151
|
Zhao Q, Elson CO. Adaptive immune education by gut microbiota antigens. Immunology 2018; 154:28-37. [PMID: 29338074 PMCID: PMC5904715 DOI: 10.1111/imm.12896] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 12/13/2022] Open
Abstract
Host-microbiota mutualism has been established during long-term co-evolution. A diverse and rich gut microbiota plays an essential role in the development and maturation of the host immune system. Education of the adaptive immune compartment by gut microbiota antigens is important in establishing immune balance. In particular, a critical time frame immediately after birth provides a 'window of opportunity' for the development of lymphoid structures, differentiation and maturation of T and B cells and, most importantly, establishment of immune tolerance to gut commensals. Depending on the colonization niche, antigen type and metabolic property of different gut microbes, CD4 T-cell responses vary greatly, which results in differentiation into distinct subsets. As a consequence, certain bacteria elicit effector-like immune responses by promoting the production of pro-inflammatory cytokines such as interferon-γ and interleukin-17A, whereas other bacteria favour the generation of regulatory CD4 T cells and provide help with gut homeostasis. The microbiota have profound effects on B cells also. Gut microbial exposure leads to a continuous diversification of B-cell repertoire and the production of T-dependent and -independent antibodies, especially IgA. These combined effects of the gut microbes provide an elegant educational process to the adaptive immune network. Contrariwise, failure of this process results in a reduced homeostasis with the gut microbiota, and an increased susceptibility to various immune disorders, both inside and outside the gut. With more definitive microbial-immune relations waiting to be discovered, modulation of the host gut microbiota has a promising future for disease intervention.
Collapse
Affiliation(s)
- Qing Zhao
- Department of MedicineThe University of Alabama at BirminghamBirminghamALUSA
| | - Charles O. Elson
- Department of MedicineThe University of Alabama at BirminghamBirminghamALUSA
| |
Collapse
|
152
|
Linehan JL, Harrison OJ, Han SJ, Byrd AL, Vujkovic-Cvijin I, Villarino AV, Sen SK, Shaik J, Smelkinson M, Tamoutounour S, Collins N, Bouladoux N, Dzutsev A, Rosshart SP, Arbuckle JH, Wang CR, Kristie TM, Rehermann B, Trinchieri G, Brenchley JM, O'Shea JJ, Belkaid Y. Non-classical Immunity Controls Microbiota Impact on Skin Immunity and Tissue Repair. Cell 2018; 172:784-796.e18. [PMID: 29358051 DOI: 10.1016/j.cell.2017.12.033] [Citation(s) in RCA: 325] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 10/17/2017] [Accepted: 12/21/2017] [Indexed: 02/02/2023]
Abstract
Mammalian barrier surfaces are constitutively colonized by numerous microorganisms. We explored how the microbiota was sensed by the immune system and the defining properties of such responses. Here, we show that a skin commensal can induce T cell responses in a manner that is restricted to non-classical MHC class I molecules. These responses are uncoupled from inflammation and highly distinct from pathogen-induced cells. Commensal-specific T cells express a defined gene signature that is characterized by expression of effector genes together with immunoregulatory and tissue-repair signatures. As such, non-classical MHCI-restricted commensal-specific immune responses not only promoted protection to pathogens, but also accelerated skin wound closure. Thus, the microbiota can induce a highly physiological and pleiotropic form of adaptive immunity that couples antimicrobial function with tissue repair. Our work also reveals that non-classical MHC class I molecules, an evolutionarily ancient arm of the immune system, can promote homeostatic immunity to the microbiota.
Collapse
Affiliation(s)
- Jonathan L Linehan
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Oliver J Harrison
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Seong-Ji Han
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Allyson L Byrd
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA; Translational and Functional Genomics Branch, NHGRI, NIH, Bethesda, MD 20892, USA; Department of Bioinformatics, Boston University, Boston, MA 02215, USA
| | - Ivan Vujkovic-Cvijin
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - Shurjo K Sen
- Cancer and Inflammation Program, NCI, NIH, Bethesda, MD 20892, USA
| | - Jahangheer Shaik
- Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Margery Smelkinson
- Biological Imaging, Research Technology Branch, NIAID, NIH, Bethesda, MD 20892, USA
| | - Samira Tamoutounour
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Nicholas Collins
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Nicolas Bouladoux
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA; NIAID Microbiome Program, NIH, Bethesda, MD 20892, USA
| | - Amiran Dzutsev
- Cancer and Inflammation Program, NCI, NIH, Bethesda, MD 20892, USA
| | - Stephan P Rosshart
- Immunology Section, Liver Diseases Branch, NIDDK, NIH, Bethesda, MD 20892, USA
| | | | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL 60611, USA
| | | | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, NIDDK, NIH, Bethesda, MD 20892, USA
| | | | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, NIAMS, NIH, Bethesda, MD 20892, USA
| | - Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
153
|
Abstract
Neuromyelitis optica (NMO) is a rare, disabling, sometimes fatal central nervous system inflammatory demyelinating disease that is associated with antibodies ("NMO IgG") that target the water channel protein aquaporin-4 (AQP4) expressed on astrocytes. There is considerable interest in identifying environmental triggers that may elicit production of NMO IgG by AQP4-reactive B cells. Although NMO is considered principally a humoral autoimmune disease, antibodies of NMO IgG are IgG1, a T-cell-dependent immunoglobulin subclass, indicating that AQP4-reactive T cells have a pivotal role in NMO pathogenesis. When AQP4-specific proliferative T cells were first identified in patients with NMO it was discovered that T cells recognizing the dominant AQP4 T-cell epitope exhibited a T helper 17 (Th17) phenotype and displayed cross-reactivity to a homologous peptide sequence within a protein of Clostridium perfringens, a commensal bacterium found in human gut flora. The initial analysis of gut microbiota in NMO demonstrated that, in comparison to healthy controls (HC) and patients with multiple sclerosis, the microbiome of NMO is distinct. Remarkably, C. perfringens was the second most significantly enriched taxon in NMO, and among bacteria identified at the species level, C. perfringens was the one most highly associated with NMO. Those discoveries, along with evidence that certain Clostridia in the gut can regulate the balance between regulatory T cells and Th17 cells, indicate that gut microbiota, and possibly C. perfringens itself, could participate in NMO pathogenesis. Collectively, the evidence linking microbiota to humoral and cellular immunity in NMO underscores the importance for further investigating this relationship.
Collapse
Affiliation(s)
- Scott S Zamvil
- Department of Neurology, University of California, San Francisco, CA, USA.
- Program in Immunology, University of California, San Francisco, CA, USA.
| | - Collin M Spencer
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Sergio E Baranzini
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Bruce A C Cree
- Department of Neurology, University of California, San Francisco, CA, USA
| |
Collapse
|
154
|
Knoop KA, Gustafsson JK, McDonald KG, Kulkarni DH, Coughlin PE, McCrate S, Kim D, Hsieh CS, Hogan SP, Elson CO, Tarr PI, Newberry RD. Microbial antigen encounter during a preweaning interval is critical for tolerance to gut bacteria. Sci Immunol 2017; 2:eaao1314. [PMID: 29246946 PMCID: PMC5759965 DOI: 10.1126/sciimmunol.aao1314] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/06/2017] [Indexed: 12/28/2022]
Abstract
We have a mutually beneficial relationship with the trillions of microorganisms inhabiting our gastrointestinal tract. However, maintaining this relationship requires recognizing these organisms as affable and restraining inflammatory responses to these organisms when encountered in hostile settings. How and when the immune system develops tolerance to our gut microbial members is not well understood. We identify a specific preweaning interval in which gut microbial antigens are encountered by the immune system to induce antigen-specific tolerance to gut bacteria. For some bacterial taxa, physiologic encounters with the immune system are restricted to this interval, despite abundance of these taxa in the gut lumen at later times outside this interval. Antigen-specific tolerance to gut bacteria induced during this preweaning interval is stable and maintained even if these taxa are encountered later in life in an inflammatory setting. However, inhibiting microbial antigen encounter during this interval or extending these encounters beyond the normal interval results in a failure to induce tolerance and robust antigen-specific effector responses to gut bacteria upon reencounter in an inflammatory setting. Thus, we have identified a defined preweaning interval critical for developing tolerance to gut bacteria and maintaining the mutually beneficial relationship with our gut microbiota.
Collapse
Affiliation(s)
- Kathryn A Knoop
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jenny K Gustafsson
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Keely G McDonald
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Devesha H Kulkarni
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Paige E Coughlin
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Stephanie McCrate
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dongyeon Kim
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chyi-Song Hsieh
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Simon P Hogan
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Charles O Elson
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Phillip I Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rodney D Newberry
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
155
|
Sharanova NE, Ninnemann J, Bondareva MA, Semin YK, Nomokonova AV, Kruglov AA. Analysis of the Specificity of IgA Antibodies Produced in the Mouse Small Intestine. Mol Biol 2017. [DOI: 10.1134/s0026893317060152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
156
|
Antigen-specific regulatory T-cell responses to intestinal microbiota. Mucosal Immunol 2017; 10:1375-1386. [PMID: 28766556 PMCID: PMC5939566 DOI: 10.1038/mi.2017.65] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 06/07/2017] [Indexed: 02/07/2023]
Abstract
The mammalian gastrointestinal tract can harbor both beneficial commensal bacteria important for host health, but also pathogenic bacteria capable of intestinal damage. It is therefore important that the host immune system mount the appropriate immune response to these divergent groups of bacteria-promoting tolerance in response to commensal bacteria and sterilizing immunity in response to pathogenic bacteria. Failure to induce tolerance to commensal bacteria may underlie immune-mediated diseases such as human inflammatory bowel disease. At homeostasis, regulatory T (Treg) cells are a key component of the tolerogenic response by adaptive immunity. This review examines the mechanisms by which intestinal bacteria influence colonic T-cells and B-cell immunoglobulin A (IgA) induction, with an emphasis on Treg cells and the role of antigen-specificity in these processes. In addition to discussing key primary literature, this review highlights current controversies and important future directions.
