101
|
Abstract
The family of innate lymphoid cells (ILCs) has attracted attention in recent years as its members are important regulators of immunity, while they can also cause pathology. In both mouse and man, ILCs were initially discovered in developing lymph nodes as lymphoid tissue inducer (LTi) cells. These cells form the prototypic members of the ILC family and play a central role in the formation of secondary lymphoid organs (SLOs). In the absence of LTi cells, lymph nodes (LN) and Peyer's Patches (PP) fail to form in mice, although the splenic white pulp can develop normally. Besides LTi cells, the ILC family encompasses helper-like ILCs with functional distinctions as seen by T-helper cells, as well as cytotoxic natural killer (NK) cells. ILCs are still present in adult SLOs where they have been shown to play a role in lymphoid tissue regeneration. Furthermore, ILCs were implicated to interact with adaptive lymphocytes and influence the adaptive immune response. Here, we review the recent literature on the role of ILCs in secondary lymphoid tissue from the formation of SLOs to mature SLOs in adults, during homeostasis and pathology.
Collapse
Affiliation(s)
- Yotam E Bar-Ephraïm
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
102
|
Abstract
The lymphatic vasculature is not considered a formal part of the immune system, but it is critical to immunity. One of its major roles is in the coordination of the trafficking of antigen and immune cells. However, other roles in immunity are emerging. Lymphatic endothelial cells, for example, directly present antigen or express factors that greatly influence the local environment. We cover these topics herein and discuss how other properties of the lymphatic vasculature, such as mechanisms of lymphatic contraction (which immunologists traditionally do not take into account), are nonetheless integral in the immune system. Much is yet unknown, and this nascent subject is ripe for exploration. We argue that to consider the impact of lymphatic biology in any given immunological interaction is a key step toward integrating immunology with organ physiology and ultimately many complex pathologies.
Collapse
Affiliation(s)
- Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110;
| | - Stoyan Ivanov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110;
| | - Bernd H Zinselmeyer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110;
| | - Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida 33612
| |
Collapse
|
103
|
Barone F, Gardner DH, Nayar S, Steinthal N, Buckley CD, Luther SA. Stromal Fibroblasts in Tertiary Lymphoid Structures: A Novel Target in Chronic Inflammation. Front Immunol 2016; 7:477. [PMID: 27877173 PMCID: PMC5100680 DOI: 10.3389/fimmu.2016.00477] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are organized aggregates of lymphocytes, myeloid, and stromal cells that provide ectopic hubs for acquired immune responses. TLS share phenotypical and functional features with secondary lymphoid organs (SLO); however, they require persistent inflammatory signals to arise and are often observed at target sites of autoimmune disease, chronic infection, cancer, and organ transplantation. Over the past 10 years, important progress has been made in our understanding of the role of stromal fibroblasts in SLO development, organization, and function. A complex and stereotyped series of events regulate fibroblast differentiation from embryonic life in SLOs to lymphoid organ architecture observed in adults. In contrast, TLS-associated fibroblasts differentiate from postnatal, locally activated mesenchyme, predominantly in settings of inflammation and persistent antigen presentation. Therefore, there are critical differences in the cellular and molecular requirements that regulate SLO versus TLS development that ultimately impact on stromal and hematopoietic cell function. These differences may contribute to the pathogenic nature of TLS in the context of chronic inflammation and malignant transformation and offer a window of opportunity for therapeutic interventions in TLS associated pathologies.
Collapse
Affiliation(s)
- Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - David H Gardner
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Nathalie Steinthal
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Christopher D Buckley
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Sanjiv A Luther
- Department of Biochemistry, Center for Immunity and Infection, University of Lausanne , Lausanne , Switzerland
| |
Collapse
|
104
|
Yadava K, Bollyky P, Lawson MA. The formation and function of tertiary lymphoid follicles in chronic pulmonary inflammation. Immunology 2016; 149:262-269. [PMID: 27441396 PMCID: PMC5046054 DOI: 10.1111/imm.12649] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/01/2016] [Accepted: 07/06/2016] [Indexed: 12/13/2022] Open
Abstract
Tertiary lymphoid follicles (TLFs) can develop in the respiratory tract in response to infections or chronic inflammation. However, their functional relevance remains unclear because they are implicated in both protective and pathological responses. In contrast to homeostatic conditions, external antigens and damage to the lung tissue may drive TLF formation in inflamed lungs, and once established, the presence of pulmonary TLFs may signal the progression of chronic lung disease. This novel concept will be discussed in light of recent work in chronic obstructive pulmonary disease and how changes in the pulmonary microbiota may drive and direct TLF formation and function. We will also discuss the cellularity of TLFs at the pulmonary mucosa, with emphasis on the potential roles of lymphoid tissue inducer cells, and B- and T-cell aggregates, and will examine the function of key chemokines and cytokines including CXCL13 and interleukin-17, in the formation and maintenance of pulmonary TLFs.
