL-21 functions to strongly augment IgA production under intestinal environment. Furthermore, IL-21 promotes intestinal B-cell homing through α4β7 expression, alone or with TGFβ and RA. Together, IL-21 from microbiota-specific Th17 and/or Tfh cells contributes to robust intestinal IgA levels by enhancing IgA(+) CSR, IgA production and B-cell trafficking into the intestine.
Collapse
Affiliation(s)
- Anthony T. Cao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Roza I. Nurieva
- Department of Immunology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030
| | - Charles O. Elson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
138
|
Abstract
Rotaviruses (RV) are ubiquitous, highly infectious, segmented double-stranded RNA genome viruses of importance in public health because of the severe acute gastroenteritis they cause in young children and many animal species. They are very well adapted to their host, with symptomatic and asymptomatic reinfections being virtually universal during the first 3 years of life. Antibodies are the major arm of the immune system responsible for protecting infants from RV reinfection. The relationship between the virus and the B cells (Bc) that produce these antibodies is complex and incompletely understood: most blood-circulating Bc that express RV-specific immunoglobulin (Ig) on their surface (RV-Ig) are naive Bc and recognize the intermediate capsid viral protein VP6 with low affinity. When compared to non-antigen-specific Bc, RV-Bc are enriched in CD27+ memory Bc (mBc) that express IgM. The Ig genes used by naive RV-Bc are different than those expressed by RV-mBc, suggesting that the latter do not primarily develop from the former. Although RV predominantly infects mature villus enterocytes, an acute systemic viremia also occurs and RV-Bc can be thought of as belonging to either the intestinal or systemic immune compartments. Serotype-specific or heterotypic RV antibodies appear to mediate protection by multiple mechanisms, including intracellular and extracellular homotypic and heterotypic neutralization. Passive administration of RV-Ig can be used either prophylactically or therapeutically. A better understanding of the Bc response generated against RV will improve our capacity to identify improved correlates of protection for RV vaccines.
Collapse
|
139
|
Ruff WE, Vieira SM, Kriegel MA. The role of the gut microbiota in the pathogenesis of antiphospholipid syndrome. Curr Rheumatol Rep 2015; 17:472. [PMID: 25475595 DOI: 10.1007/s11926-014-0472-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Infectious triggers are associated with the induction of transient antiphospholipid antibodies. One therefore wonders if microbes that permanently colonize us play a role in the pathogenesis of antiphospholipid syndrome (APS). The microbiota represents the collection of all microorganisms colonizing humans and is necessary for normal host physiology. The microbiota, however, is a constant stress on the immune system, which is tasked with recognizing and eliminating pathogenic microbes while tolerating commensal populations. A growing body of literature supports a critical role for the commensal-immune axis in the development of autoimmunity against colonized barriers (e.g., gut or skin) and sterile organs (e.g., pancreas or joints). Whether these interactions affect the development and sustainment of autoreactive CD4(+) T cells and pathogenic autoantibodies in APS is unknown. This review provides an overview of the current understanding of the commensal-immune axis in autoimmunity with a focus on the potential relevance to APS. Additionally, we discuss emerging findings supporting the involvement of the gut microbiota in a spontaneous model of APS, the (NZW × BXSB)F1 hybrid, and formalize hypotheses to explain how interactions between the immune system and the microbiota may influence human APS etiopathogenesis.
Collapse
Affiliation(s)
- William E Ruff
- Department of Immunobiology, Yale University School of Medicine, 300 George St, Suite 353G, New Haven, CT, 06511, USA,
| | | | | |
Collapse
|
140
|
The bilateral responsiveness between intestinal microbes and IgA. Trends Immunol 2015; 36:460-70. [PMID: 26169256 DOI: 10.1016/j.it.2015.06.006] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/01/2015] [Accepted: 06/19/2015] [Indexed: 12/30/2022]
Abstract
The immune system has developed strategies to maintain a homeostatic relationship with the resident microbiota. IgA is central in holding this relationship, as the most dominant immunoglobulin isotype at the mucosal surface of the intestine. Recent studies report a role for IgA in shaping the composition of the intestinal microbiota and exploit strategies to characterise IgA-binding bacteria for their inflammatory potential. We review these findings here, and place them in context of the current understanding of the range of microorganisms that contribute to the IgA repertoire and the pathways that determine the quality of the IgA response. We examine why only certain intestinal microbes are coated with IgA, and discuss how understanding the determinants of this specific responsiveness may provide insight into diseases associated with dysbiosis.