Collapse
|
157
|
Lycke NY, Bemark M. The regulation of gut mucosal IgA B-cell responses: recent developments. Mucosal Immunol 2017; 10:1361-1374. [PMID: 28745325 DOI: 10.1038/mi.2017.62] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/20/2017] [Indexed: 02/04/2023]
Abstract
The majority of activated B cells differentiate into IgA plasma cells, with the gut being the largest producer of immunoglobulin in the body. Secretory IgA antibodies have numerous critical functions of which protection against infections and the role for establishing a healthy microbiota appear most important. Expanding our knowledge of the regulation of IgA B-cell responses and how effective mucosal vaccines can be designed are of critical importance. Here we discuss recent developments in the field that shed light on the uniqueness and complexity of mucosal IgA responses and the control of protective IgA responses in the gut, specifically.
Collapse
Affiliation(s)
- N Y Lycke
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - M Bemark
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
158
|
A Gut Microbial Mimic that Hijacks Diabetogenic Autoreactivity to Suppress Colitis. Cell 2017; 171:655-667.e17. [DOI: 10.1016/j.cell.2017.09.022] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 08/10/2017] [Accepted: 09/15/2017] [Indexed: 12/24/2022]
|
159
|
Fay KT, Ford ML, Coopersmith CM. The intestinal microenvironment in sepsis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2574-2583. [PMID: 28286161 PMCID: PMC5589488 DOI: 10.1016/j.bbadis.2017.03.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/16/2017] [Accepted: 03/06/2017] [Indexed: 12/17/2022]
Abstract
The gastrointestinal tract has long been hypothesized to function as "the motor" of multiple organ dysfunction syndrome. The gastrointestinal microenvironment is comprised of a single cell layer epithelia, a local immune system, and the microbiome. These three components of the intestine together play a crucial role in maintaining homeostasis during times of health. However, the gastrointestinal microenvironment is perturbed during sepsis, resulting in pathologic changes that drive both local and distant injury. In this review, we seek to characterize the relationship between the epithelium, gastrointestinal lymphocytes, and commensal bacteria during basal and pathologic conditions and how the intestinal microenvironment may be targeted for therapeutic gain in septic patients.
Collapse
Affiliation(s)
- Katherine T Fay
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Mandy L Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States; Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Craig M Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States; Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States.
| |
Collapse
|
160
|
Jung MK, Kwak JE, Shin EC. IL-17A-Producing Foxp3 + Regulatory T Cells and Human Diseases. Immune Netw 2017; 17:276-286. [PMID: 29093649 PMCID: PMC5662777 DOI: 10.4110/in.2017.17.5.276] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/29/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
CD4+Foxp3+ regulatory T (Treg) cells play major roles in immune homeostasis. While CD4+Foxp3+ Treg cells act to suppress other immune effector cells, there is growing evidence that they also produce pro-inflammatory cytokines, such as IL-17A, in inflammatory conditions. The pro-inflammatory cytokine milieu, toll-like receptor (TLR) signaling, and specific transcription factors are important for the production of IL-17A by CD4+Foxp3+ Treg cells. In particular, IL-17A-producing CD4+Foxp3+ Treg cells express RORγt, the T helper (Th) 17-specific transcription factor, in addition to Foxp3. IL-17A-producing CD4+Foxp3+ Treg cells are also involved in the pathogenesis of various diseases. Here we review the mechanisms underlying the induction of IL-17A-producing CD4+Foxp3+ Treg cells and the roles of these cells in human disease.
Collapse
Affiliation(s)
- Min Kyung Jung
- Laboratory of Immunology & Infectious Diseases, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Korea
| | - Jeong-Eun Kwak
- BioMedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141, Korea
| | - Eui-Cheol Shin
- Laboratory of Immunology & Infectious Diseases, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Korea
- BioMedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141, Korea
| |
Collapse
|
161
|
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: 339] [Impact Index Per Article: 42.4] [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
|
162
|
Mei HE, Hahne S, Redlin A, Hoyer BF, Wu K, Baganz L, Lisney AR, Alexander T, Rudolph B, Dörner T. Plasmablasts With a Mucosal Phenotype Contribute to Plasmacytosis in Systemic Lupus Erythematosus. Arthritis Rheumatol 2017. [DOI: 10.1002/art.40181] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Henrik E. Mei
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Stefanie Hahne
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Andreas Redlin
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Bimba F. Hoyer
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Kaiyin Wu
- Charité University Medicine Berlin; Berlin Germany
| | - Lisa Baganz
- German Rheumatism Research Center Berlin; Berlin Germany
| | - Anna R. Lisney
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | - Tobias Alexander
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| | | | - Thomas Dörner
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin; Berlin Germany
| |
Collapse
|
163
|
McCoy KD, Ronchi F, Geuking MB. Host-microbiota interactions and adaptive immunity. Immunol Rev 2017; 279:63-69. [DOI: 10.1111/imr.12575] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Kathy D. McCoy
- Department of Physiology and Pharmacology; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases; Cumming School of Medicine; University of Calgary; Calgary AB Canada
| | - Francesca Ronchi
- Maurice Müller Laboratories; Department of Clinical Research (DKF); UVCM; University Hospital; Bern Switzerland
| | - Markus B. Geuking
- Department of Microbiology, Immunology and Infectious Diseases; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases; Cumming School of Medicine; University of Calgary; Calgary AB Canada
| |
Collapse
|
164
|
Weingartner E, Golding A. Direct control of B cells by Tregs: An opportunity for long-term modulation of the humoral response. Cell Immunol 2017; 318:8-16. [DOI: 10.1016/j.cellimm.2017.05.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/20/2017] [Accepted: 05/28/2017] [Indexed: 12/23/2022]
|
165
|
Alexander KL, Katz J, Elson CO. CBirTox is a selective antigen-specific agonist of the Treg-IgA-microbiota homeostatic pathway. PLoS One 2017; 12:e0181866. [PMID: 28750075 PMCID: PMC5531474 DOI: 10.1371/journal.pone.0181866] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 07/07/2017] [Indexed: 01/16/2023] Open
Abstract
Cultivating an environment of mutualism between host cells and the microbiota is vital, and dysregulation of this relationship is associated with multiple immune disorders including metabolic and skin diseases, asthma, allergy, and Inflammatory Bowel Disease (IBD). One prominent mechanism for maintaining homeostasis is the protective regulatory T cell (Treg)- Immunoglobulin A (IgA) pathway toward microbiota antigens, in which Tregs maintain homeostasis and provide critical survival factors to IgA+ B cells. In order to amplify the Treg-IgA pathway, we have generated a fusion protein, CBirTox, comprised of a portion of the carboxy terminus of CBir1, a microbiota flagellin, genetically coupled to Cholera Toxin B subunit (CTB) via the A2 linker of CT. Both dendritic cells (DCs) and B cells pulsed with CBirTox selectively induced functional CD4+Foxp3+ Tregs in vitro, and CBirTox augmented CD4+Foxp3+ cell numbers in vivo. The induced Foxp3 expression was independent of retinoic acid (RA) signaling but was inhibited by neutralization of TGF-β. CBirTox treatment of B cells downregulated mammalian target of rapamycin (mTOR) signaling. Furthermore, CBirTox-pulsed DCs induced substantial production of IgA from naïve B cells. Collectively these data demonstrate that CBirTox represents a novel approach to bolstering the Treg-IgA pathway at the host-microbiota interface.
Collapse
Affiliation(s)
- Katie L. Alexander
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Jannet Katz
- Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Charles O. Elson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- * E-mail:
| |
Collapse
|
166
|
Chai JN, Peng Y, Rengarajan S, Solomon BD, Ai TL, Shen Z, Perry JSA, Knoop KA, Tanoue T, Narushima S, Honda K, Elson CO, Newberry RD, Stappenbeck TS, Kau AL, Peterson DA, Fox JG, Hsieh CS. Helicobacter species are potent drivers of colonic T cell responses in homeostasis and inflammation. Sci Immunol 2017; 2:2/13/eaal5068. [PMID: 28733471 DOI: 10.1126/sciimmunol.aal5068] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 06/02/2017] [Indexed: 12/16/2022]
Abstract
Specific gut commensal bacteria improve host health by eliciting mutualistic regulatory T (Treg) cell responses. However, the bacteria that induce effector T (Teff) cells during inflammation are unclear. We addressed this by analyzing bacterial-reactive T cell receptor (TCR) transgenic cells and TCR repertoires in a murine colitis model. Unexpectedly, we found that mucosal-associated Helicobacter species triggered both Treg cell responses during homeostasis and Teff cell responses during colitis, as suggested by an increased overlap between the Teff/Treg TCR repertoires with colitis. Four of six Treg TCRs tested recognized mucosal-associated Helicobacter species in vitro and in vivo. By contrast, the marked expansion of luminal Bacteroides species seen during colitis did not trigger a commensurate Teff cell response. Unlike other Treg cell-inducing bacteria, Helicobacter species are known pathobionts and cause disease in immunodeficient mice. Thus, our study suggests a model in which mucosal bacteria elicit context-dependent Treg or Teff cell responses to facilitate intestinal tolerance or inflammation.