Collapse
Affiliation(s)
- Koshika Yadava
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Paul Bollyky
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | |
Collapse
|
105
|
Jing F, Choi EY. Potential of Cells and Cytokines/Chemokines to Regulate Tertiary Lymphoid Structures in Human Diseases. Immune Netw 2016; 16:271-280. [PMID: 27799872 PMCID: PMC5086451 DOI: 10.4110/in.2016.16.5.271] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/22/2016] [Accepted: 08/27/2016] [Indexed: 02/06/2023] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopic lymphoid tissues involved in chronic inflammation, autoimmune diseases, transplant rejection and cancer. They exhibit almost all the characteristics of secondary lymphoid organs (SLO), which are associated with adaptive immune responses; as such, they contain organized B-cell follicles with germinal centers, distinct areas containing T cells and dendritic cells, high endothelial venules, and lymphatics. In this review, we briefly describe the formation of SLO, and describe the cellular subsets and molecular cues involved in the formation and maintenance of TLS. Finally, we discuss the associations of TLS with human diseases, especially autoimmune diseases, and the potential for therapeutic targeting.
Collapse
Affiliation(s)
- Feifeng Jing
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
106
|
Lymphoid Aggregates Remodel Lymphatic Collecting Vessels that Serve Mesenteric Lymph Nodes in Crohn Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:3066-3073. [PMID: 27746181 DOI: 10.1016/j.ajpath.2016.07.026] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 07/07/2016] [Accepted: 07/25/2016] [Indexed: 12/22/2022]
Abstract
Early pathological descriptions of Crohn disease (CD) argued for a potential defect in lymph transport; however, this concept has not been thoroughly investigated. In mice, poor healing in response to infection-induced tissue damage can cause hyperpermeable lymphatic collecting vessels in mesenteric adipose tissue that impair antigen and immune cell access to mesenteric lymph nodes (LNs), which normally sustain appropriate immunity. To investigate whether analogous changes might occur in human intestinal disease, we established a three-dimensional imaging approach to characterize the lymphatic vasculature in mesenteric tissue from controls or patients with CD. In CD specimens, B-cell-rich aggregates resembling tertiary lymphoid organs (TLOs) impinged on lymphatic collecting vessels that enter and exit LNs. In areas of creeping fat, which characterizes inflammation-affected areas of the bowel in CD, we observed B cells and apparent innate lymphoid cells that had invaded the lymphatic vessel wall, suggesting these cells may be mediators of lymphatic remodeling. Although TLOs have been described in many chronic inflammatory states, their anatomical relationship to preestablished LNs has never been revealed. Our data indicate that, at least in the CD-affected mesentery, TLOs are positioned along collecting lymphatic vessels in a manner expected to affect delivery of lymph to LNs.
Collapse
|
107
|
Tait Wojno ED, Artis D. Emerging concepts and future challenges in innate lymphoid cell biology. J Exp Med 2016; 213:2229-2248. [PMID: 27811053 PMCID: PMC5068238 DOI: 10.1084/jem.20160525] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 09/26/2016] [Indexed: 12/15/2022] Open
Abstract
Innate lymphoid cells (ILCs) are innate immune cells that are ubiquitously distributed in lymphoid and nonlymphoid tissues and enriched at mucosal and barrier surfaces. Three major ILC subsets are recognized in mice and humans. Each of these subsets interacts with innate and adaptive immune cells and integrates cues from the epithelium, the microbiota, and pathogens to regulate inflammation, immunity, tissue repair, and metabolic homeostasis. Although intense study has elucidated many aspects of ILC development, phenotype, and function, numerous challenges remain in the field of ILC biology. In particular, recent work has highlighted key new questions regarding how these cells communicate with their environment and other cell types during health and disease. This review summarizes new findings in this rapidly developing field that showcase the critical role ILCs play in directing immune responses through their ability to interact with a variety of hematopoietic and nonhematopoietic cells. In addition, we define remaining challenges and emerging questions facing the field. Finally, this review discusses the potential application of basic studies of ILC biology to the development of new treatments for human patients with inflammatory and infectious diseases in which ILCs play a role.
