301
|
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: 126] [Impact Index Per Article: 15.8] [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
|
302
|
Belkaid Y, Harrison OJ. Homeostatic Immunity and the Microbiota. Immunity 2017; 46:562-576. [PMID: 28423337 DOI: 10.1016/j.immuni.2017.04.008] [Citation(s) in RCA: 799] [Impact Index Per Article: 99.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 04/04/2017] [Accepted: 04/05/2017] [Indexed: 12/27/2022]
Abstract
The microbiota plays a fundamental role in the induction, education, and function of the host immune system. In return, the host immune system has evolved multiple means by which to maintain its symbiotic relationship with the microbiota. The maintenance of this dialogue allows the induction of protective responses to pathogens and the utilization of regulatory pathways involved in the sustained tolerance to innocuous antigens. The ability of microbes to set the immunological tone of tissues, both locally and systemically, requires tonic sensing of microbes and complex feedback loops between innate and adaptive components of the immune system. Here we review the dominant cellular mediators of these interactions and discuss emerging themes associated with our current understanding of the homeostatic immunological dialogue between the host and its microbiota.
Collapse
Affiliation(s)
- Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA; NIAID Microbiome Program, NIH, Bethesda, MD 20892, USA.
| | - Oliver J Harrison
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
303
|
Jones AP, Trend S, Byrne SN, Fabis-Pedrini MJ, Geldenhuys S, Nolan D, Booth DR, Carroll WM, Lucas RM, Kermode AG, Hart PH. Altered regulatory T-cell fractions and Helios expression in clinically isolated syndrome: clues to the development of multiple sclerosis. Clin Transl Immunology 2017; 6:e143. [PMID: 28690849 PMCID: PMC5493587 DOI: 10.1038/cti.2017.18] [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: 03/09/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 02/06/2023] Open
Abstract
Development of multiple sclerosis (MS) is frequently preceded by an acute or subacute neurological disturbance referred to as clinically isolated syndrome (CIS). The specific immunological disturbances present in CIS remain underexamined. This study analysed peripheral blood mononuclear cells from n=18 treatment-naive individuals with recently diagnosed CIS (<120 days) for disturbances in the phenotype of T regulatory (Treg), follicular T regulatory (Tfr), T helper (Th), follicular T helper (Tfh) and B cells. Relative to healthy controls (n=19), CIS was associated with lower proportions of suppressive CD45RA+FoxP3lo Treg and Tfr cells and greater proportions of non-suppressive CD45RA−FoxP3lo and Th17-like Treg and Tfr. Lower Helios expression (maen fluorescent intensity) was measured across all Treg and Tfr fractions in the CIS group, suggesting less potent regulatory function. Greater frequencies of activated, efficient B-cell helper Tfh subsets and a trend for a higher proportion of IgD−CD27− B cells was also detected in the CIS group, characteristics that were positively correlated with Treg and Tfr Helios expression. These results indicate that Treg and Tfr impairment is an early feature in MS.
Collapse
Affiliation(s)
- Anderson P Jones
- Telethon Kids Institute, University of Western Australia, West Perth, Western Australia, Australia
| | - Stephanie Trend
- Telethon Kids Institute, University of Western Australia, West Perth, Western Australia, Australia
| | - Scott N Byrne
- Cellular Photoimmunology Group, Infectious Diseases & Immunology, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia.,Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Marzena J Fabis-Pedrini
- Centre for Neuromuscular and Neurological Disorders, Western Australian Neuroscience Research Institute, University of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Sian Geldenhuys
- Telethon Kids Institute, University of Western Australia, West Perth, Western Australia, Australia
| | - David Nolan
- Institute for Immunology and Infectious Disease, Murdoch University, Perth, Western Australia, Australia.,Immunology Department, Royal Perth Hospital, Perth, Western Australia, Australia
| | - David R Booth
- Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - William M Carroll
- Centre for Neuromuscular and Neurological Disorders, Western Australian Neuroscience Research Institute, University of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Robyn M Lucas
- National Centre for Epidemiology and Population Health, Research School of Population Health, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Allan G Kermode
- Centre for Neuromuscular and Neurological Disorders, Western Australian Neuroscience Research Institute, University of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,Institute for Immunology and Infectious Disease, Murdoch University, Perth, Western Australia, Australia
| | - Prue H Hart
- Telethon Kids Institute, University of Western Australia, West Perth, Western Australia, Australia
| |
Collapse
|
304
|