Collapse
|
141
|
Berkowska MA, Schickel JN, Grosserichter-Wagener C, de Ridder D, Ng YS, van Dongen JJM, Meffre E, van Zelm MC. Circulating Human CD27-IgA+ Memory B Cells Recognize Bacteria with Polyreactive Igs. THE JOURNAL OF IMMUNOLOGY 2015; 195:1417-26. [PMID: 26150533 DOI: 10.4049/jimmunol.1402708] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 06/08/2015] [Indexed: 01/01/2023]
Abstract
The vast majority of IgA production occurs in mucosal tissue following T cell-dependent and T cell-independent Ag responses. To study the nature of each of these responses, we analyzed the gene-expression and Ig-reactivity profiles of T cell-dependent CD27(+)IgA(+) and T cell-independent CD27(-)IgA(+) circulating memory B cells. Gene-expression profiles of IgA(+) subsets were highly similar to each other and to IgG(+) memory B cell subsets, with typical upregulation of activation markers and downregulation of inhibitory receptors. However, we identified the mucosa-associated CCR9 and RUNX2 genes to be specifically upregulated in CD27(-)IgA(+) B cells. We also found that CD27(-)IgA(+) B cells expressed Abs with distinct Ig repertoire and reactivity compared with those from CD27(+)IgA(+) B cells. Indeed, Abs from CD27(-)IgA(+) B cells were weakly mutated, often used Igλ chain, and were enriched in polyreactive clones recognizing various bacterial species. Hence, T cell-independent IgA responses are likely involved in the maintenance of gut homeostasis through the production of polyreactive mutated IgA Abs with cross-reactive anti-commensal reactivity.
Collapse
Affiliation(s)
- Magdalena A Berkowska
- Department of Immunology, Erasmus MC, University Medical Center, 3015 CN Rotterdam, the Netherlands
| | - Jean-Nicolas Schickel
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06511; and
| | | | - Dick de Ridder
- The Delft Bioinformatics Lab, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, 2628 CD Delft, the Netherlands
| | - Yen Shing Ng
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06511; and
| | - Jacques J M van Dongen
- Department of Immunology, Erasmus MC, University Medical Center, 3015 CN Rotterdam, the Netherlands
| | - Eric Meffre
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06511; and
| | - Menno C van Zelm
- Department of Immunology, Erasmus MC, University Medical Center, 3015 CN Rotterdam, the Netherlands;
| |
Collapse
|
142
|
Abstract
Immunoglobulin A (IgA) is the most abundantly produced immunoglobulin found primarily on mucosal surfaces. The generation of IgA and its involvement in mucosal immune responses have been intensely studied over the past years. IgA can be generated in T cell-dependent and T cell-independent pathways, and it has an important impact on maintaining homeostasis within the mucosal immune system. There is good evidence that B-1 cells contribute substantially to the production of mucosal IgA and thus play an important role in regulating commensal microbiota. However, whether B-1 cells produce antigen-specific or only nonspecific IgA remains to be determined. This review will discuss what is currently known about IgA production by B-1 cells and the functional relevance of B-1 cell-derived IgA both in vitro and in vivo.