Collapse
Affiliation(s)
- Elia D Tait Wojno
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853 .,Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10065.,Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10065.,Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065
| |
Collapse
|
108
|
Jones GW, Hill DG, Jones SA. Understanding Immune Cells in Tertiary Lymphoid Organ Development: It Is All Starting to Come Together. Front Immunol 2016; 7:401. [PMID: 27752256 PMCID: PMC5046062 DOI: 10.3389/fimmu.2016.00401] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/21/2016] [Indexed: 01/28/2023] Open
Abstract
Tertiary lymphoid organs (TLOs) are frequently observed in tissues affected by non-resolving inflammation as a result of infection, autoimmunity, cancer, and allograft rejection. These highly ordered structures resemble the cellular composition of lymphoid follicles typically associated with the spleen and lymph node compartments. Although TLOs within tissues show varying degrees of organization, they frequently display evidence of segregated T and B cell zones, follicular dendritic cell networks, a supporting stromal reticulum, and high endothelial venules. In this respect, they mimic the activities of germinal centers and contribute to the local control of adaptive immune responses. Studies in various disease settings have described how these structures contribute to either beneficial or deleterious outcomes. While the development and architectural organization of TLOs within inflamed tissues requires homeostatic chemokines, lymphoid and inflammatory cytokines, and adhesion molecules, our understanding of the cells responsible for triggering these events is still evolving. Over the past 10–15 years, novel immune cell subsets have been discovered that have more recently been implicated in the control of TLO development and function. In this review, we will discuss the contribution of these cell types and consider the potential to develop new therapeutic strategies that target TLOs.
Collapse
Affiliation(s)
- Gareth W Jones
- Division of Infection and Immunity, Systems Immunity URI, The School of Medicine, Cardiff University , Cardiff , UK
| | - David G Hill
- Division of Infection and Immunity, Systems Immunity URI, The School of Medicine, Cardiff University , Cardiff , UK
| | - Simon A Jones
- Division of Infection and Immunity, Systems Immunity URI, The School of Medicine, Cardiff University , Cardiff , UK
| |
Collapse
|
109
|
Buettner M, Lochner M. Development and Function of Secondary and Tertiary Lymphoid Organs in the Small Intestine and the Colon. Front Immunol 2016; 7:342. [PMID: 27656182 PMCID: PMC5011757 DOI: 10.3389/fimmu.2016.00342] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/23/2016] [Indexed: 01/25/2023] Open
Abstract
The immune system of the gut has evolved a number of specific lymphoid structures that contribute to homeostasis in the face of microbial colonization and food-derived antigenic challenge. These lymphoid organs encompass Peyer’s patches (PP) in the small intestine and their colonic counterparts that develop in a programed fashion before birth. In addition, the gut harbors a network of lymphoid tissues that is commonly designated as solitary intestinal lymphoid tissues (SILT). In contrast to PP, SILT develop strictly after birth and consist of a dynamic continuum of structures ranging from small cryptopatches (CP) to large, mature isolated lymphoid follicles (ILF). Although the development of PP and SILT follow similar principles, such as an early clustering of lymphoid tissue inducer (LTi) cells and the requirement for lymphotoxin beta (LTβ) receptor-mediated signaling, the formation of CP and their further maturation into ILF is associated with additional intrinsic and environmental signals. Moreover, recent data also indicate that specific differences exist in the regulation of ILF formation between the small intestine and the colon. Importantly, intestinal inflammation in both mice and humans is associated with a strong expansion of the lymphoid network in the gut. Recent experiments in mice suggest that these structures, although they resemble large, mature ILF in appearance, may represent de novo-induced tertiary lymphoid organs (TLO). While, so far, it is not clear whether intestinal TLO contribute to the exacerbation of inflammatory pathology, it has been shown that ILF provide the critical microenvironment necessary for the induction of an effective host response upon infection with enteric bacterial pathogens. Regarding the importance of ILF for intestinal immunity, interfering with the development and maturation of these lymphoid tissues may offer novel means for manipulating the immune response during intestinal infection or inflammation.