Niemz J, Kliche S, Pils MC, Morrison E, Manns A, Freund C, Crittenden JR, Graybiel AM, Galla M, Jänsch L, Huehn J. The Guanine-Nucleotide Exchange Factor Caldag Gefi Fine-Tunes Functional Properties of Regulatory T Cells. Eur J Microbiol Immunol (Bp) 2017; 7:112-126. [PMID: 28690878 PMCID: PMC5495083 DOI: 10.1556/1886.2017.00007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 04/27/2017] [Indexed: 12/12/2022] Open
Abstract
Using quantitative phosphopeptide sequencing of unstimulated versus stimulated primary murine Foxp3+ regulatory and Foxp3– conventional T cells (Tregs and Tconv, respectively), we detected a novel and differentially regulated tyrosine phosphorylation site within the C1 domain of the guanine-nucleotide exchange factor CalDAG GEFI. We hypothesized that the Treg-specific and activation-dependent reduced phosphorylation at Y523 allows binding of CalDAG GEFI to diacylglycerol, thereby impacting the formation of a Treg-specific immunological synapse. However, diacylglycerol binding assays of phosphomutant C1 domains of CalDAG GEFI could not confirm this hypothesis. Moreover, CalDAG GEFI–/– mice displayed normal Treg numbers in thymus and secondary lymphoid organs, and CalDAG GEFI–/– Tregs showed unaltered in vitro suppressive capacity when compared to CalDAG GEFI+/+ Tregs. Interestingly, when tested in vivo, CalDAG GEFI–/– Tregs displayed a slightly reduced suppressive ability in the transfer colitis model when compared to CalDAG GEFI+/+ Tregs. Additionally, CRISPR-Cas9-generated CalDAG GEFI–/– Jurkat T cell clones showed reduced adhesion to ICAM-1 and fibronectin when compared to CalDAG GEFI-competent Jurkat T cells. Therefore, we speculate that deficiency in CalDAG GEFI impairs adherence of Tregs to antigen-presenting cells, thereby impeding formation of a fully functional immunological synapse, which finally results in a reduced suppressive potential.
Collapse
Affiliation(s)
- Jana Niemz
- Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Stefanie Kliche
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Marina C Pils
- Mousepathology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Eliot Morrison
- Institute for Chemistry and Biochemistry, Free University Berlin, 14195 Berlin, Germany
| | - Annika Manns
- Institute for Chemistry and Biochemistry, Free University Berlin, 14195 Berlin, Germany
| | - Christian Freund
- Institute for Chemistry and Biochemistry, Free University Berlin, 14195 Berlin, Germany
| | - Jill R Crittenden
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Ann M Graybiel
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Lothar Jänsch
- Cellular Proteomics, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| |
Collapse
|
305
|
Stephen-Victor E, Bosschem I, Haesebrouck F, Bayry J. The Yin and Yang of regulatory T cells in infectious diseases and avenues to target them. Cell Microbiol 2017; 19. [DOI: 10.1111/cmi.12746] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Emmanuel Stephen-Victor
- Institut National de la Santé et de la Recherche Médicale; Paris France
- Centre de Recherche des Cordeliers; Equipe-Immunopathologie et Immunointervention Thérapeutique; Paris France
- Sorbonne Universités; Université Pierre et Marie Curie; Paris France
| | - Iris Bosschem
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - Freddy Haesebrouck
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale; Paris France
- Centre de Recherche des Cordeliers; Equipe-Immunopathologie et Immunointervention Thérapeutique; Paris France
- Sorbonne Universités; Université Pierre et Marie Curie; Paris France
- Université Paris Descartes; Sorbonne Paris Cité; Paris France
| |
Collapse
|
306
|
Adamczyk A, Gageik D, Frede A, Pastille E, Hansen W, Rueffer A, Buer J, Büning J, Langhorst J, Westendorf AM. Differential expression of GPR15 on T cells during ulcerative colitis. JCI Insight 2017; 2:90585. [PMID: 28422750 DOI: 10.1172/jci.insight.90585] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 03/14/2017] [Indexed: 12/16/2022] Open
Abstract
G protein-coupled receptor 15 (GPR15) was recently highlighted as a colon-homing receptor for murine and human CD4+ T cells. The aim of this study was to explore the functional phenotype of human GPR15+CD4+ T cells, focusing on Tregs and effector T cells (Teffs), and to determine whether GPR15 is the driver for the migration of T cells to the colon during ulcerative colitis (UC). In the peripheral blood, GPR15 was expressed on Tregs and Teffs; both GPR15+ T cell subsets produced less IFN-γ and IL-4 but more IL-17 after stimulation and showed a higher migration activity compared with GPR15-CD4+ T cells. In UC patients, GPR15 expression was increased on Tregs in the peripheral blood but not on Teffs. Interestingly, the expression of GPR15 was significantly enhanced on colonic T cells of UC patients in noninflamed biopsies but not in inflamed biopsies. The differential expression of GPR15 in UC patients was accompanied by a significant reduction of bacterial immunoregulatory metabolites in the feces. In conclusion, GPR15 expression on CD4+ T cells is altered in UC patients, which may have implications for the development of therapeutic approaches to target T cell trafficking to the colon.