Collapse
Affiliation(s)
- Almut Meyer-Bahlburg
- Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
143
|
How the Intricate Interaction among Toll-Like Receptors, Microbiota, and Intestinal Immunity Can Influence Gastrointestinal Pathology. J Immunol Res 2015; 2015:489821. [PMID: 26090491 PMCID: PMC4452102 DOI: 10.1155/2015/489821] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 10/01/2014] [Accepted: 10/27/2014] [Indexed: 12/12/2022] Open
Abstract
The gut is able to maintain tolerance to microbial and food antigens. The intestine minimizes the number of harmful bacteria by shaping the microbiota through a symbiotic relationship. In healthy human intestine, a constant homeostasis is maintained by the perfect regulation of microbial load and the immune response generated against it. Failure of this balance may result in various pathological conditions. Innate immune sensors, such as Toll-like receptors (TLRs), may be considered an interface among intestinal epithelial barrier, microbiota, and immune system. TLRs pathway, activated by pathogens, is involved in the pathogenesis of several infectious and inflammatory diseases. The alteration of the homeostasis between physiologic and pathogenic bacteria of intestinal flora causes a condition called dysbiosis. The breakdown of homeostasis by dysbiosis may increase susceptibility to inflammatory bowel diseases. It is evident that environment, genetics, and host immunity form a highly interactive regulatory triad that controls TLR function. Imbalanced relationships within this triad may promote aberrant TLR signaling, critically contributing to acute and chronic intestinal inflammatory processes, such as in IBD, colitis, and colorectal cancer. The study of interactions between different components of the immune systems and intestinal microbiota will open new horizons in the knowledge of gut inflammation.
Collapse
|
144
|
Lorin V, Mouquet H. Efficient generation of human IgA monoclonal antibodies. J Immunol Methods 2015; 422:102-10. [PMID: 25910833 DOI: 10.1016/j.jim.2015.04.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 04/15/2015] [Indexed: 11/17/2022]
Abstract
Immunoglobulin A (IgA) is the most abundant antibody isotype produced in humans. IgA antibodies primarily ensure immune protection of mucosal surfaces against invading pathogens, but also circulate and are present in large quantities in blood. IgAs are heterogeneous at a molecular level, with two IgA subtypes and the capacity to form multimers by interacting with the joining (J) chain. Here, we have developed an efficient strategy to rapidly generate human IgA1 and IgA2 monoclonal antibodies in their monomeric and dimeric forms. Recombinant monomeric and dimeric IgA1/IgA2 counterparts of a prototypical IgG1 monoclonal antibody, 10-1074, targeting the HIV-1 envelope protein, were produced in large amounts after expression cloning and transient transfection of 293-F cells. 10-1074 IgAs were FPLC-purified using a novel affinity-based resin engrafted with anti-IgA chimeric Fabs, followed by a monomers/multimers separation using size exclusion-based FPLC. ELISA binding experiments confirmed that the artificial IgA class switching of 10-1074 did not alter its antigen recognition. In summary, our technical approach allows the very efficient production of various forms of purified recombinant human IgA molecules, which are precious tools in dissecting IgA B-cell responses in physiological and pathophysiological conditions, and studying the biology, function and therapeutic potential of IgAs.
Collapse
Affiliation(s)
- Valérie Lorin
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris, 75015, France; CNRS-URA 1961, Paris, 75015, France
| | - Hugo Mouquet
- Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris, 75015, France; CNRS-URA 1961, Paris, 75015, France.