Collapse
Affiliation(s)
- Jiani N Chai
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yangqing Peng
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sunaina Rengarajan
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Benjamin D Solomon
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Teresa L Ai
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zeli Shen
- Division of Comparative Medicine, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Justin S A Perry
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kathryn A Knoop
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Takeshi Tanoue
- RIKEN Center for Integrative Medical Sciences, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Seiko Narushima
- RIKEN Center for Integrative Medical Sciences, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Kenya Honda
- RIKEN Center for Integrative Medical Sciences, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Charles O Elson
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Rodney D Newberry
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thaddeus S Stappenbeck
- Department of Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew L Kau
- Center for Women's Infectious Disease Research and Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - James G Fox
- Division of Comparative Medicine, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chyi-Song Hsieh
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
167
|
Zhao Z, Liang Y, Liu Y, Xu P, Flamme-Wiese MJ, Sun D, Sun J, Mullins RF, Chen Y, Cai J. Choroidal γδ T cells in protection against retinal pigment epithelium and retinal injury. FASEB J 2017; 31:4903-4916. [PMID: 28729290 DOI: 10.1096/fj.201700533r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 07/05/2017] [Indexed: 12/17/2022]
Abstract
γδ T cells located near the epithelial barrier are integral components of local inflammatory and innate immune responses. We have previously reported the presence of choroidal γδ T cells in a model of chronic degeneration of the retinal pigment epithelium (RPE). The goals of the current study were to further define the functions of choroidal γδ T cells and to explore the underlying mechanisms of their action. Our data demonstrate that choroidal γδ T cells are activated by RPE injury in response to NaIO3 treatment, and that they express genes that encode immunosuppressive cytokines, such as IL-4 and IL-10. γδ-T-cell-deficient mice developed profound RPE and retinal damage at doses that caused minimal effects in wild-type mice, and adoptive transfer of γδ T cells prevented sensitization. Intravitreal injection of IL-4 and IL-10 ameliorated RPE toxicity that was induced by NaIO3Ex vivo coculture of γδ T cells with RPE explants activated the production of anti-inflammatory cytokines via an aryl hydrocarbon receptor (AhR)-dependent mechanism. AhR deficiency abolished the protective effects of γδ T cells after adoptive transfer. Collectively, these findings define important roles for choroid γδ T cells in maintaining tissue homeostasis in the outer retina.-Zhao, Z., Liang, Y., Liu, Y., Xu, P., Flamme-Wiese, M. J., Sun, D., Sun, J., Mullins, R. F., Chen, Y., Cai, J. Choroidal γδ T cells in protection against retinal pigment epithelium and retinal injury.
Collapse
Affiliation(s)
- Zhenyang Zhao
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yin Liu
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Pei Xu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Miles J Flamme-Wiese
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, Iowa, USA
| | - Deming Sun
- Doheny Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Robert F Mullins
- Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, Iowa, USA
| | - Yan Chen
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jiyang Cai
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA;
| |
Collapse
|
168
|
Wu W, Sun M, Chen F, Cao AT, Liu H, Zhao Y, Huang X, Xiao Y, Yao S, Zhao Q, Liu Z, Cong Y. Microbiota metabolite short-chain fatty acid acetate promotes intestinal IgA response to microbiota which is mediated by GPR43. Mucosal Immunol 2017; 10:946-956. [PMID: 27966553 PMCID: PMC5471141 DOI: 10.1038/mi.2016.114] [Citation(s) in RCA: 335] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 10/28/2016] [Indexed: 02/04/2023]
Abstract
Intestinal IgA, which is regulated by gut microbiota, has a crucial role in maintenance of intestinal homeostasis and in protecting the intestines from inflammation. However, the means by which microbiota promotes intestinal IgA responses remain unclear. Emerging evidence suggests that the host can sense gut bacterial metabolites in addition to pathogen-associated molecular patterns and that recognition of these small molecules influences host immune response in the intestines and beyond. We reported here that microbiota metabolite short-chain fatty acid acetate promoted intestinal IgA responses, which was mediated by "metabolite-sensing" GPR43. GPR43-/- mice demonstrated lower levels of intestinal IgA and IgA+ gut bacteria compared with those in wild type (WT) mice. Feeding WT but not GPR43-/- mice acetate but not butyrate promoted intestinal IgA response independent of T cells. Acetate promoted B-cell IgA class switching and IgA production in vitro in the presence of WT but not GPR43-/- dendritic cells (DCs). Mechanistically, acetate-induced DC expression of Aldh1a2, which converts Vitamin A into its metabolite retinoic acid (RA). Moreover, blockade of RA signaling inhibited the acetate induction of B-cell IgA production. Our studies thus identified a new pathway by which microbiota promotes intestinal IgA response through its metabolites.
Collapse
Affiliation(s)
- Wei Wu
- Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Mingming Sun
- Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Feidi Chen
- Department of Pathology, University of Texas Medical Branch, Galveston, TX
| | - Anthony T Cao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Han Liu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Ye Zhao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Xiangsheng Huang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Yi Xiao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | | | - Zhanju Liu
- Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China,Corresponding authors: Yingzi Cong, Ph.D., Department of Microbiology and Immunology, University of Texas Medical Branch, 4.142C Medical Research Building, 301 University Blvd, Galveston, TX 77555-1019. Phone: (409) 772-4902. Fax: (409) 772-5065. or Dr. Zhanju Liu, Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China; ; fax: (86) 21-6630-3983
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX,Department of Pathology, University of Texas Medical Branch, Galveston, TX,Corresponding authors: Yingzi Cong, Ph.D., Department of Microbiology and Immunology, University of Texas Medical Branch, 4.142C Medical Research Building, 301 University Blvd, Galveston, TX 77555-1019. Phone: (409) 772-4902. Fax: (409) 772-5065. or Dr. Zhanju Liu, Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China; ; fax: (86) 21-6630-3983
| |
Collapse
|
169
|
Larsen JM. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology 2017; 151:363-374. [PMID: 28542929 DOI: 10.1111/imm.12760] [Citation(s) in RCA: 857] [Impact Index Per Article: 107.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/10/2017] [Accepted: 05/15/2017] [Indexed: 02/07/2023] Open
Abstract
The microbiota plays a central role in human health and disease by shaping immune development, immune responses and metabolism, and by protecting from invading pathogens. Technical advances that allow comprehensive characterization of microbial communities by genetic sequencing have sparked the hunt for disease-modulating bacteria. Emerging studies in humans have linked the increased abundance of Prevotella species at mucosal sites to localized and systemic disease, including periodontitis, bacterial vaginosis, rheumatoid arthritis, metabolic disorders and low-grade systemic inflammation. Intriguingly, Prevotella abundance is reduced within the lung microbiota of patients with asthma and chronic obstructive pulmonary disease. Increased Prevotella abundance is associated with augmented T helper type 17 (Th17) -mediated mucosal inflammation, which is in line with the marked capacity of Prevotella in driving Th17 immune responses in vitro. Studies indicate that Prevotella predominantly activate Toll-like receptor 2, leading to production of Th17-polarizing cytokines by antigen-presenting cells, including interleukin-23 (IL-23) and IL-1. Furthermore, Prevotella stimulate epithelial cells to produce IL-8, IL-6 and CCL20, which can promote mucosal Th17 immune responses and neutrophil recruitment. Prevotella-mediated mucosal inflammation leads to systemic dissemination of inflammatory mediators, bacteria and bacterial products, which in turn may affect systemic disease outcomes. Studies in mice support a causal role of Prevotella as colonization experiments promote clinical and inflammatory features of human disease. When compared with strict commensal bacteria, Prevotella exhibit increased inflammatory properties, as demonstrated by augmented release of inflammatory mediators from immune cells and various stromal cells. These findings indicate that some Prevotella strains may be clinically important pathobionts that can participate in human disease by promoting chronic inflammation.
Collapse
Affiliation(s)
- Jeppe Madura Larsen
- Department of Technology, Faculty of Health and Technology, Metropolitan University College, Copenhagen, Denmark.,National Food Institute, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
170
|
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: 796] [Impact Index Per Article: 99.5] [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
|
171
|
Zhao Q, Harbour SN, Kolde R, Latorre IJ, Tun HM, Schoeb TR, Turner H, Moon JJ, Khafipour E, Xavier RJ, Weaver CT, Elson CO. Selective Induction of Homeostatic Th17 Cells in the Murine Intestine by Cholera Toxin Interacting with the Microbiota. THE JOURNAL OF IMMUNOLOGY 2017; 199:312-322. [PMID: 28539431 DOI: 10.4049/jimmunol.1700171] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023]
Abstract
Th17 cells play a role as an inflammation mediator in a variety of autoimmune disorders, including inflammatory bowel disease, and thus are widely considered to be pathogenic. However, Th17 cells are present in the normal intestine and show a homeostatic phenotype; that is, they participate in the maintenance of intestinal homeostasis rather than inducing inflammation. We observed an enlarged Th17 population in the small intestine of C57BL/6.IgA-/- mice compared with wild-type mice, which was further amplified with cholera toxin (CT) immunization without causing intestinal inflammation. The increased Th17 induction and the correspondingly 10-fold higher CT B subunit-specific serum IgG response in IgA-/- mice after CT immunization was microbiota dependent and was associated with increased segmented filamentous bacteria in the small intestine of IgA-/- mice. Oral administration of vancomycin greatly dampened both CT immunogenicity and adjuvanticity, and the differential CT responses in IgA-/- and wild-type mice disappeared after intestinal microbiota equalization. Using gnotobiotic mouse models, we found that CT induction of homeostatic intestinal Th17 responses was supported not only by segmented filamentous bacteria, but also by other commensal bacteria. Furthermore, transcriptome analysis using IL-17AhCD2 reporter mice revealed a similar gene expression profile in CT-induced intestinal Th17 cells and endogenous intestinal Th17 cells at homeostasis, with upregulated expression of a panel of immune-regulatory genes, which was distinctly different from the gene expression profile of pathogenic Th17 cells. Taken together, we identified a nonpathogenic signature of intestinal homeostatic Th17 cells, which are actively regulated by the commensal microbiota and can be selectively stimulated by CT.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Stacey N Harbour
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Raivo Kolde
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | | | - Hein M Tun
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Trenton R Schoeb
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Henrietta Turner
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - James J Moon
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Ehsan Khafipour
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Ramnik J Xavier
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114.,Broad Institute of MIT and Harvard, Cambridge, MA 02142.,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114; and
| | - Casey T Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294.,Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Charles O Elson
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294; .,Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
172
|
Oh-oka K, Kojima Y, Uchida K, Yoda K, Ishimaru K, Nakajima S, Hemmi J, Kano H, Fujii-Kuriyama Y, Katoh R, Ito H, Nakao A. Induction of Colonic Regulatory T Cells by Mesalamine by Activating the Aryl Hydrocarbon Receptor. Cell Mol Gastroenterol Hepatol 2017; 4:135-151. [PMID: 28593185 PMCID: PMC5453907 DOI: 10.1016/j.jcmgh.2017.03.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 03/31/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Mesalamine is a first-line drug for treatment of inflammatory bowel diseases (IBD). However, its mechanisms are not fully understood. CD4+ Foxp3+ regulatory T cells (Tregs) play a potential role in suppressing IBD. This study determined whether the anti-inflammatory activity of mesalamine is related to Treg induction in the colon. METHODS We examined the frequencies of Tregs in the colons of wild-type mice, mice deficient for aryl hydrocarbon receptor (AhR-/- mice), and bone marrow-chimeric mice lacking AhR in hematopoietic cells (BM-AhR-/- mice), following oral treatment with mesalamine. We also examined the effects of mesalamine on transforming growth factor (TGF)-β expression in the colon. RESULTS Treatment of wild-type mice with mesalamine increased the accumulation of Tregs in the colon and up-regulated the AhR target gene Cyp1A1, but this effect was not observed in AhR-/- or BM-AhR-/- mice. In addition, mesalamine promoted in vitro differentiation of naive T cells to Tregs, concomitant with AhR activation. Mice treated with mesalamine exhibited increased levels of the active form of TGF-β in the colon in an AhR-dependent manner and blockade of TGF-β signaling suppressed induction of Tregs by mesalamine in the colon. Furthermore, mice pretreated with mesalamine acquired resistance to dextran sodium sulfate-induced colitis. CONCLUSIONS We propose a novel anti-inflammatory mechanism of mesalamine for colitis: induction of Tregs in the colon via the AhR pathway, followed by TGF-β activation.