Collapse
Affiliation(s)
- Alexandra Adamczyk
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Daniel Gageik
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Annika Frede
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Eva Pastille
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jürgen Büning
- Department of Internal Medicine I, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Jost Langhorst
- Center of Integrative Gastroenterology, Kliniken Essen-Mitte, Essen, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
307
|
Bauché D, Marie JC. Transforming growth factor β: a master regulator of the gut microbiota and immune cell interactions. Clin Transl Immunology 2017; 6:e136. [PMID: 28523126 PMCID: PMC5418590 DOI: 10.1038/cti.2017.9] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 02/20/2017] [Accepted: 02/20/2017] [Indexed: 12/12/2022] Open
Abstract
The relationship between host organisms and their microbiota has co-evolved towards an inter-dependent network of mutualistic interactions. This interplay is particularly well studied in the gastrointestinal tract, where microbiota and host immune cells can modulate each other directly, as well as indirectly, through the production and release of chemical molecules and signals. In this review, we define the functional impact of transforming growth factor-beta (TGF-β) on this complex interplay, especially through its modulation of the activity of local regulatory T cells (Tregs), type 17 helper (Th17) cells, innate lymphoid cells (ILCs) and B cells.
Collapse
Affiliation(s)
- David Bauché
- Department of Immunology, Virology and Inflammation, Cancer Research Center of Lyon UMR INSERM1052, CNRS 5286, Centre Léon Bérard Hospital, Université de Lyon, Equipe labellisée LIGUE, Lyon, France.,TGF-β and Immuno-evasion Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julien C Marie
- Department of Immunology, Virology and Inflammation, Cancer Research Center of Lyon UMR INSERM1052, CNRS 5286, Centre Léon Bérard Hospital, Université de Lyon, Equipe labellisée LIGUE, Lyon, France.,TGF-β and Immuno-evasion Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
308
|
Barthels C, Ogrinc A, Steyer V, Meier S, Simon F, Wimmer M, Blutke A, Straub T, Zimber-Strobl U, Lutgens E, Marconi P, Ohnmacht C, Garzetti D, Stecher B, Brocker T. CD40-signalling abrogates induction of RORγt + Treg cells by intestinal CD103 + DCs and causes fatal colitis. Nat Commun 2017; 8:14715. [PMID: 28276457 PMCID: PMC5347138 DOI: 10.1038/ncomms14715] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 01/25/2017] [Indexed: 12/23/2022] Open
Abstract
Immune homeostasis in intestinal tissues depends on the generation of regulatory T (Treg) cells. CD103+ dendritic cells (DCs) acquire microbiota-derived material from the gut lumen for transport to draining lymph nodes and generation of receptor-related orphan γt+ (RORγt+) Helios−-induced Treg (iTreg) cells. Here we show CD40-signalling as a microbe-independent signal that can induce migration of CD103+ DCs from the lamina propria (LP) to the mesenteric lymph nodes. Transgenic mice with constitutive CD11c-specific CD40-signalling have reduced numbers of CD103+ DCs in LP and a low frequency of RORγt+Helios− iTreg cells, exacerbated inflammatory Th1/Th17 responses, high titres of microbiota-specific immunoglobulins, dysbiosis and fatal colitis, but no pathology is detected in other tissues. Our data demonstrate a CD40-dependent mechanism capable of abrogating iTreg cell induction by DCs, and suggest that the CD40L/CD40-signalling axis might be able to intervene in the generation of new iTreg cells in order to counter-regulate immune suppression to enhance immunity. CD103+ dendritic cells induce iTreg cells to maintain immune balance in the gut, but how CD40-signalling regulates this process is unclear. Here the authors show that mice with constitutive CD11c-specific CD40-signalling have altered CD103+ dendritic cell migration, reduced iTreg cell induction, and fatal colitis.