| |
Collapse
|
145
|
Trama AM, Moody MA, Alam SM, Jaeger FH, Lockwood B, Parks R, Lloyd KE, Stolarchuk C, Scearce R, Foulger A, Marshall DJ, Whitesides JF, Jeffries TL, Wiehe K, Morris L, Lambson B, Soderberg K, Hwang KK, Tomaras GD, Vandergrift N, Jackson KJL, Roskin KM, Boyd SD, Kepler TB, Liao HX, Haynes BF. HIV-1 envelope gp41 antibodies can originate from terminal ileum B cells that share cross-reactivity with commensal bacteria. Cell Host Microbe 2015; 16:215-226. [PMID: 25121750 DOI: 10.1016/j.chom.2014.07.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/08/2014] [Accepted: 07/01/2014] [Indexed: 12/23/2022]
Abstract
Monoclonal antibodies derived from blood plasma cells of acute HIV-1-infected individuals are predominantly targeted to the HIV Env gp41 and cross-reactive with commensal bacteria. To understand this phenomenon, we examined anti-HIV responses in ileum B cells using recombinant antibody technology and probed their relationship to commensal bacteria. The dominant ileum B cell response was to Env gp41. Remarkably, a majority (82%) of the ileum anti-gp41 antibodies cross-reacted with commensal bacteria, and of those, 43% showed non-HIV-1 antigen polyreactivity. Pyrosequencing revealed shared HIV-1 antibody clonal lineages between ileum and blood. Mutated immunoglobulin G antibodies cross-reactive with both Env gp41 and microbiota could also be isolated from the ileum of HIV-1 uninfected individuals. Thus, the gp41 commensal bacterial antigen cross-reactive antibodies originate in the intestine, and the gp41 Env response in HIV-1 infection can be derived from a preinfection memory B cell pool triggered by commensal bacteria that cross-react with Env.
Collapse
Affiliation(s)
- Ashley M Trama
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA.
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Pediatrics, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - S Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Frederick H Jaeger
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Bradley Lockwood
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Krissey E Lloyd
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Christina Stolarchuk
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Richard Scearce
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Andrew Foulger
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Dawn J Marshall
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - John F Whitesides
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Thomas L Jeffries
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Lynn Morris
- Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg 2131, South Africa
| | - Bronwen Lambson
- Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg 2131, South Africa
| | - Kelly Soderberg
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Kwan-Ki Hwang
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Surgery, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Nathan Vandergrift
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | | | - Krishna M Roskin
- Department of Pathology, Stanford University, Palo Alto, CA 94305, USA
| | - Scott D Boyd
- Department of Pathology, Stanford University, Palo Alto, CA 94305, USA
| | - Thomas B Kepler
- Department of Microbiology, Boston University, Boston, MA 02215, USA
| | - Hua-Xin Liao
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine and Duke Global Health Institute, Durham, NC 27710, USA.
| |
Collapse
|
146
|
Peterson DA, Planer JD, Guruge JL, Xue L, Downey-Virgin W, Goodman AL, Seedorf H, Gordon JI. Characterizing the interactions between a naturally primed immunoglobulin A and its conserved Bacteroides thetaiotaomicron species-specific epitope in gnotobiotic mice. J Biol Chem 2015; 290:12630-49. [PMID: 25795776 DOI: 10.1074/jbc.m114.633800] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Indexed: 12/13/2022] Open
Abstract
The adaptive immune response to the human gut microbiota consists of a complex repertoire of antibodies interacting with a broad range of taxa. Fusing intestinal lamina propria lymphocytes from mice monocolonized with Bacteroides thetaiotaomicron to a myeloma fusion partner allowed us to recover hybridomas that captured naturally primed, antigen-specific antibody responses representing multiple isotypes, including IgA. One of these hybridomas, 260.8, produced a monoclonal antibody that recognizes an epitope specific for B. thetaiotaomicron isolates in a large panel of hospital- and community-acquired Bacteroides. Whole genome transposon mutagenesis revealed a 19-gene locus, involved in LPS O-antigen polysaccharide synthesis and conserved among multiple B. thetaiotaomicron isolates, that is required for 260.8 epitope expression. Mutants in this locus exhibited marked fitness defects in vitro during growth in rich medium and in gnotobiotic mice colonized with defined communities of human gut symbionts. Expression of the 260.8 epitope was sustained during 10 months of daily passage in vitro and during 14 months of monocolonization of gnotobiotic wild-type, Rag1-/-, or Myd88-/- mice. Comparison of gnotobiotic Rag1-/- mice with and without subcutaneous 260.8 hybridomas disclosed that this IgA did not affect B. thetaiotaomicron population density or suppress 260.8 epitope production but did affect bacterial gene expression in ways emblematic of a diminished host innate immune response. Our study illustrates an approach for (i) generating diagnostic antibodies, (ii) characterizing IgA responses along a continuum of specificity/degeneracy that defines the IgA repertoire to gut symbionts, and (iii) identifying immunogenic epitopes that affect competitiveness and help maintain host-microbe mutualism.