Collapse
Key Words
- AhR, aryl hydrocarbon receptor
- Aryl Hydrocarbon Receptor
- BM, bone marrow
- DSS, dextran sodium sulfate
- ELISA, enzyme-linked immunosorbent assay
- FBS, fetal bovine serum
- FITC, fluorescein isothiocyanate
- IBD, inflammatory bowel disease
- IFN, interferon
- IL, interleukin
- LPL, lamina propria lymphocytes
- MLN, mesenteric lymph nodes
- Mesalamine
- PBS, phosphate-buffered saline
- Q-PCR, quantitative polymerase chain reaction
- RPMI, Roswell Park Memorial Institute
- Regulatory T Cells
- TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin
- TGF, transforming growth factor
- TGF-β
- TNF, tumor necrosis factor
- Tregs, regulatory T cells
- WT, wild-type
- XRE, xenobiotic responsive element
- mAb, monoclonal antibody
Collapse
Affiliation(s)
- Kyoko Oh-oka
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yuko Kojima
- The Laboratory of Morphology and Image Analysis, Research Support Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Koichiro Uchida
- Atopy Research Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Kimiko Yoda
- Department of Pathology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kayoko Ishimaru
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Shotaro Nakajima
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Jun Hemmi
- Food Science Research Laboratories, Division of Research and Development, Meiji Co, Ltd, Kanagawa, Japan
| | - Hiroshi Kano
- Food Science Research Laboratories, Division of Research and Development, Meiji Co, Ltd, Kanagawa, Japan
| | | | - Ryohei Katoh
- Department of Pathology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Hiroyuki Ito
- Food Science Research Laboratories, Division of Research and Development, Meiji Co, Ltd, Kanagawa, Japan
| | - Atsuhito Nakao
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan,Atopy Research Center, Juntendo University School of Medicine, Tokyo, Japan,Correspondence Address correspondence to: Atsuhito Nakao, MD, PhD, Department of Immunology, Faculty of Medicine, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan. fax: 81-55-273-9542.Department of ImmunologyFaculty of MedicineUniversity of Yamanashi1110, ShimokatoChuoYamanashi 409-3898Japan
| |
Collapse
|
173
|
Kelly A, Houston SA, Sherwood E, Casulli J, Travis MA. Regulation of Innate and Adaptive Immunity by TGFβ. Adv Immunol 2017; 134:137-233. [PMID: 28413021 DOI: 10.1016/bs.ai.2017.01.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Immune regulation by cytokines is crucial in maintaining immune homeostasis, promoting responses to infection, resolving inflammation, and promoting immunological memory. Additionally, cytokine responses drive pathology in immune-mediated disease. A crucial cytokine in the regulation of all aspects of an immune response is transforming growth factor beta (TGFβ). Although best known as a crucial regulator of T cell responses, TGFβ plays a vital role in regulating responses mediated by virtually every innate and adaptive immune cell, including dendritic cells, B cells, NK cells, innate lymphoid cells, and granulocytes. Here, we review our current knowledge of how TGFβ regulates the immune system, highlighting the multifunctional nature of TGFβ and how its function can change depending on location and context of action.
Collapse
Affiliation(s)
- Aoife Kelly
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom; Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom; Manchester Immunology Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Stephanie A Houston
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom; Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom; Manchester Immunology Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Eleanor Sherwood
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom; Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom; Manchester Immunology Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Joshua Casulli
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom; Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom; Manchester Immunology Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Mark A Travis
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom; Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom; Manchester Immunology Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
174
|
Kim DH, Cheon JH. Pathogenesis of Inflammatory Bowel Disease and Recent Advances in Biologic Therapies. Immune Netw 2017; 17:25-40. [PMID: 28261018 PMCID: PMC5334120 DOI: 10.4110/in.2017.17.1.25] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/24/2017] [Accepted: 02/07/2017] [Indexed: 02/08/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory disorder with an unknown etiology. IBD is composed of two different disease entities: Crohn's disease (CD) and ulcerative colitis (UC). IBD has been thought to be idiopathic but has two main attributable causes that include genetic and environmental factors. The gastrointestinal tract in which this disease occurs is central to the immune system, and the innate and the adaptive immune systems are balanced in complex interactions with intestinal microbes under homeostatic conditions. However, in IBD, this homeostasis is disrupted and uncontrolled intestinal inflammation is perpetuated. Recently, the pathogenesis of IBD has become better understood owing to advances in genetic and immunologic technology. Moreover, new therapeutic strategies are now being implemented that accurately target the pathogenesis of IBD. Beyond conventional immunesuppressive therapy, the development of biological agents that target specific disease mechanisms has resulted in more frequent and deeper remission in IBD patients, with mucosal healing as a treatment goal of therapy. Future novel biologics should overcome the limitations of current therapies and ensure that individual patients can be treated with optimal drugs that are safe and precisely target IBD.
Collapse
Affiliation(s)
- Duk Hwan Kim
- Digestive Disease Center, CHA Bundang Hospital, CHA University, Seongnam 13496, Korea
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul 03722, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
175
|
Immunization with Salmonella Enteritidis secreting mucosal adjuvant labile toxin confers protection against wild type challenge via augmentation of CD3 + CD4 + T-cell proliferation and enhancement of IFN-γ, IL-6 and IL-10 expressions in chicken. Vaccine 2017; 35:767-773. [DOI: 10.1016/j.vaccine.2016.12.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 12/16/2016] [Accepted: 12/19/2016] [Indexed: 01/06/2023]
|
176
|
An expanding stage for commensal microbes in host immune regulation. Cell Mol Immunol 2017; 14:339-348. [PMID: 28065939 DOI: 10.1038/cmi.2016.64] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/12/2016] [Accepted: 10/12/2016] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal commensal microbiota is a concentrated mix of microbial life forms, including bacteria, fungi, archaea and viruses. These life forms are targets of host antimicrobial defense in order to establish a homeostatic symbiosis inside the host. However, they are also instrumental in shaping the functions of our immune system via a diverse set of communication mechanisms. In the gut, T helper 17, regulatory T and B cells are continuously tuned by specific microbial strains and metabolic processes. These cells in return help to establish a mutually beneficial exchange with the gut microbial contents. Imbalances in this symbiosis lead to dysregulations in the host's ability to control infections and the development of autoimmune diseases. In addition, the commensal microbiota has a significant and obligatory role in shaping both gut intrinsic and distal lymphoid organs, casting a large impact on the overall immune landscape in the host. This review discusses the major components of the microbial community in the gut and how its members collectively and individually exert regulatory roles in the host immune system and lymphoid structure development, as well as the functions of several major immune cell types.
Collapse
|
177
|
Hodzic Z, Bolock AM, Good M. The Role of Mucosal Immunity in the Pathogenesis of Necrotizing Enterocolitis. Front Pediatr 2017; 5:40. [PMID: 28316967 PMCID: PMC5334327 DOI: 10.3389/fped.2017.00040] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/15/2017] [Indexed: 12/29/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is the most devastating gastrointestinal disease of prematurity. Although the precise cause is not well understood, the main risk factors thought to contribute to NEC include prematurity, formula feeding, and bacterial colonization. Recent evidence suggests that NEC develops as a consequence of intestinal hyper-responsiveness to microbial ligands upon bacterial colonization in the preterm infant, initiating a cascade of aberrant signaling events, and a robust pro-inflammatory mucosal immune response. We now have a greater understanding of important mechanisms of disease pathogenesis, such as the role of cytokines, immunoglobulins, and immune cells in NEC. In this review, we will provide an overview of the mucosal immunity of the intestine and the relationship between components of the mucosal immune system involved in the pathogenesis of NEC, while highlighting recent advances in the field that have promise as potential therapeutic targets. First, we will describe the cellular components of the intestinal epithelium and mucosal immune system and their relationship to NEC. We will then discuss the relationship between the gut microbiota and cell signaling that underpins disease pathogenesis. We will conclude our discussion by highlighting notable therapeutic advancements in NEC that target the intestinal mucosal immunity.
Collapse
Affiliation(s)
- Zerina Hodzic
- University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Alexa M Bolock
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| | - Misty Good
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine , St. Louis, MO , USA
| |
Collapse
|
178
|
Rice TC, Armocida SM, Kuethe JW, Midura EF, Jain A, Hildeman DA, Healy DP, Gulbins E, Caldwell CC. Burn injury influences the T cell homeostasis in a butyrate-acid sphingomyelinase dependent manner. Cell Immunol 2016; 313:25-31. [PMID: 28063598 DOI: 10.1016/j.cellimm.2016.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/23/2016] [Accepted: 12/24/2016] [Indexed: 01/12/2023]
Abstract
Following burn injury, a key factor for patients susceptible to opportunistic infections is immune suppression. Butyrate levels are important in maintaining a functional immune system and these levels can be altered after injury. The acid sphingomyelinase (Asm) lipid signaling system has been implicated in a T cell actions with some evidence of being influenced by butyrate. Here, we hypothesized that burn-injury changes in butyrate levels would mediate Asm activity and, consequently, T cell homeostasis. We demonstrate that burn injury temporally decreases butyrate levels. We further determined that T cell Asm activity is increased by butyrate and decreased after burn injury. We additionally observed decreased T cell numbers in Asm-deficient, burn-injured, and microbiota-depleted mice. Finally, we demonstrate that butyrate reduced T cell death in an Asm-dependent manner. These data suggest that restoration of butyrate after burn injury may ameliorate the T cell lost observed in burn-injured patients by Asm regulation.