Collapse
Affiliation(s)
- Christian Barthels
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Ana Ogrinc
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Verena Steyer
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Stefanie Meier
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Ferdinand Simon
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Maria Wimmer
- Center of Allergy Environment (ZAUM), Helmholtz Center and TU Munich, Neuherberg 85764, Germany
| | - Andreas Blutke
- Section of Animal Pathology, Department of Veterinary Clinical Sciences, LMU Munich, Munich 80539, Germany
| | - Tobias Straub
- Bioinformatics core unit, BMC, LMU Munich, Großhaderner Strasse 9, Planegg-Munich 82152, Germany
| | | | - Esther Lutgens
- Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, LMU Munich, Munich 80336, Germany.,Department of Medical Biochemistry, AMC, Amsterdam 1105AZ, The Netherlands
| | - Peggy Marconi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara 44121, Italy
| | - Caspar Ohnmacht
- Center of Allergy Environment (ZAUM), Helmholtz Center and TU Munich, Neuherberg 85764, Germany
| | - Debora Garzetti
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, German Center for Infection Research (DZIF), Partner Site Munich, LMU Munich, Munich 80336, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, German Center for Infection Research (DZIF), Partner Site Munich, LMU Munich, Munich 80336, Germany
| | - Thomas Brocker
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| |
Collapse
|
309
|
Ferreira RC, Rainbow DB, Rubio García A, Pekalski ML, Porter L, Oliveira JJ, Waldron-Lynch F, Wicker LS, Todd JA. Human IL-6R hiTIGIT - CD4 +CD127 lowCD25 + T cells display potent in vitro suppressive capacity and a distinct Th17 profile. Clin Immunol 2017; 179:25-39. [PMID: 28284938 PMCID: PMC5471606 DOI: 10.1016/j.clim.2017.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/03/2017] [Accepted: 03/07/2017] [Indexed: 12/12/2022]
Abstract
To date many clinical studies aim to increase the number and/or fitness of CD4+ CD127lowCD25+ regulatory T cells (Tregs) in vivo to harness their regulatory potential in the context of treating autoimmune disease. Here, we sought to define the phenotype and function of Tregs expressing the highest levels of IL-6 receptor (IL-6R). We have identified a population of CD4+ CD127lowCD25+ TIGIT− T cells distinguished by their elevated IL-6R expression that lacked expression of HELIOS, showed higher CTLA-4 expression, and displayed increased suppressive capacity compared to IL-6RhiTIGIT+ Tregs. IL-6RhiTIGIT− CD127lowCD25+ T cells contained a majority of cells demethylated at FOXP3 and displayed a Th17 transcriptional signature, including RORC (RORγt) and the capacity of producing both pro- and anti-inflammatory cytokines, such as IL-17, IL-22 and IL-10. We propose that in vivo, in the presence of IL-6-associated inflammation, the suppressive function of CD4+ CD127lowCD25+ FOXP3+ IL-6RhiTIGIT− T cells is temporarily disarmed allowing further activation of the effector functions and potential pathogenic tissue damage. IL-6R is highly expressed in certain Treg subsets. IL-6RhiTIGIT− CD127lowCD25+ T cells contain a subset of antigen-experienced Tregs with potent suppression capacity. IL-6RhiTIGIT− Tregs display a Th17 transcriptional profile ex vivo, and the capacity to migrate to the gut. IL-2 treatment in humans elicits the trafficking and expansion of Tregs in circulation.