Collapse
Affiliation(s)
- Manuela Buettner
- Central Animal Facility, Institute of Laboratory Animal Science, Hannover Medical School , Hannover , Germany
| | - Matthias Lochner
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI) , Hannover , Germany
| |
Collapse
|
110
|
Olivier BJ, Cailotto C, van der Vliet J, Knippenberg M, Greuter MJ, Hilbers FW, Konijn T, Te Velde AA, Nolte MA, Boeckxstaens GE, de Jonge WJ, Mebius RE. Vagal innervation is required for the formation of tertiary lymphoid tissue in colitis. Eur J Immunol 2016; 46:2467-2480. [PMID: 27457277 DOI: 10.1002/eji.201646370] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 06/07/2016] [Accepted: 07/19/2016] [Indexed: 01/20/2023]
Abstract
Tertiary lymphoid tissue (TLT) is lymphoid tissue that forms in adult life as a result of chronic inflammation in a tissue or organ. TLT has been shown to form in a variety of chronic inflammatory diseases, though it is not clear if and how TLT develops in the inflamed colon during inflammatory bowel disease. Here, we show that TLT develops as newly formed lymphoid tissue in the colon following dextran sulphate sodium induced colitis in C57BL/6 mice, where it can be distinguished from the preexisting colonic patches and solitary intestinal lymphoid tissue. TLT in the inflamed colon develops following the expression of lymphoid tissue-inducing chemokines and adhesion molecules, such as CXCL13 and VCAM-1, respectively, which are produced by stromal organizer cells. Surprisingly, this process of TLT formation was independent of the lymphotoxin signaling pathway, but rather under neuronal control, as we demonstrate that selective surgical ablation of vagus nerve innervation inhibits CXCL13 expression and abrogates TLT formation without affecting colitis. Sympathetic neuron denervation does not affect TLT formation. Hence, we reveal that inflammation in the colon induces the formation of TLT, which is controlled by innervation through the vagus nerve.
Collapse
Affiliation(s)
- Brenda J Olivier
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands.,Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands.,Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, ,University of Amsterdam, Amsterdam, The Netherlands
| | - Cathy Cailotto
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - Jan van der Vliet
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - Marlene Knippenberg
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Mascha J Greuter
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Francisca W Hilbers
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - Tanja Konijn
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Anje A Te Velde
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - Martijn A Nolte
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, ,University of Amsterdam, Amsterdam, The Netherlands
| | - Guy E Boeckxstaens
- Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Department of Gastroenterology, University Hospital Leuven, Leuven, Belgium
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands.
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
111
|
Neyt K, GeurtsvanKessel CH, Deswarte K, Hammad H, Lambrecht BN. Early IL-1 Signaling Promotes iBALT Induction after Influenza Virus Infection. Front Immunol 2016; 7:312. [PMID: 27579026 PMCID: PMC4985557 DOI: 10.3389/fimmu.2016.00312] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/02/2016] [Indexed: 11/13/2022] Open
Abstract
Inducible bronchus-associated lymphoid tissue (iBALT) is a long lasting tertiary lymphoid tissue that can be induced following influenza A virus (IAV) infection. Previous studies have shown that iBALT structures containing germinal center (GC) B cells protect against repeated infection by contributing locally to the cellular and humoral immune response. If we are to exploit this in vaccination strategies, we need a better understanding on how iBALT structures are induced. One hypothesis is that the strength of the initial innate response dictates induction of iBALT. In the present study, we investigated the role of interleukin (IL)-1 and IL-1R signaling on iBALT formation. Mice lacking the IL-1R had a delayed viral clearance and, thus, a prolonged exposure to viral replication, leading to increased disease severity, compared to wild-type mice. Contradictorily, iBALT formation following clearance of the virus was heavily compromised in Il1r1−/− mice. Quantification of gene induction after IAV infection demonstrated induction of IL-1α and to a much lesser extent of IL-1β. Administration of recombinant IL-1α to the lungs of wild-type mice, early but not late, after IAV infection led to more pronounced iBALT formation and an increased amount of GC B cells in the lungs. Bone marrow chimeric mice identified the stromal compartment as the crucial IL-1 responsive cell for iBALT induction. Mechanistically, Q-PCR analysis of lung homogenates revealed a strongly diminished production of CXCL13, a B cell-attracting chemokine, in Il1r−/− mice during the early innate phase of IAV infection. These experiments demonstrate that appropriate innate IL-1α–IL-1R signaling is necessary for IAV clearance and at the same time instructs the formation of organized tertiary lymphoid tissues through induction of CXCL13 early after infection. These findings are discussed in the light of recent insights on the pathogenesis of tertiary lymphoid organ formation in the lung in various diseases where the IL-1 axis is hyperactive, such as rheumatoid arthritis and COPD.