Collapse
Affiliation(s)
- Teresa C Rice
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Stephanie M Armocida
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Joshua W Kuethe
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Emily F Midura
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Ayushi Jain
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, USA
| | - Daniel P Healy
- James L. Winkle College of Pharmacy, Division of Pharmacy Practice and Administrative Sciences, University of Cincinnati Academic Health Center, Cincinnati, OH, USA
| | - Erich Gulbins
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Charles C Caldwell
- Division of Research, Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
179
|
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
|
180
|
Kwong Chung CKC, Ronchi F, Geuking MB. Detrimental effect of systemic antimicrobial CD4 + T-cell reactivity on gut epithelial integrity. Immunology 2016; 150:221-235. [PMID: 27779311 PMCID: PMC5214769 DOI: 10.1111/imm.12682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/09/2016] [Accepted: 10/17/2016] [Indexed: 12/18/2022] Open
Abstract
Healthy host-microbe mutualism relies on compartmentalization and proper regulation of systemic and mucosal immune responses. Nevertheless, the systemic immune system is frequently exposed to bouts of bacteraemia, which can trigger systemic antimicrobial immune reactivity including CD4+ T cells. Low-level bacteraemia can occur when immune compartmentalization is compromised, for example in the presence of innate immune deficiency or following use of non-steroidal anti-inflammatory drugs. We generated an Escherichia coli strain expressing a defined T helper neo-epitope to study systemic antigen-specific antimicrobial CD4+ T cells and their potential involvement in the pathogenisis of inflammatory bowel diseases. We found that the dose of bacteria required for the induction of systemic antimicrobial CD4+ T-cell proliferation was high and not easily reached under physiological conditions. Importantly, however, when intestinal barrier function was compromised by induced damage to the intestinal epithelium, the presence of systemic antimicrobial CD4+ T cells specific for a single neo-antigen resulted in dramatically increased levels of bacterial translocation. This study therefore demonstrates that systemic antimicrobial CD4+ T-cell reactivity might impact adversely on the mucosa under conditions of reduced barrier function and that despite strong mucosal immune regulation, antigen-specific recognition is still sensitive.
Collapse
Affiliation(s)
- Cheong K C Kwong Chung
- Department of Clinical Research (DKF), Mucosal Immunology Laboratory, University of Bern, Bern, Switzerland
| | - Francesca Ronchi
- Department of Clinical Research (DKF), Mucosal Immunology Laboratory, University of Bern, Bern, Switzerland
| | - Markus B Geuking
- Department of Clinical Research (DKF), Mucosal Immunology Laboratory, University of Bern, Bern, Switzerland.,Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
181
|
Sutherland DB, Suzuki K, Fagarasan S. Fostering of advanced mutualism with gut microbiota by Immunoglobulin A. Immunol Rev 2016; 270:20-31. [PMID: 26864102 DOI: 10.1111/imr.12384] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immunoglobulin A (IgA), the most abundantly secreted antibody isotype in mammals, not only provides direct immune protection to neonates via maternal milk but also helps program the infant immune system by regulating the microbiota. IgA continues to maintain dynamic interactions with the gut microbiota throughout life and this influences immune system homeostasis as well as other physiological processes. The secretory IgA produced independently of T-cell selection are commonly referred to as natural or innate antibodies. Our studies have shown that innate-IgA, while effective at excluding microorganisms from the gut, does not promote mutualism with the microbiota in the same way as adaptive-IgA that is selected in T cell-dependent germinal center reactions. Adaptive-IgA fosters more advanced mutualism with the microbiota than innate-IgA by selecting and diversifying beneficial microbial communities. In this review, we suggest that the diversified microbiota resulting from adaptive-IgA pressure was pivotal in promoting ecological adaptability and speciation potential of mammals.
Collapse
Affiliation(s)
- Duncan B Sutherland
- Laboratory of Intestinal Immunology, Global Health Institute, School of Life Sciences, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Keiichiro Suzuki
- Center for Innovation in Immunoregulative Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sidonia Fagarasan
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences (IMS), RIKEN Yokohama Institute, Yokohama, Japan
| |
Collapse
|
182
|
Chinen T, Kannan AK, Levine AG, Fan X, Klein U, Zheng Y, Gasteiger G, Feng Y, Fontenot JD, Rudensky AY. An essential role for the IL-2 receptor in T reg cell function. Nat Immunol 2016; 17:1322-1333. [PMID: 27595233 PMCID: PMC5071159 DOI: 10.1038/ni.3540] [Citation(s) in RCA: 631] [Impact Index Per Article: 70.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 07/27/2016] [Indexed: 12/12/2022]
Abstract
Regulatory T cells (Treg cells), which have abundant expression of the interleukin 2 receptor (IL-2R), are reliant on IL-2 produced by activated T cells. This feature indicates a key role for a simple network based on the consumption of IL-2 by Treg cells in their suppressor function. However, congenital deficiency in IL-2R results in reduced expression of the Treg cell lineage-specification factor Foxp3, which has confounded experimental efforts to understand the role of IL-2R expression and signaling in the suppressor function of Treg cells. Using genetic gain- and loss-of-function approaches, we found that capture of IL-2 was dispensable for the control of CD4+ T cells but was important for limiting the activation of CD8+ T cells, and that IL-2R-dependent activation of the transcription factor STAT5 had an essential role in the suppressor function of Treg cells separable from signaling via the T cell antigen receptor.
Collapse
Affiliation(s)
- Takatoshi Chinen
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Andrew G Levine
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xiying Fan
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ulf Klein
- Herbert Irving Comprehensive Cancer Center, Department of Pathology and Cell Biology, and Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Ye Zheng
- Nomis Foundation Laboratories for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Georg Gasteiger
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Institute for Medical Microbiology and Hygiene, University of Mainz Medical Centre, Mainz, Germany
| | - Yongqiang Feng
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Alexander Y Rudensky
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
183
|
Bronson PG, Chang D, Bhangale T, Seldin MF, Ortmann W, Ferreira RC, Urcelay E, Pereira LF, Martin J, Plebani A, Lougaris V, Friman V, Freiberger T, Litzman J, Thon V, Pan-Hammarström Q, Hammarström L, Graham RR, Behrens TW. Common variants at PVT1, ATG13-AMBRA1, AHI1 and CLEC16A are associated with selective IgA deficiency. Nat Genet 2016; 48:1425-1429. [PMID: 27723758 PMCID: PMC5086090 DOI: 10.1038/ng.3675] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/24/2016] [Indexed: 12/18/2022]
Abstract
Selective immunoglobulin A deficiency (IgAD) is the most common primary immunodeficiency in Europeans. Our genome-wide association study (GWAS) meta-analysis of 1,635 patients with IgAD and 4,852 controls identified four new significant (P < 5 × 10-8) loci and association with a rare IFIH1 variant (p.Ile923Val). Peak new variants (PVT1, P = 4.3 × 10-11; ATG13-AMBRA1, P = 6.7 × 10-10; AHI1, P = 8.4 × 10-10; CLEC16A, P = 1.4 × 10-9) overlapped with autoimmune markers (3/4) and correlated with 21 putative regulatory variants, including expression quantitative trait loci (eQTLs) for AHI1 and DEXI and DNase hypersensitivity sites in FOXP3+ regulatory T cells. Pathway analysis of the meta-analysis results showed striking association with the KEGG pathway for IgA production (pathway P < 0.0001), with 22 of the 30 annotated pathway genes containing at least one variant with P ≤ 0.05 in the IgAD meta-analysis. These data suggest that a complex network of genetic effects, including genes known to influence the biology of IgA production, contributes to IgAD.
Collapse
Affiliation(s)
- Paola G. Bronson
- Department of Human Genetics, Genentech, Inc., South San
Francisco, CA, USA
| | - Diana Chang
- Department of Human Genetics, Genentech, Inc., South San
Francisco, CA, USA
| | - Tushar Bhangale
- Department of Bioinformatics and Computational Biology,
Genentech, Inc., South San Francisco, CA, USA
| | - Michael F. Seldin
- Department of Biochemistry, School of Medicine, University
of California, Davis, CA, USA
| | - Ward Ortmann
- Department of Human Genetics, Genentech, Inc., South San
Francisco, CA, USA
| | - Ricardo C. Ferreira
- Juvenile Diabetes Research Foundation/Wellcome Trust
Diabetes and Inflammation Laboratory, Cambridge Institute for Medical Research,
Cambridge, UK
| | - Elena Urcelay
- Department of Immunology, Instituto de Investigación
Sanitaria del Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | | | - Javier Martin
- Instituto de Parasitología y Biomedicina
López-Neyra, CSIC, Granada, Spain
| | - Alessandro Plebani
- Pediatrics Clinic, Department of Clinical and Experimental
Sciences, University of Brescia, Spedali Civili di Brescia, Italy
- Institute for Molecular Medicine, A. Nocivelli, Department
of Clinical and Experimental Sciences, University of Brescia, Spedali Civili di
Brescia, Italy
| | - Vassilios Lougaris
- Pediatrics Clinic, Department of Clinical and Experimental
Sciences, University of Brescia, Spedali Civili di Brescia, Italy
- Institute for Molecular Medicine, A. Nocivelli, Department
of Clinical and Experimental Sciences, University of Brescia, Spedali Civili di
Brescia, Italy
| | - Vanda Friman
- Department of Infectious Diseases, University of
Gothenburg, Gothenburg, Sweden
| | - Tomáš Freiberger
- Molecular Genetics Laboratory, Centre for Cardiovascular
Surgery and Transplantation, Brno, Czech Republic
- Central European Institute of Technology, Masaryk
University, Brno, Czech Republic
| | - Jiri Litzman
- Department of Clinical Immunology and Allergy, Faculty of
Medicine, Masaryk University, St. Anne’s Univ. Hospital, Brno, Czech
Republic
| | - Vojtech Thon
- Department of Clinical Immunology and Allergy, Faculty of
Medicine, Masaryk University, St. Anne’s Univ. Hospital, Brno, Czech
Republic
- Research Centre for Toxic Compounds in the Environment,
Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Qiang Pan-Hammarström
- Division of Clinical Immunology & Transfusion
Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lennart Hammarström
- Division of Clinical Immunology & Transfusion
Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Robert R. Graham
- Department of Human Genetics, Genentech, Inc., South San
Francisco, CA, USA
| | - Timothy W. Behrens
- Department of Human Genetics, Genentech, Inc., South San
Francisco, CA, USA
| |
Collapse
|
184
|
The Microbiome, Timing, and Barrier Function in the Context of Allergic Disease. Immunity 2016; 44:728-38. [PMID: 27096316 DOI: 10.1016/j.immuni.2016.02.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Indexed: 12/14/2022]
Abstract
Allergic disease affects millions. Despite many advances in our understanding of the immune system in the past century, the physiologic underpinning for the existence of allergy remains largely mysterious. Food allergies, in particular, have increased dramatically in recent years, adding a new sense of urgency to unraveling this mystery. The concurrence of significant lifestyle changes in Western societies with increasing disease prevalence implies a causal link. Demographic variables that influence the composition and function of the commensal microbiota early in life seem to be most important. Identifying the evolutionary and physiologic foundations of allergic disease and defining what about our modern environment is responsible for its increased incidence will provide insights critical to the development of new approaches to prevention and treatment.