Collapse
Affiliation(s)
- Ricardo C Ferreira
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.
| | - Daniel B Rainbow
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Arcadio Rubio García
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Marcin L Pekalski
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Linsey Porter
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - João J Oliveira
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Frank Waldron-Lynch
- Experimental Medicine and Immunotherapeutics, Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK; NIHR Cambridge Clinical Trial Unit, Cambridge NHS University Hospitals Trust, Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Linda S Wicker
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - John A Todd
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
310
|
Kluger MA, Nosko A, Ramcke T, Goerke B, Meyer MC, Wegscheid C, Luig M, Tiegs G, Stahl RAK, Steinmetz OM. RORγt expression in T regs promotes systemic lupus erythematosus via IL-17 secretion, alteration of T reg phenotype and suppression of Th2 responses. Clin Exp Immunol 2017; 188:63-78. [PMID: 27880975 DOI: 10.1111/cei.12905] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2016] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a common autoimmune disorder with a complex and poorly understood immunopathogenesis. However, a pathogenic role for the T helper type 17 (Th17) axis was demonstrated by many studies, while regulatory T cells (Tregs ) were shown to mediate protection. Recently, we and others characterized a novel and independent T cell population expressing both the Treg characteristic transcription factor forkhead box protein 3 (FoxP3) and the Th17-defining retinoic acid receptor-related orphan nuclear receptor γt (RORγt). Studies in a model of acute glomerulonephritis unveiled potent regulatory, but also proinflammatory, functions of RORγt+ FoxP3+ Tregs . This bi-functional nature prompted us to suggest the name 'biTregs '. Importantly, the pathogenic biTreg effects were dependent upon expression of RORγt. We thus aimed to evaluate the contribution of RORγt+ FoxP3+ biTregs to pristane-induced SLE and explored the therapeutic potential of interference with RORγt activation. Our analyses revealed expansion of IL-17 producing biTregs in a distinctive time-course and organ-specific pattern, coincident with the development of autoimmunity and tissue injury. Importantly, specific ablation of RORγt activation in endogenous biTregs resulted in significant amelioration of pristane-induced pulmonary vasculitis and lupus nephritis. As potential mechanisms underlying the observed protection, we found that secretion of IL-17 by biTregs was abrogated completely in FoxP3Cre × RORCfl/fl mice. Furthermore, Tregs showed a more activated phenotype after cell-specific inactivation of RORγt signalling. Finally, and remarkably, biTregs were found to potently suppress anti-inflammatory Th2 immunity in a RORγt-dependent manner. Our study thus identifies biTregs as novel players in SLE and advocates RORγt-directed interventions as promising therapeutic strategies.
Collapse
Affiliation(s)
- M A Kluger
- III Medizinische Klinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - A Nosko
- III Medizinische Klinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - T Ramcke
- III Medizinische Klinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - B Goerke
- III Medizinische Klinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - M C Meyer
- III Medizinische Klinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - C Wegscheid
- Institut für experimentelle Immunologie und Hepatologie, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - M Luig
- III Medizinische Klinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - G Tiegs
- Institut für experimentelle Immunologie und Hepatologie, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - R A K Stahl
- III Medizinische Klinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| | - O M Steinmetz
- III Medizinische Klinik, Universitätsklinikum Eppendorf, Hamburg, Germany
| |
Collapse
|
311
|
Eberl G. RORγt, a multitask nuclear receptor at mucosal surfaces. Mucosal Immunol 2017; 10:27-34. [PMID: 27706126 DOI: 10.1038/mi.2016.86] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/30/2016] [Indexed: 02/07/2023]
Abstract
RORγt is a nuclear hormone receptor that has followed an exponential success carrier. Its modest origins as an orphan receptor cloned from human pancreas blossomed within 15 years into a critical regulator of anti-microbial immunity and a major target in the fight against inflammatory pathologies. Here, I review its role as a transcription factor required for the generation of type 3 lymphoid cells, which induce the development of lymphoid tissues, provide resistance of epithelial stem cells to injury, maintain homeostasis with the symbiotic microbiota, orchestrate defense against extracellular microbes, and regulate allergic responses. RORγt is also an intriguing molecule that is regulated by the circadian rhythm and includes cholesterol metabolites as ligands. RORγt therefore links anti-microbial immunity with circadian rhythms and steroids, the logic of which remains to be understood.