Collapse
Affiliation(s)
- Katrijn Neyt
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | | | - Kim Deswarte
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Hamida Hammad
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
112
|
McNamee EN, Rivera-Nieves J. Ectopic Tertiary Lymphoid Tissue in Inflammatory Bowel Disease: Protective or Provocateur? Front Immunol 2016; 7:308. [PMID: 27579025 PMCID: PMC4985530 DOI: 10.3389/fimmu.2016.00308] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/29/2016] [Indexed: 12/15/2022] Open
Abstract
Organized lymphoid tissues like the thymus first appeared in jawed vertebrates around 500 million years ago and have evolved to equip the host with a network of specialized sites, strategically located to orchestrate strict immune-surveillance and efficient immune responses autonomously. The gut-associated lymphoid tissues maintain a mostly tolerant environment to dampen our responses to daily dietary and microbial products in the intestine. However, when this homeostasis is perturbed by chronic inflammation, the intestine is able to develop florid organized tertiary lymphoid tissues (TLT), which heralds the onset of regional immune dysregulation. While TLT are a pathologic hallmark of Crohn's disease (CD), their role in the overall process remains largely enigmatic. A critical question remains; are intestinal TLT generated by the immune infiltrated intestine to modulate immune responses and rebuild tolerance to the microbiota or are they playing a more sinister role by generating dysregulated responses that perpetuate disease? Herein, we discuss the main theories of intestinal TLT neogenesis and focus on the most recent findings that open new perspectives to their role in inflammatory bowel disease.
Collapse
Affiliation(s)
- Eóin N McNamee
- Mucosal Inflammation Program, Department of Anesthesiology, School of Medicine, University of Colorado - Anschutz Medical Campus , Aurora, CO , USA
| | - Jesús Rivera-Nieves
- Division of Gastroenterology, Inflammatory Bowel Disease Center, San Diego VAMC, University of California San Diego , La Jolla, CA , USA
| |
Collapse
|
113
|
Microbiota, regulatory T cell subsets, and allergic disorders. ACTA ACUST UNITED AC 2016; 25:114-123. [PMID: 27656354 PMCID: PMC5016534 DOI: 10.1007/s40629-016-0118-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 01/26/2016] [Indexed: 02/07/2023]
|
114
|
Ohnmacht C. Microbiota, regulatory T cell subsets, and allergic disorders. ALLERGO JOURNAL 2016. [DOI: 10.1007/s15007-016-1137-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
115
|
Abstract
The classical model of immunity posits that the immune system reacts to pathogens and injury and restores homeostasis. Indeed, a century of research has uncovered the means and mechanisms by which the immune system recognizes danger and regulates its own activity. However, this classical model does not fully explain complex phenomena, such as tolerance, allergy, the increased prevalence of inflammatory pathologies in industrialized nations and immunity to multiple infections. In this Essay, I propose a model of immunity that is based on equilibrium, in which the healthy immune system is always active and in a state of dynamic equilibrium between antagonistic types of response. This equilibrium is regulated both by the internal milieu and by the microbial environment. As a result, alteration of the internal milieu or microbial environment leads to immune disequilibrium, which determines tolerance, protective immunity and inflammatory pathology.
Collapse
Affiliation(s)
- Gérard Eberl
- Institut Pasteur, Microenvironment and Immunity Unit, 75724 Paris, France, and the Institut National de la Santé et de la Recherche Médicale (INSERM) U1224, 75724 Paris, France
| |
Collapse
|
116
|
New insights into B cell biology in systemic lupus erythematosus and Sjögren's syndrome. Curr Opin Rheumatol 2016; 27:461-7. [PMID: 26164595 DOI: 10.1097/bor.0000000000000201] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Our understanding of the physiological and pathogenic functions of B cells in systemic lupus erythematosus (SLE) and Primary Sjögren's syndrome (pSS) continues to expand. In this review, we discuss novel insights published in the last 18 months into the roles of B cells in systemic autoimmunity. RECENT FINDINGS Data have continued to expand regarding the diverse mechanisms by which innate immune signals including Toll-like receptors (TLRs) regulate the B cell compartment. Localized B cells and long-lived plasma cells have been identified as playing an important role in target tissue including the development of ectopic lymphoid structures in kidney and salivary gland. In addition to pathogenic roles for B cells, there is mounting evidence for regulatory B cell subsets that play a protective role and new insights into the signals that regulate their development. SUMMARY The past few years have provided insights into the multiple paths by which innate immune signals can lead to B cell activation in SLE and pSS and the increasingly diverse ways in which B cells contribute to disease expression. Further understanding the imbalance between protective and pathogenic functions for B cells in disease including in understudied target tissue should yield new treatment approaches.
Collapse
|
117
|
Ohnmacht C. Tolerance to the Intestinal Microbiota Mediated by ROR(γt)(+) Cells. Trends Immunol 2016; 37:477-486. [PMID: 27255270 DOI: 10.1016/j.it.2016.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/21/2022]
Abstract
Harmless microbes colonizing the gut require the establishment of a well-equilibrated symbiosis between this microbiota and its host. However, the immune system is primed to recognize both conserved microbial patterns and foreign antigens, and therefore developed strong tolerance mechanisms to prevent potential fatal immune reactivity to symbiotic microbes. The transcription factor RAR-related orphan-like γt [ROR(γt); encoded by Rorc] plays a key role in the gut for lymphoid tissue organogenesis, development of innate lymphoid cells type 3 (ILC3s) and proinflammatory type 17 T helper (Th17) cells. Surprisingly, recent research has revealed a contribution of ROR(γt)-expressing cells in a variety of tolerance mechanisms in both the innate and adaptive immune system.