Collapse
|
185
|
Santisteban MM, Kim S, Pepine CJ, Raizada MK. Brain-Gut-Bone Marrow Axis: Implications for Hypertension and Related Therapeutics. Circ Res 2016; 118:1327-36. [PMID: 27081113 DOI: 10.1161/circresaha.116.307709] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/18/2016] [Indexed: 02/06/2023]
Abstract
Hypertension is the most prevalent modifiable risk factor for cardiovascular disease and disorders directly influencing cardiovascular disease morbidity and mortality, such as diabetes mellitus, chronic kidney disease, obstructive sleep apnea, etc. Despite aggressive attempts to influence lifestyle modifications and advances in pharmacotherapeutics, a large percentage of patients still do not achieve recommended blood pressure control worldwide. Thus, we think that mechanism-based novel strategies should be considered to significantly improve control and management of hypertension. The overall objective of this review is to summarize implications of peripheral- and neuroinflammation as well as the autonomic nervous system-bone marrow communication in hematopoietic cell homeostasis and their impact on hypertension pathophysiology. In addition, we discuss the novel and emerging field of intestinal microbiota and roles of gut permeability and dysbiosis in cardiovascular disease and hypertension. Finally, we propose a brain-gut-bone marrow triangular interaction hypothesis and discuss its potential in the development of novel therapies for hypertension.
Collapse
Affiliation(s)
- Monica M Santisteban
- From the Department of Physiology and Functional Genomics (M.M.S., S.K., M.K.R.) and Division of Cardiovascular Medicine, Department of Medicine (C.J.P.), College of Medicine, University of Florida, Gainesville
| | - Seungbum Kim
- From the Department of Physiology and Functional Genomics (M.M.S., S.K., M.K.R.) and Division of Cardiovascular Medicine, Department of Medicine (C.J.P.), College of Medicine, University of Florida, Gainesville
| | - Carl J Pepine
- From the Department of Physiology and Functional Genomics (M.M.S., S.K., M.K.R.) and Division of Cardiovascular Medicine, Department of Medicine (C.J.P.), College of Medicine, University of Florida, Gainesville
| | - Mohan K Raizada
- From the Department of Physiology and Functional Genomics (M.M.S., S.K., M.K.R.) and Division of Cardiovascular Medicine, Department of Medicine (C.J.P.), College of Medicine, University of Florida, Gainesville.
| |
Collapse
|
186
|
Hand TW. The Role of the Microbiota in Shaping Infectious Immunity. Trends Immunol 2016; 37:647-658. [PMID: 27616558 DOI: 10.1016/j.it.2016.08.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 02/07/2023]
Abstract
Humans are meta-organisms that maintain a diverse population of microorganisms on their barrier surfaces, collectively named the microbiota. Since most pathogens either cross or inhabit barrier surfaces, the microbiota plays a critical and often protective role during infections, both by modulating immune system responses and by mediating colonization resistance. However, the microbiota can also act as a reservoir for opportunistic microorganisms that can 'bloom', significantly complicating diseases of barrier surfaces by contributing to inflammatory immune responses. This review discusses our current understanding of the complex interactions between the host, its microbiota, and pathogenic organisms, focusing in particular on the intestinal mucosa.
Collapse
Affiliation(s)
- Timothy W Hand
- Richard King Mellon Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| |
Collapse
|
187
|
Sapir-Koren R, Livshits G. Rheumatoid arthritis onset in postmenopausal women: Does the ACPA seropositive subset result from genetic effects, estrogen deficiency, skewed profile of CD4(+) T-cells, and their interactions? Mol Cell Endocrinol 2016; 431:145-63. [PMID: 27178986 DOI: 10.1016/j.mce.2016.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 05/04/2016] [Accepted: 05/09/2016] [Indexed: 12/28/2022]
Abstract
Rheumatoid arthritis (RA) incidence displays a differentiated age-dependent female-to-male ratio in which women outnumber men. Evidence that the peak incidence of RA in women coincides with menopause age, suggests a potential estrogenic role to disease etiology. Estrogens exert physiologically both stimulatory and inhibitory effects on the immune system. Epidemiologic and animal model studies with estrogen deprivation or supplementation suggested estrogens as to play, mainly, a protective role in RA immunopathology. In this review, we propose that some yet unidentified disturbances associated with estrogen circulating levels, differentiated by the menopausal status, play a major role in women's RA susceptibility. We focus on the interaction between estrogen deprivation and genetic risk alleles for anti-citrullinated protein antibodies (ACPA) seropositive RA, as a major driving force for increased immune reactivity and RA susceptibility, in postmenopausal women. This opens up new fields for research concerning the association among different irregular estrogenic conditions, the cytokine milieu, and age/menopausal status bias in RA.
Collapse
Affiliation(s)
- Rony Sapir-Koren
- Human Population Biology Research Group, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gregory Livshits
- Human Population Biology Research Group, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Lilian and Marcel Pollak Chair of Biological Anthropology, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
188
|
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: 1249] [Impact Index Per Article: 138.8] [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
|
189
|
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: 1.8] [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
|
190
|
Wymore Brand M, Wannemuehler MJ, Phillips GJ, Proctor A, Overstreet AM, Jergens AE, Orcutt RP, Fox JG. The Altered Schaedler Flora: Continued Applications of a Defined Murine Microbial Community. ILAR J 2016; 56:169-78. [PMID: 26323627 DOI: 10.1093/ilar/ilv012] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal (GI) microbiota forms a mutualistic relationship with the host through complex and dynamic interactions. Because of the complexity and interindividual variation of the GI microbiota, investigating how members of the microbiota interact with each other, as well as with the host, is daunting. The altered Schaedler flora (ASF) is a model community of eight microorganisms that was developed by R.P. Orcutt and has been in use since the late 1970s. The eight microorganisms composing the ASF were all derived from mice, can be cultured in vitro, and are stably passed through multiple generations (at least 15 years or more by the authors) in gnotobiotic mice continually bred in isolator facilities. With the limitations associated with conventional, mono- or biassociated, and germfree mice, use of mice colonized with a consortium of known bacteria that naturally inhabit the murine gut offers a powerful system to investigate mechanisms governing host-microbiota relationships, and how members of the GI microbiota interact with one another. The ASF community offers significant advantages to study homeostatic as well as disease-related interactions by taking advantage of a well-defined, limited community of microorganisms. For example, quantification and spatial distribution of individual members, microbial genetic manipulation, genomic-scale analysis, and identification of microorganism-specific host immune responses are all achievable using the ASF model. This review compiles highlights associated with the 37-year history of the ASF, including descriptions of its continued use in biomedical research to elucidate the complexities of host-microbiome interactions in health and disease.