Collapse
Affiliation(s)
- G Eberl
- Institut Pasteur, Microenvironment & Immunity Unit, Department of Immunology, Paris, France.,INSERM U1224, Paris, France
| |
Collapse
|
312
|
Ren J, Li B. The Functional Stability of FOXP3 and RORγt in Treg and Th17 and Their Therapeutic Applications. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 107:155-189. [PMID: 28215223 DOI: 10.1016/bs.apcsb.2016.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The balance of CD4+CD25+FOXP3+ regulatory T cells (Tregs) and effector T cells plays a key role in maintaining immune homeostasis, while the imbalance of them is related to many inflammatory diseases in both human and mice. Here we discuss about the plasticity of Tregs and Th17 cells, and the related human diseases resulted from the imbalance of them. Further, we will focus on the mechanisms regulating the plasticity between Tregs and Th17 cells and the potential therapeutic strategies by targeting regulators of the expression and activity of FOXP3 and RORγt or regulators of Treg/Th17 balance in autoimmune diseases, allergy, infection, and cancer.
Collapse
Affiliation(s)
- J Ren
- Key Laboratory of Molecular Virology and Immunology, CAS Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Universities and Colleges Admissions Service, Shanghai, PR China
| | - B Li
- Key Laboratory of Molecular Virology and Immunology, CAS Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Universities and Colleges Admissions Service, Shanghai, PR China; Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
313
|
CD4 + T Cell Fate in Glomerulonephritis: A Tale of Th1, Th17, and Novel Treg Subtypes. Mediators Inflamm 2016; 2016:5393894. [PMID: 27974866 PMCID: PMC5126430 DOI: 10.1155/2016/5393894] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 06/17/2016] [Accepted: 10/12/2016] [Indexed: 12/12/2022] Open
Abstract
Multiple studies have identified CD4+ T cells as central players of glomerulonephritis (GN). Cells of the Th1 and Th17 responses cause renal tissue damage, while Tregs mediate protection. Recently, a high degree of plasticity among these T cell lineages was proposed. During inflammation, Th17 cells were shown to have the potential of transdifferentiation into Th1, Th2, or alternatively anti-inflammatory Tr1 cells. Currently available data from studies in GN, however, do not indicate relevant Th17 to Th1 or Th2 conversion, leaving the Th17 cell fate enigmatic. Tregs, on the other hand, were speculated to transdifferentiate into Th17 cells. Again, data from GN do not support this concept. Rather, it seems that previously unrecognized subspecialized effector Treg lineages exist. These include Th1 specific Treg1 as well as Th17 directed Treg17 cells. Furthermore, a bifunctional Treg subpopulation was recently identified in GN, which secrets IL-17 and coexpresses Foxp3 together with the Th17 characteristic transcription factor RORγt. Similarities between these different and highly specialized effector Treg subpopulations with the corresponding T helper effector cell lineages might have resulted in previous misinterpretation as Treg transdifferentiation. In summary, Th17 cells have a relatively stable phenotype during GN, while, in the case of Tregs, currently available data suggest lineage heterogeneity rather than plasticity.
Collapse
|
314
|
Induced Regulatory T Cells: Their Development, Stability, and Applications. Trends Immunol 2016; 37:803-811. [DOI: 10.1016/j.it.2016.08.012] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/11/2016] [Accepted: 08/17/2016] [Indexed: 12/25/2022]
|
315
|
Solomon BD, Hsieh CS. Antigen-Specific Development of Mucosal Foxp3+RORγt+ T Cells from Regulatory T Cell Precursors. THE JOURNAL OF IMMUNOLOGY 2016; 197:3512-3519. [PMID: 27671109 DOI: 10.4049/jimmunol.1601217] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/02/2016] [Indexed: 12/22/2022]
Abstract
Foxp3+retinoic acid-related orphan receptor (ROR)γt+ T cells have recently been characterized as an immunoregulatory population highly enriched in the colon lamina propria. However, their developmental origin and relationship to RORγt- regulatory T and Th17 cells remain unclear. In this study, we use a fixed TCRβ system to show that the TCR repertoire of the Foxp3+RORγt+ population is mostly distinct compared with other colonic T cell subsets. However, of these TCRs, a fraction is also found in the Th17 subset, suggesting that TCR repertoire overlap may contribute to the reported ability of Foxp3+RORγt+ cells to regulate Th17 immunity. Naive transgenic T cells expressing a Foxp3+RORγt+-restricted TCR first acquire a Foxp3+RORγt- phenotype before coexpressing RORγt, suggesting that Foxp3+RORγt+ cell development can occur via an RORγt- regulatory T cell intermediate.