Collapse
Affiliation(s)
- Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technische Universität and Helmholtz Center Munich, Biedersteiner Strasse 29, 80802 Munich, Germany.
| |
Collapse
|
118
|
Mian F, Kandiah N, Chew M, Ashkar A, Bienenstock J, Forsythe P, Karimi K. A probiotic provides protection against acute salmonellosis in mice: Possible role of innate lymphid NKP46+ cells. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.02.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
119
|
Foxp3(+) T cells expressing RORγt represent a stable regulatory T-cell effector lineage with enhanced suppressive capacity during intestinal inflammation. Mucosal Immunol 2016; 9:444-57. [PMID: 26307665 DOI: 10.1038/mi.2015.74] [Citation(s) in RCA: 310] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/07/2015] [Indexed: 02/04/2023]
Abstract
Foxp3 (forkhead box P3 transcription factor)-expressing regulatory T cells (Tregs) are essential for immunological tolerance, best illustrated by uncontrolled effector T-cell responses and autoimmunity upon loss of Foxp3 expression. Tregs can adopt specific effector phenotypes upon activation, reflecting the diversity of functional demands in the different tissues of the body. Here, we report that Foxp3(+)CD4(+) T cells coexpressing retinoic acid-related orphan receptor-γt (RORγt), the master transcription factor for T helper type 17 (Th17) cells, represent a stable effector Treg lineage. Transcriptomic and epigenetic profiling revealed that Foxp3(+)RORγt(+) T cells display signatures of both Tregs and Th17 cells, although the degree of similarity was higher to Foxp3(+)RORγt(-) Tregs than to Foxp3(-)RORγt(+) T cells. Importantly, Foxp3(+)RORγt(+) T cells were significantly demethylated at Treg-specific epigenetic signature genes such as Foxp3, Ctla-4, Gitr, Eos, and Helios, suggesting that these cells have a stable regulatory rather than inflammatory function. Indeed, adoptive transfer of Foxp3(+)RORγt(+) T cells in the T-cell transfer colitis model confirmed their Treg function and lineage stability in vivo, and revealed an enhanced suppressive capacity as compared with Foxp3(+)RORγt(-) Tregs. Thus, our data suggest that RORγt expression in Tregs contributes to an optimal suppressive capacity during gut-specific immune responses, rendering Foxp3(+)RORγt(+) T cells as an important effector Treg subset in the intestinal system.
Collapse
|
120
|
Caballero-Franco C, Guma M, Choo MK, Sano Y, Enzler T, Karin M, Mizoguchi A, Park JM. Epithelial Control of Gut-Associated Lymphoid Tissue Formation through p38α-Dependent Restraint of NF-κB Signaling. THE JOURNAL OF IMMUNOLOGY 2016; 196:2368-76. [PMID: 26792803 DOI: 10.4049/jimmunol.1501724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/18/2015] [Indexed: 01/03/2023]
Abstract
The protein kinase p38α mediates cellular responses to environmental and endogenous cues that direct tissue homeostasis and immune responses. Studies of mice lacking p38α in several different cell types have demonstrated that p38α signaling is essential to maintaining the proliferation-differentiation balance in developing and steady-state tissues. The mechanisms underlying these roles involve cell-autonomous control of signaling and gene expression by p38α. In this study, we show that p38α regulates gut-associated lymphoid tissue (GALT) formation in a noncell-autonomous manner. From an investigation of mice with intestinal epithelial cell-specific deletion of the p38α gene, we find that p38α serves to limit NF-κB signaling and thereby attenuate GALT-promoting chemokine expression in the intestinal epithelium. Loss of this regulation results in GALT hyperplasia and, in some animals, mucosa-associated B cell lymphoma. These anomalies occur independently of luminal microbial stimuli and are most likely driven by direct epithelial-lymphoid interactions. Our study illustrates a novel p38α-dependent mechanism preventing excessive generation of epithelial-derived signals that drive lymphoid tissue overgrowth and malignancy.