Collapse
Affiliation(s)
- Daniel A Peterson
- From the Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63108 and Department of Pathology, The Johns Hopkins University, Baltimore, Maryland 21205
| | - Joseph D Planer
- From the Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63108 and
| | - Janaki L Guruge
- From the Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63108 and
| | - Lai Xue
- From the Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63108 and
| | - Whitt Downey-Virgin
- From the Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63108 and
| | - Andrew L Goodman
- From the Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63108 and
| | - Henning Seedorf
- From the Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63108 and
| | - Jeffrey I Gordon
- From the Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63108 and
| |
Collapse
|
147
|
Ruff WE, Kriegel MA. Autoimmune host-microbiota interactions at barrier sites and beyond. Trends Mol Med 2015; 21:233-44. [PMID: 25771098 DOI: 10.1016/j.molmed.2015.02.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/21/2015] [Accepted: 02/12/2015] [Indexed: 02/07/2023]
Abstract
The microbiota is considered to be an important factor influencing the pathogenesis of autoimmunity at both barrier sites and internal organs. Impinging on innate and adaptive immunity, commensals exert protective or detrimental effects on various autoimmune animal models. Human microbiome studies of autoimmunity remain largely descriptive, but suggest a role for dysbiosis in autoimmune disease. Humanized gnotobiotic approaches have advanced our understanding of immune-commensal interactions, but little is known about the mechanisms in autoimmunity. We propose that, similarly to infectious agents, the microbiota mediates autoimmunity via bystander activation, epitope spread, and, particularly under homeostatic conditions, via crossreactivity. This review presents an overview of the current literature concluding with outstanding questions in this field.
Collapse
Affiliation(s)
- William E Ruff
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Martin A Kriegel
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Section of Rheumatology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
148
|
Van Praet JT, Donovan E, Vanassche I, Drennan MB, Windels F, Dendooven A, Allais L, Cuvelier CA, van de Loo F, Norris PS, Kruglov AA, Nedospasov SA, Rabot S, Tito R, Raes J, Gaboriau-Routhiau V, Cerf-Bensussan N, Van de Wiele T, Eberl G, Ware CF, Elewaut D. Commensal microbiota influence systemic autoimmune responses. EMBO J 2015; 34:466-74. [PMID: 25599993 PMCID: PMC4331001 DOI: 10.15252/embj.201489966] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/12/2014] [Accepted: 12/01/2014] [Indexed: 12/22/2022] Open
Abstract
Antinuclear antibodies are a hallmark feature of generalized autoimmune diseases, including systemic lupus erythematosus and systemic sclerosis. However, the processes underlying the loss of tolerance against nuclear self-constituents remain largely unresolved. Using mice deficient in lymphotoxin and Hox11, we report that approximately 25% of mice lacking secondary lymphoid organs spontaneously develop specific antinuclear antibodies. Interestingly, we find this phenotype is not caused by a defect in central tolerance. Rather, cell-specific deletion and in vivo lymphotoxin blockade link these systemic autoimmune responses to the formation of gut-associated lymphoid tissue in the neonatal period of life. We further demonstrate antinuclear antibody production is influenced by the presence of commensal gut flora, in particular increased colonization with segmented filamentous bacteria, and IL-17 receptor signaling. Together, these data indicate that neonatal colonization of gut microbiota influences generalized autoimmunity in adult life.