Collapse
Affiliation(s)
- Benjamin D Solomon
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Chyi-Song Hsieh
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
316
|
Abstract
The immune system has evolved to defend the organism against an almost infinite number of pathogens in a locally confined and antigen-specific manner while at the same time preserving tolerance to harmless antigens and self. Regulatory T (Treg) cells essentially contribute to an immunoregulatory network preventing excessive immune responses and immunopathology. There is emerging evidence that Treg cells not only operate in secondary lymphoid tissue but also regulate immune responses directly at the site of inflammation. Hence, the classification of Treg cells might need to be further extended by Treg cell subsets that are functionally and phenotypically polarized by their residency. In this review, we discuss recent findings on these tissue-resident Treg cell subsets and how these cells may operate in a tissue- and context-dependent manner.
Collapse
|
317
|
Yang J, Xu L. Elevated IL-23R Expression and Foxp3+Rorgt+ Cells in Intestinal Mucosa During Acute and Chronic Colitis. Med Sci Monit 2016; 22:2785-92. [PMID: 27498708 PMCID: PMC4988365 DOI: 10.12659/msm.896827] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background IL-23/IL-23R signaling plays a pivotal role during the course of inflammatory bowel diseases (IBD). However, the underlying mechanisms are poorly characterized. Foxp3+ regulatory T cells are critical in the maintenance of gut immune homeostasis and therefore are important in preventing the development of IBD. This study was performed to clarify the association between IL-23/IL-23R signaling and Foxp3+ regulatory T cells in colitis. Material/Methods Acute and chronic mouse colitis models were established by administering mice DSS in drinking water. IL-23R, IL-23, IL-I7, and IFN-γ expression level, as well as regulatory T cell, Th17-, and Th1-related transcription factors Foxp3, RORγt, and T-bet were assayed by real-time PCR. The frequency of Foxp3+ RORγt+ cells in a Foxp3+ cell population in colon mucosa during acute and chronic colitis was evaluated through flow cytometry. The signaling pathway mediated by IL-23R in the colon mucosa from acute colitis mice and chronic colitis mice was monitored by Western blot analysis. Results We detected elevated IL-23R, IL-23, and IFN-γ expression in colon mucosa during acute and chronic colitis and found increased IL-17 in acute colitis mice. Transcription factors Foxp3 and T-bet were elevated in colon mucosa during acute and chronic colitis. Phosphorylation of Stat3 was greatly enhanced, indicating the activation of IL-23R function in colitis mice. The percentage of Foxp3+ T cells in acute and chronic colitis mice was comparable to control mice, but there was a 2-fold increase of Foxp3+ RORγt+ cells among the Foxp3+ cell population in acute and chronic colitis mice compared to control mice. Conclusions These findings indicate that the induction of Foxp3+ RORγt+ T cells could be enhanced during inflammation in the intestine where IL-23R expression is greatly induced. Our study highlights the importance of IL-23R expression level and the instability of Foxp3+ regulatory T cells in the development of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Jiayin Yang
- Division of Gastroenterology, Zhongshan hospital, Fudan University, Shanghai, China (mainland)
| | - Lili Xu
- Division of Gastroenterology, Zhongshan hospital, Fudan University, Shanghai, China (mainland)
| |
Collapse
|
318
|
Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature 2016; 535:75-84. [PMID: 27383982 DOI: 10.1038/nature18848] [Citation(s) in RCA: 1250] [Impact Index Per Article: 138.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/25/2016] [Indexed: 12/12/2022]
Abstract
In the mucosa, the immune system's T cells and B cells have position-specific phenotypes and functions that are influenced by the microbiota. These cells play pivotal parts in the maintenance of immune homeostasis by suppressing responses to harmless antigens and by enforcing the integrity of the barrier functions of the gut mucosa. Imbalances in the gut microbiota, known as dysbiosis, can trigger several immune disorders through the activity of T cells that are both near to and distant from the site of their induction. Elucidation of the mechanisms that distinguish between homeostatic and pathogenic microbiota-host interactions could identify therapeutic targets for preventing or modulating inflammatory diseases and for boosting the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Kenya Honda
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan.,RIKEN Center for Integrative Medical Sciences, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,AMED-CREST, Chiyoda, Tokyo 100-0004, Japan