Collapse
Affiliation(s)
- Celia Caballero-Franco
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129
| | - Monica Guma
- Department of Pharmacology, Laboratory of Gene Regulation and Signal Transduction, School of Medicine, University of California, San Diego, La Jolla, CA 92093; Division of Rheumatology, Allergy, and Immunology, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Min-Kyung Choo
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129
| | - Yasuyo Sano
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129
| | - Thomas Enzler
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129; Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724
| | - Michael Karin
- Department of Pharmacology, Laboratory of Gene Regulation and Signal Transduction, School of Medicine, University of California, San Diego, La Jolla, CA 92093; Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Atsushi Mizoguchi
- Department of Pathology, Molecular Pathology Unit, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129
| | - Jin Mo Park
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129;
| |
Collapse
|
121
|
Goc J, Hepworth MR, Sonnenberg GF. Group 3 innate lymphoid cells: regulating host-commensal bacteria interactions in inflammation and cancer. Int Immunol 2016; 28:43-52. [PMID: 26451009 PMCID: PMC5891988 DOI: 10.1093/intimm/dxv056] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/28/2015] [Indexed: 12/15/2022] Open
Abstract
A delicate balance exists between the mammalian immune system and normally beneficial commensal bacteria that colonize the gastrointestinal tract, which is necessary to maintain tissue homeostasis. Dysregulation of these interactions between the host and commensal bacteria is causally associated with chronic inflammation and the development of cancer. In contrast, recent reports have highlighted that commensal bacteria also play an essential role in promoting anti-tumor immune responses in several contexts, highlighting a paradox whereby interactions between the host and commensal bacteria can influence both pro- and anti-tumor immunity. Given the critical roles for group 3 innate lymphoid cells (ILC3s) in regulating inflammation, tissue repair and host-microbe interactions in the intestine, here we discuss new evidence that ILC3s may profoundly influence the development, progression and control of tumors. In this review, we provide an overview of recent advances in understanding the impact of commensal bacteria on tumorigenesis, discuss recent findings identifying ILC3s as critical regulators of host-microbe interactions and highlight the emerging role of this immune cell population in cancer and their potential implication as a therapeutic target.
Collapse
Affiliation(s)
- Jeremy Goc
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Department of Microbiology and Immunology and The Jill Robert's Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, Cornell University, 413 East 69th Street, Belfer Research Building 512, Box 190, New York, NY 10021, USA
| | - Matthew R Hepworth
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Department of Microbiology and Immunology and The Jill Robert's Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, Cornell University, 413 East 69th Street, Belfer Research Building 512, Box 190, New York, NY 10021, USA
| | - Gregory F Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Department of Microbiology and Immunology and The Jill Robert's Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, Cornell University, 413 East 69th Street, Belfer Research Building 512, Box 190, New York, NY 10021, USA
| |
Collapse
|
122
|
Bostick JW, Zhou L. Innate lymphoid cells in intestinal immunity and inflammation. Cell Mol Life Sci 2016; 73:237-52. [PMID: 26459449 PMCID: PMC11108440 DOI: 10.1007/s00018-015-2055-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 09/25/2015] [Accepted: 09/28/2015] [Indexed: 12/18/2022]
Abstract
Innate lymphoid cells (ILCs) are a new and distinct family of innate immune cells that play an important role in immunity and inflammation. In this review, we focus on the role of ILCs in mucosal tissues, especially in the gut, in health and disease. ILCs support intestinal homeostasis by protecting the intestine from pathogens, contributing to the development of gut lymphoid tissue, and helping to repair injuries. By cooperating with epithelial cells and other innate and adaptive immune cells, ILCs participate in the control of pathogens and tolerance of commensal bacteria. The development and maintenance of ILCs are influenced by nutrients and metabolites sourced from diet and/or gut bacteria. ILCs have been shown to be involved in host metabolism and to participate in various diseases of the intestine including infectious and chronic inflammatory diseases, and cancer. Thus, the elucidation of ILC biology provides an exciting potential for development of novel therapeutic means to modulate immune responses in various disease settings.
Collapse
Affiliation(s)
- John W Bostick
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Liang Zhou
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
123
|
Jones GW, Jones SA. Ectopic lymphoid follicles: inducible centres for generating antigen-specific immune responses within tissues. Immunology 2015; 147:141-51. [PMID: 26551738 PMCID: PMC4717241 DOI: 10.1111/imm.12554] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 10/28/2015] [Accepted: 11/01/2015] [Indexed: 02/06/2023] Open
Abstract
Lymphoid neogenesis is traditionally viewed as a pre‐programmed process that promotes the formation of lymphoid organs during development. Here, the spatial organization of T and B cells in lymph nodes and spleen into discrete structures regulates antigen‐specific responses and adaptive immunity following immune challenge. However, lymphoid neogenesis is also triggered by chronic or persistent inflammation. Here, ectopic (or tertiary) lymphoid organs frequently develop in inflamed tissues as a response to infection, auto‐immunity, transplantation, cancer or environmental irritants. Although these structures affect local immune responses, the contribution of these lymphoid aggregates to the underlining pathology are highly context dependent and can elicit either protective or deleterious outcomes. Here we review the cellular and molecular mechanisms responsible for ectopic lymphoid neogenesis and consider the relevance of these structures in human disease.