Collapse
Affiliation(s)
- Meghan Wymore Brand
- Meghan Wymore Brand, DVM, is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Michael J. Wannemuehler, MS, PhD, is Professor and Chair in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Gregory J. Phillips, MA, PhD, is a professor in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Alexandra Proctor is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Anne-Marie Overstreet, PhD, is a postdoctoral fellow in the Department of Microbiology and Immunology at Indiana University School of Medicine-South Bend in South Bend, Indiana. Albert E. Jergens, DVM, MS, PhD, is Professor and Associate Chair for Research and Graduate Studies in the Department of Veterinary Clinical Sciences at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Roger P. Orcutt, PhD, is a consultant at Biomedical Research Associates in Dunkirk, New York. James G. Fox, MS, DVM, is Director of the Division of Comparative Medicine and Professor in the Department of Biological Engineering at Massachusetts Institute of Technology in Cambridge, Massachusetts
| | - Michael J Wannemuehler
- Meghan Wymore Brand, DVM, is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Michael J. Wannemuehler, MS, PhD, is Professor and Chair in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Gregory J. Phillips, MA, PhD, is a professor in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Alexandra Proctor is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Anne-Marie Overstreet, PhD, is a postdoctoral fellow in the Department of Microbiology and Immunology at Indiana University School of Medicine-South Bend in South Bend, Indiana. Albert E. Jergens, DVM, MS, PhD, is Professor and Associate Chair for Research and Graduate Studies in the Department of Veterinary Clinical Sciences at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Roger P. Orcutt, PhD, is a consultant at Biomedical Research Associates in Dunkirk, New York. James G. Fox, MS, DVM, is Director of the Division of Comparative Medicine and Professor in the Department of Biological Engineering at Massachusetts Institute of Technology in Cambridge, Massachusetts
| | - Gregory J Phillips
- Meghan Wymore Brand, DVM, is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Michael J. Wannemuehler, MS, PhD, is Professor and Chair in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Gregory J. Phillips, MA, PhD, is a professor in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Alexandra Proctor is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Anne-Marie Overstreet, PhD, is a postdoctoral fellow in the Department of Microbiology and Immunology at Indiana University School of Medicine-South Bend in South Bend, Indiana. Albert E. Jergens, DVM, MS, PhD, is Professor and Associate Chair for Research and Graduate Studies in the Department of Veterinary Clinical Sciences at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Roger P. Orcutt, PhD, is a consultant at Biomedical Research Associates in Dunkirk, New York. James G. Fox, MS, DVM, is Director of the Division of Comparative Medicine and Professor in the Department of Biological Engineering at Massachusetts Institute of Technology in Cambridge, Massachusetts
| | - Alexandra Proctor
- Meghan Wymore Brand, DVM, is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Michael J. Wannemuehler, MS, PhD, is Professor and Chair in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Gregory J. Phillips, MA, PhD, is a professor in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Alexandra Proctor is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Anne-Marie Overstreet, PhD, is a postdoctoral fellow in the Department of Microbiology and Immunology at Indiana University School of Medicine-South Bend in South Bend, Indiana. Albert E. Jergens, DVM, MS, PhD, is Professor and Associate Chair for Research and Graduate Studies in the Department of Veterinary Clinical Sciences at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Roger P. Orcutt, PhD, is a consultant at Biomedical Research Associates in Dunkirk, New York. James G. Fox, MS, DVM, is Director of the Division of Comparative Medicine and Professor in the Department of Biological Engineering at Massachusetts Institute of Technology in Cambridge, Massachusetts
| | - Anne-Marie Overstreet
- Meghan Wymore Brand, DVM, is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Michael J. Wannemuehler, MS, PhD, is Professor and Chair in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Gregory J. Phillips, MA, PhD, is a professor in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Alexandra Proctor is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Anne-Marie Overstreet, PhD, is a postdoctoral fellow in the Department of Microbiology and Immunology at Indiana University School of Medicine-South Bend in South Bend, Indiana. Albert E. Jergens, DVM, MS, PhD, is Professor and Associate Chair for Research and Graduate Studies in the Department of Veterinary Clinical Sciences at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Roger P. Orcutt, PhD, is a consultant at Biomedical Research Associates in Dunkirk, New York. James G. Fox, MS, DVM, is Director of the Division of Comparative Medicine and Professor in the Department of Biological Engineering at Massachusetts Institute of Technology in Cambridge, Massachusetts
| | - Albert E Jergens
- Meghan Wymore Brand, DVM, is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Michael J. Wannemuehler, MS, PhD, is Professor and Chair in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Gregory J. Phillips, MA, PhD, is a professor in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Alexandra Proctor is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Anne-Marie Overstreet, PhD, is a postdoctoral fellow in the Department of Microbiology and Immunology at Indiana University School of Medicine-South Bend in South Bend, Indiana. Albert E. Jergens, DVM, MS, PhD, is Professor and Associate Chair for Research and Graduate Studies in the Department of Veterinary Clinical Sciences at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Roger P. Orcutt, PhD, is a consultant at Biomedical Research Associates in Dunkirk, New York. James G. Fox, MS, DVM, is Director of the Division of Comparative Medicine and Professor in the Department of Biological Engineering at Massachusetts Institute of Technology in Cambridge, Massachusetts
| | - Roger P Orcutt
- Meghan Wymore Brand, DVM, is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Michael J. Wannemuehler, MS, PhD, is Professor and Chair in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Gregory J. Phillips, MA, PhD, is a professor in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Alexandra Proctor is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Anne-Marie Overstreet, PhD, is a postdoctoral fellow in the Department of Microbiology and Immunology at Indiana University School of Medicine-South Bend in South Bend, Indiana. Albert E. Jergens, DVM, MS, PhD, is Professor and Associate Chair for Research and Graduate Studies in the Department of Veterinary Clinical Sciences at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Roger P. Orcutt, PhD, is a consultant at Biomedical Research Associates in Dunkirk, New York. James G. Fox, MS, DVM, is Director of the Division of Comparative Medicine and Professor in the Department of Biological Engineering at Massachusetts Institute of Technology in Cambridge, Massachusetts
| | - James G Fox
- Meghan Wymore Brand, DVM, is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Michael J. Wannemuehler, MS, PhD, is Professor and Chair in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Gregory J. Phillips, MA, PhD, is a professor in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Alexandra Proctor is a graduate student in the Department of Veterinary Microbiology and Preventive Medicine at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Anne-Marie Overstreet, PhD, is a postdoctoral fellow in the Department of Microbiology and Immunology at Indiana University School of Medicine-South Bend in South Bend, Indiana. Albert E. Jergens, DVM, MS, PhD, is Professor and Associate Chair for Research and Graduate Studies in the Department of Veterinary Clinical Sciences at the College of Veterinary Medicine at Iowa State University in Ames, Iowa. Roger P. Orcutt, PhD, is a consultant at Biomedical Research Associates in Dunkirk, New York. James G. Fox, MS, DVM, is Director of the Division of Comparative Medicine and Professor in the Department of Biological Engineering at Massachusetts Institute of Technology in Cambridge, Massachusetts
| |
Collapse
|
191
|
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
|
192
|
Chen F, Cao A, Yao S, Evans-Marin HL, Liu H, Wu W, Carlsen ED, Dann SM, Soong L, Sun J, Zhao Q, Cong Y. mTOR Mediates IL-23 Induction of Neutrophil IL-17 and IL-22 Production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:4390-9. [PMID: 27067005 PMCID: PMC4868807 DOI: 10.4049/jimmunol.1501541] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 03/11/2016] [Indexed: 12/31/2022]
Abstract
It has been shown recently that neutrophils are able to produce IL-22 and IL-17, which differentially regulate the pathogenesis of inflammatory bowel disease. However, it is still largely unknown how the neutrophil production of IL-22 and IL-17 is regulated, and their role in the pathogenesis of inflammatory bowel disease. In this study, we found that IL-23 promoted neutrophil production of IL-17 and IL-22. IL-23 stimulated the neutrophil expression of IL-23R as well as rorc and ahr. Retinoid acid receptor-related orphan receptor γ t and aryl-hydrocarbon receptor differentially regulated IL-23 induction of neutrophil IL-17 and IL-22. In addition, IL-23 induced the activation of mTOR in neutrophils. Blockade of the mTOR pathway inhibited IL-23-induced expression of rorc and ahr, as well as IL-17 and IL-22 production. By using a microbiota Ag-specific T cell-mediated colitis model, we demonstrated that depletion of neutrophils, as well as blockade of IL-22, resulted in a significant increase in the severity of colitis, thereby indicating a protective role of neutrophils and IL-22 in chronic colitis. Collectively, our data revealed that neutrophils negatively regulate microbiota Ag-specific T cell induction of colitis, and IL-23 induces neutrophil production of IL-22 and IL-17 through induction of rorc and ahr, which is mediated by the mTOR pathway.
Collapse
MESH Headings
- Animals
- Cecum/pathology
- Cell Differentiation
- Colitis/immunology
- Colitis/pathology
- Colon/pathology
- Interleukin-17/biosynthesis
- Interleukin-23/metabolism
- Interleukin-23/pharmacology
- Interleukins/biosynthesis
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neutrophils/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Receptors, Interleukin/genetics
- Receptors, Interleukin/metabolism
- Signal Transduction
- TOR Serine-Threonine Kinases/genetics
- Th17 Cells/immunology
- Interleukin-22
Collapse
Affiliation(s)
- Feidi Chen
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Anthony 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
| | - Heather L Evans-Marin
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Han Liu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555; Department of Gastroenterology, The Qilu Hospital, Shandong University, Shandong 250012, China
| | - Wei Wu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555; Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Eric D Carlsen
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555; Department of Medicine, University of Texas Medical Branch, Galveston, TX 77555; and
| | - Sara M Dann
- Department of Medicine, University of Texas Medical Branch, Galveston, TX 77555; and
| | - Lynn Soong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | | | - Yingzi Cong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555;
| |
Collapse
|
193
|
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: 414] [Impact Index Per Article: 46.0] [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
|
194
|
Gan P, Jin D, Zhao X, Gao Z, Wang S, Du P, Qi G. Bacillus-produced surfactin attenuates chronic inflammation in atherosclerotic lesions of ApoE(-/-) mice. Int Immunopharmacol 2016; 35:226-234. [PMID: 27082998 DOI: 10.1016/j.intimp.2016.03.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 03/10/2016] [Accepted: 03/30/2016] [Indexed: 10/21/2022]
Abstract
Bacillus-produced surfactin can inhibit acute inflammation in vitro and in vivo. However, there is no report whether surfactin could inhibit chronic inflammation in the atherosclerotic lesions. Apoliprotein E deficient (ApoE(-/-)) mice (fed on atherogenic diet) were intragastrically administered with surfactin for 9 doses, then the athero-protective effect of surfactin was determined in vivo. The results showed surfactin could induce anti-inflammatory factors such as IgA, transforming growth factor (TGF)-β and interleukin (IL)-10 in the intestine. Further investigation discovered that surfactin also systemically induced CD4(+)CD25(+)FoxP3(+) Tregs in spleen, which could inhibit T cells to produce pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α and interferon (IFN)-γ. The IgG subclass pattern with high titer of IgG1 (Th2-type) but low titer of IgG2a (Th1-type) was also found in the surfactin-treated mice. As a result, the attenuation of chronic inflammation was observed in the surfactin-treated groups accompanying with less TNF-α but more IL-10 in the atherosclerotic lesions. Moreover, surfactin could reduce serum total cholesterol and cholesterol in low-density lipoprotein, and increase serum cholesterol in high-density lipoprotein in mice. Collectively, surfactin could significantly attenuate atherosclerotic lesions on the aorta by restoration of the delicate balance of Th1/Th2 response in mice.