| | - Dan R Littman
- The Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York 10016, USA.,The Howard Hughes Medical Institute, New York University School of Medicine, New York, New York 10016, USA
| |
Collapse
|
319
|
Microbiota, regulatory T cell subsets, and allergic disorders. ACTA ACUST UNITED AC 2016; 25:114-123. [PMID: 27656354 PMCID: PMC5016534 DOI: 10.1007/s40629-016-0118-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 01/26/2016] [Indexed: 02/07/2023]
|
320
|
Ohnmacht C. Tolerance to the Intestinal Microbiota Mediated by ROR(γt)(+) Cells. Trends Immunol 2016; 37:477-486. [PMID: 27255270 DOI: 10.1016/j.it.2016.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/21/2022]
Abstract
Harmless microbes colonizing the gut require the establishment of a well-equilibrated symbiosis between this microbiota and its host. However, the immune system is primed to recognize both conserved microbial patterns and foreign antigens, and therefore developed strong tolerance mechanisms to prevent potential fatal immune reactivity to symbiotic microbes. The transcription factor RAR-related orphan-like γt [ROR(γt); encoded by Rorc] plays a key role in the gut for lymphoid tissue organogenesis, development of innate lymphoid cells type 3 (ILC3s) and proinflammatory type 17 T helper (Th17) cells. Surprisingly, recent research has revealed a contribution of ROR(γt)-expressing cells in a variety of tolerance mechanisms in both the innate and adaptive immune system.
Collapse
Affiliation(s)
- Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technische Universität and Helmholtz Center Munich, Biedersteiner Strasse 29, 80802 Munich, Germany.
| |
Collapse
|
321
|
Abstract
The immune system is responsible for defending an organism against the myriad of microbial invaders it constantly confronts. It has become increasingly clear that the immune system has a second major function: the maintenance of organismal homeostasis. Foxp3(+)CD4(+) regulatory T cells (Tregs) are important contributors to both of these critical activities, defense being the primary purview of Tregs circulating through lymphoid organs, and homeostasis ensured mainly by their counterparts residing in parenchymal tissues. This review focuses on so-called tissue Tregs. We first survey existing information on the phenotype, function, sustaining factors, and human equivalents of the three best-characterized tissue-Treg populations-those operating in visceral adipose tissue, skeletal muscle, and the colonic lamina propria. We then attempt to distill general principles from this body of work-as concerns the provenance, local adaptation, molecular sustenance, and targets of action of tissue Tregs, in particular.
Collapse
Affiliation(s)
- Marisella Panduro
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02115; , ,
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, Massachusetts 02115
- Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Christophe Benoist
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02115; , ,
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, Massachusetts 02115
- Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Diane Mathis
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02115; , ,
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, Massachusetts 02115
- Brigham and Women's Hospital, Boston, Massachusetts 02115
| |
Collapse
|
322
|
Tanoue T, Atarashi K, Honda K. Development and maintenance of intestinal regulatory T cells. Nat Rev Immunol 2016; 16:295-309. [PMID: 27087661 DOI: 10.1038/nri.2016.36] [Citation(s) in RCA: 415] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gut-resident forkhead box P3 (FOXP3)(+)CD4(+) regulatory T cells (Treg cells) are distinct from those in other organs and have gut-specific phenotypes and functions. Whereas Treg cells in other organs have T cell receptors (TCRs) specific for self antigens, intestinal Treg cells have a distinct set of TCRs that are specific for intestinal antigens, and these cells have pivotal roles in the suppression of immune responses against harmless dietary antigens and commensal microorganisms. The differentiation, migration and maintenance of intestinal Treg cells are controlled by specific signals from the local environment. In particular, certain members of the microbiota continuously provide antigens and immunoregulatory small molecules that modulate intestinal Treg cells. Understanding the development and the maintenance of intestinal Treg cells provides important insights into disease-relevant host-microorganism interactions.
Collapse
Affiliation(s)
- Takeshi Tanoue
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan
| | - Koji Atarashi
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kenya Honda
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| |
Collapse
|