Collapse
Affiliation(s)
- Jens T Van Praet
- Laboratory for Molecular Immunology and Inflammation, Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Erin Donovan
- Laboratory for Molecular Immunology and Inflammation, Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Inge Vanassche
- Laboratory for Molecular Immunology and Inflammation, Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Michael B Drennan
- Laboratory for Molecular Immunology and Inflammation, Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Fien Windels
- Laboratory for Molecular Immunology and Inflammation, Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Amélie Dendooven
- Department of Pathology, University Medical Center, Utrecht, the Netherlands
| | - Liesbeth Allais
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | | | - Fons van de Loo
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Paula S Norris
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Andrey A Kruglov
- German Rheumatism Research Center (DRFZ), A Leibniz Institute, Berlin, Germany Belozersky Institute of Physico-Chemical Biology and Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| | - Sergei A Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, and Lomonosov Moscow State University, Moscow, Russia
| | - Sylvie Rabot
- INRA, UMR1319 Micalis, Jouy-en-Josas, France AgroParisTech Micalis, Jouy-en-Josas, France
| | - Raul Tito
- Bioinformatics and (eco-)systems Biology Laboratory, Department of Microbiology and Immunology, Rega Institute VIB Center for the Biology of Disease, KU Leuven, Belgium
| | - Jeroen Raes
- Bioinformatics and (eco-)systems Biology Laboratory, Department of Microbiology and Immunology, Rega Institute VIB Center for the Biology of Disease, KU Leuven, Belgium
| | - Valerie Gaboriau-Routhiau
- INRA, UMR1319 Micalis, Jouy-en-Josas, France INSERM UMR1163, Laboratory of Intestinal Immunity, Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, Paris, France
| | - Nadine Cerf-Bensussan
- INSERM UMR1163, Laboratory of Intestinal Immunity, Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, Paris, France
| | - Tom Van de Wiele
- Laboratory of Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | - Gérard Eberl
- Lymphoid Tissue Development Group, Institut Pasteur, Paris, France
| | - Carl F Ware
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Dirk Elewaut
- Laboratory for Molecular Immunology and Inflammation, Department of Rheumatology, Ghent University Hospital, Ghent, Belgium VIB Inflammation Research Center Ghent University, Ghent, Belgium
| |
Collapse
|
149
|
Gutzeit C, Magri G, Cerutti A. Intestinal IgA production and its role in host-microbe interaction. Immunol Rev 2015; 260:76-85. [PMID: 24942683 DOI: 10.1111/imr.12189] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Complex and diverse communities of bacteria establish mutualistic and symbiotic relationships with the gut after birth. The intestinal immune system responds to bacterial colonization by acquiring a state of hypo-responsiveness against commensals and active readiness against pathogens. The resulting homeostatic balance involves a continuous dialog between the microbiota and lymphocytes with the intermediation of epithelial and dendritic cells. This dialog causes massive production of immunoglobulin A (IgA), a non-inflammatory antibody specialized in mucosal protection. Here, we discuss recent advances on the regulation of intestinal IgA responses and their role in host-microbe interaction.
Collapse
Affiliation(s)
- Cindy Gutzeit
- Immunology Institute, Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | |
Collapse
|
150
|
Antibody repertoire diversification through VH gene replacement in mice cloned from an IgA plasma cell. Proc Natl Acad Sci U S A 2015; 112:E450-7. [PMID: 25609671 DOI: 10.1073/pnas.1417988112] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In mammals, VDJ recombination is responsible for the establishment of a highly diversified preimmune antibody repertoire. Acquisition of a functional Ig heavy (H) chain variable (V) gene rearrangement is thought to prevent further recombination at the IgH locus. Here, we describe VHQ52(NT); Vκgr32(NT) Ig monoclonal mice reprogrammed from the nucleus of an intestinal IgA(+) plasma cell. In VHQ52(NT) mice, IgA replaced IgM to drive early B-cell development and peripheral B-cell maturation. In VHQ52(NT) animals, over 20% of mature B cells disrupted the single productive, nonautoimmune IgH rearrangement through VH replacement and exchanged it with a highly diversified pool of IgH specificities. VH replacement occurred in early pro-B cells, was independent of pre-B-cell receptor signaling, and involved predominantly one adjacent VH germ-line gene. VH replacement was also identified in 5% of peripheral B cells of mice inheriting a different productive VH rearrangement expressed in the form of an IgM H chain. In summary, editing of a productive IgH rearrangement through VH replacement can account for up to 20% of the IgH repertoire expressed by mature B cells.
Collapse
|