Collapse
Affiliation(s)
- Ping Gan
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Dong Jin
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiuyun Zhao
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhenqiu Gao
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; School of Pharmacy, Yancheng Teachers' University, Xiwang Road, Yancheng 224051, China
| | - Shengying Wang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Peng Du
- College of Life Science, Hubei University, 430062, China
| | - Gaofu Qi
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
195
|
The microbiota as a component of the celiac disease and non-celiac gluten sensitivity. CLINICAL NUTRITION EXPERIMENTAL 2016. [DOI: 10.1016/j.yclnex.2016.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
196
|
Magen E, Waitman DA, Goldstein N, Schlesinger M, Dickstein Y, Kahan NR. Helicobacter pylori infection in patients with selective immunoglobulin a deficiency. Clin Exp Immunol 2016; 184:332-7. [PMID: 26749258 DOI: 10.1111/cei.12765] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/12/2015] [Accepted: 12/14/2015] [Indexed: 12/24/2022] Open
Abstract
Selective immunoglobulin A (IgA) deficiency (IgAD) is the most common primary immunodeficiency in the western world. The aim of the study was to investigate the prevalence and clinical characteristics of Helicobacter pylori-infected dyspeptic patients with IgAD. Case samples were drawn from all subjects ≥ 12 years of age (n = 104729) who had undergone serum total IgA measurements during 2004-14 for any reason at Leumit Healthcare Services (Israel) and had serum total IgA < 0·07 g/l. The control group was comprised of a random sample of remaining patients with a case-control ratio of 10 controls for each case. The dyspeptic diseases were identified and retrieved from Leumit Health Care Services electronic database using specific ICD-9-CM diagnostic codes. The case group included 347 subjects and the control group 3470 subjects. There were no significant differences in the prevalence of patients with dyspepsia [84 (24·2%) versus 821 (23·6%) for cases and controls, respectively]. Additionally, there was no difference in a proportion of dyspeptic H. pylori-positive subjects [59 (17·1%) versus 524 (15·1%)] between the case and control groups. Only 59 (17%) among the 347 IgAD patients underwent gastroscopy. A significantly larger proportion of case subjects experienced several forms of gastritis [13 (61·9%) versus 38 (21·6%), P < 0·001), duodenal ulcers [seven (33·3%) versus 19 (10·8%); P = 0·01] and nodular lymphoid hyperplasia (NLH) [two (9·5%) versus none; P = 0·011]. IgAD is not associated with increased prevalence of H. pylori-associated dyspepsia; nevertheless, H. pylori-infected dyspeptic IgAD subjects experience more EGD-proved gastritis, duodenal ulcers and NLH.
Collapse
Affiliation(s)
- E Magen
- Leumit Health Services, Ashdod, Israel.,Clinical Immunology and Allergy Unit, Barzilai University Medical Center, Ben Gurion University of the Negev, Ashkelon, Israel
| | | | - N Goldstein
- Clinical Immunology and Allergy Unit, Barzilai University Medical Center, Ben Gurion University of the Negev, Ashkelon, Israel
| | - M Schlesinger
- Clinical Immunology and Allergy Unit, Barzilai University Medical Center, Ben Gurion University of the Negev, Ashkelon, Israel
| | | | - N R Kahan
- Leumit Health Services, Ashdod, Israel.,School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
197
|
Secretory IgA in complex with Lactobacillus rhamnosus potentiates mucosal dendritic cell-mediated Treg cell differentiation via TLR regulatory proteins, RALDH2 and secretion of IL-10 and TGF-β. Cell Mol Immunol 2016; 14:546-556. [PMID: 26972771 DOI: 10.1038/cmi.2015.110] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/02/2015] [Accepted: 12/02/2015] [Indexed: 12/16/2022] Open
Abstract
The importance of secretory IgA in controlling the microbiota is well known, yet how the antibody affects the perception of the commensals by the local immune system is still poorly defined. We have previously shown that the transport of secretory IgA in complex with bacteria across intestinal microfold cells results in an association with dendritic cells in Peyer's patches. However, the consequences of such an interaction on dendritic cell conditioning have not been elucidated. In this study, we analyzed the impact of the commensal Lactobacillus rhamnosus, alone or associated with secretory IgA, on the responsiveness of dendritic cells freshly recovered from mouse Peyer's patches, mesenteric lymph nodes, and spleen. Lactobacillus rhamnosus-conditioned mucosal dendritic cells are characterized by increased expression of Toll-like receptor regulatory proteins [including single immunoglobulin interleukin-1 receptor-related molecule, suppressor of cytokine signaling 1, and Toll-interacting molecule] and retinaldehyde dehydrogenase 2, low surface expression of co-stimulatory markers, high anti- versus pro-inflammatory cytokine production ratios, and induction of T regulatory cells with suppressive function. Association with secretory IgA enhanced the anti-inflammatory/regulatory Lactobacillus rhamnosus-induced conditioning of mucosal dendritic cells, particularly in Peyer's patches. At the systemic level, activation of splenic dendritic cells exposed to Lactobacillus rhamnosus was partially dampened upon association with secretory IgA. These data suggest that secretory IgA, through coating of commensal bacteria, contributes to the conditioning of mucosal dendritic cells toward tolerogenic profiles essential for the maintenance of intestinal homeostasis.
Collapse
|
198
|
Abstract
Obesity and insulin resistance are associated with chronic inflammation in metabolic tissues such as adipose tissue and the liver. Recently, growing evidence has implicated the intestinal immune system as an important contributor to metabolic disease. Obesity predisposes to altered intestinal immunity and is associated with changes to the gut microbiota, intestinal barrier function, gut-residing innate and adaptive immune cells, and oral tolerance to luminal antigens. Accordingly, the gut immune system may represent a novel therapeutic target for systemic inflammation in insulin resistance. This review discusses the emerging field of intestinal immunity in obesity-related insulin resistance and how it affects metabolic disease.
Collapse
Affiliation(s)
- Daniel A Winer
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Pathology, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada.
| | - Helen Luck
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Sue Tsai
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Shawn Winer
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Laboratory Medicine, St. Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada.
| |
Collapse
|
199
|
Withers DR, Hepworth MR, Wang X, Mackley EC, Halford EE, Dutton EE, Marriott CL, Brucklacher-Waldert V, Veldhoen M, Kelsen J, Baldassano RN, Sonnenberg GF. Transient inhibition of ROR-γt therapeutically limits intestinal inflammation by reducing TH17 cells and preserving group 3 innate lymphoid cells. Nat Med 2016; 22:319-23. [PMID: 26878233 PMCID: PMC4948756 DOI: 10.1038/nm.4046] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/13/2016] [Indexed: 12/14/2022]
Abstract
RAR-related orphan receptor-γt (ROR-γt) directs differentiation of proinflammatory T helper 17 (TH17) cells and is a potential therapeutic target in chronic autoimmune and inflammatory diseases. However, ROR-γt-dependent group 3 innate lymphoid cells ILC3s provide essential immunity and tissue protection in the intestine, suggesting that targeting ROR-γt could also result in impaired host defense after infection or enhanced tissue damage. Here, we demonstrate that transient chemical inhibition of ROR-γt in mice selectively reduces cytokine production from TH17 but not ILCs in the context of intestinal infection with Citrobacter rodentium, resulting in preserved innate immunity. Temporal deletion of Rorc (encoding ROR-γt) in mature ILCs also did not impair cytokine response in the steady state or during infection. Finally, pharmacologic inhibition of ROR-γt provided therapeutic benefit in mouse models of intestinal inflammation and reduced the frequency of TH17 cells but not ILCs isolated from primary intestinal samples of individuals with inflammatory bowel disease (IBD). Collectively, these results reveal differential requirements for ROR-γt in the maintenance of TH17 cell and ILC3 responses and suggest that transient inhibition of ROR-γt is a safe and effective therapeutic approach during intestinal inflammation.
Collapse
Affiliation(s)
- David R. Withers
- MRC Centre for Immune Regulation, Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Matthew R. Hepworth
- Joan and Sanford I. Weill Department of Medicine, Gastroenterology Division, Weill Cornell Medicine, New York, New York, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, USA
- The Jill Robert’s Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, New York, USA
| | - Xinxin Wang
- Joan and Sanford I. Weill Department of Medicine, Gastroenterology Division, Weill Cornell Medicine, New York, New York, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, USA
- The Jill Robert’s Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, New York, USA
| | - Emma C. Mackley
- MRC Centre for Immune Regulation, Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Emily E. Halford
- MRC Centre for Immune Regulation, Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Emma E. Dutton
- MRC Centre for Immune Regulation, Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Clare L. Marriott
- MRC Centre for Immune Regulation, Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, UK
| | | | - Marc Veldhoen
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Judith Kelsen
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Robert N. Baldassano
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Gregory F. Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Gastroenterology Division, Weill Cornell Medicine, New York, New York, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, USA
- The Jill Robert’s Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
200
|
TLR5 mediates CD172α(+) intestinal lamina propria dendritic cell induction of Th17 cells. Sci Rep 2016; 6:22040. [PMID: 26907705 PMCID: PMC4764953 DOI: 10.1038/srep22040] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 02/05/2016] [Indexed: 12/30/2022] Open
Abstract
Multiple mechanisms exist in regulation of host responses to massive challenges from microbiota to maintain immune homeostasis in the intestines. Among these is the enriched Th17 cells in the intestines, which regulates intestinal homeostasis through induction of antimicrobial peptides and secretory IgA among others. However, the means by which Th17 cells develop in response to microbiota is still not completely understood. Although both TLR5 and CD172α(+) lamina propria dendritic cells (LPDC) have been shown to promote Th17 cell development, it is still unclear whether TLR5 mediates the CD172α(+)LPDC induction of Th17 cells. By using a microbiota antigen-specific T cell reporter mouse system, we demonstrated that microbiota antigen-specific T cells developed into Th17 cells in the intestinal LP, but not in the spleen when transferred into TCRβxδ(-/-) mice. LPDCs expressed high levels of TLR5, and most CD172α(+)LPDCs also co-expressed TLR5. LPDCs produced high levels of IL-23, IL-6 and TGFβ when stimulated with commensal flagellin and promoted Th17 cell development when cultured with full-length CBir1 flagellin but not CBir1 peptide. Wild-type CD172α(+), but not CD172α(-), LPDCs induced Th17 cells, whereas TLR5-deficient LPDC did not induce Th17 cells. Our data thereby demonstrated that TLR5 mediates CD172α(+)LPDC induction of Th17 cells in the intestines.
Collapse
|