Collapse
Affiliation(s)
- Gareth W Jones
- Division of Infection and Immunity, The School of Medicine, Cardiff University, Cardiff, UK
| | - Simon A Jones
- Division of Infection and Immunity, The School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
124
|
Abstract
PURPOSE OF REVIEW Innate lymphoid cells (ILCs) are a newly-identified population of immune cells prevalent in, but not limited to, mucosal tissues that not only play a significant role in immune homeostasis and host defense, but also in disease pathogenesis. This review highlights the importance of type 3 ILCs (ILC3s) and their interactions with the intestinal microflora, both in maintaining gut health and in the development of inflammatory bowel disease (IBD). RECENT FINDINGS Distinct lineages of ILCs are defined based on the presence of cell surface proteins, secretion of effector cytokines and expression of master transcription factors that determine their differentiation and inflammatory behavior. These ILC subgroups mirror corresponding CD4 T-cell subsets, with which they share many phenotypic, morphologic and functional attributes. ILC3s, in particular, through direct and indirect interactions with the gut microbiota, have been identified to promote protection and maintenance of epithelial integrity, as well as to regulate intestinal inflammation and fibrosis, such as that observed in IBD. SUMMARY Gut mucosal ILCs respond to environmental cues, such as diet and microflora composition, which can shape downstream immune function. As such, ILCs represent attractive targets for the development of therapeutic modalities to maintain gut health and to potentially treat IBD.
Collapse
|
125
|
Wichner K, Stauss D, Kampfrath B, Krüger K, Müller G, Rehm A, Lipp M, Höpken UE. Dysregulated development of IL-17- and IL-21-expressing follicular helper T cells and increased germinal center formation in the absence of RORγt. FASEB J 2015; 30:761-74. [PMID: 26499265 DOI: 10.1096/fj.15-274001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 10/13/2015] [Indexed: 01/14/2023]
Abstract
Interleukin 17-producing helper T (Th17) cells have been widely defined by the lineage transcription factor retinoid-related orphan receptor (ROR)γt. Pathophysiologically, these cells play a crucial role in autoimmune diseases and have been linked to dysregulated germinal center (GC) reactions and autoantibody production. In this study, we used gene expression and flow cytometric analyses for the characterization of Rorγt(-/-) and Rorγt(-/-)Il21(RFP/+) mice to demonstrate a previously unknown transcriptional flexibility in the development of IL-17-producing Th-cell subsets. We found an accumulation of follicular Th (Tfh) cells by 5.2-fold, spontaneous 13-fold higher GC formation, decreased frequency of follicular Foxp3(+) T-regulatory (Treg) cells (50%), and a 3.4-fold increase in the number of proliferating follicular B cells in RORγt-deficient vs. wild-type mice. Dysregulated B-cell responses were associated with enhanced production of IL-17 (6.4-fold), IL-21 (2.2-fold), and B-cell-activating factor (BAFF) (2-fold) and were partially rescued by adoptive transfer of Treg cells. In an unexpected finding, we detected RORγt-independent IL-17 expression in ICOS(+)CXCR5(+)Tfh and in ICOS(+)CXCR5(-)Th cells. Based on the observed high Irf4 and Batf gene expression, we suggest that CD4(+) T-cell transcription factors other than RORγt can cooperatively induce differentiation of IL-17-producing Th cells, including Th17-like Tfh-cell subsets. We conclude that the occurrence of aberrant Tfh and follicular Treg cells support spontaneous GC formation and dysregulated B-cell responses in RORγt-deficient mice.
Collapse
Affiliation(s)
- Katharina Wichner
- *Department of Tumor Genetics and Immunogenetics and Department of Hematology, Oncology, and Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Dennis Stauss
- *Department of Tumor Genetics and Immunogenetics and Department of Hematology, Oncology, and Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Branka Kampfrath
- *Department of Tumor Genetics and Immunogenetics and Department of Hematology, Oncology, and Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Kerstin Krüger
- *Department of Tumor Genetics and Immunogenetics and Department of Hematology, Oncology, and Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Gerd Müller
- *Department of Tumor Genetics and Immunogenetics and Department of Hematology, Oncology, and Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Armin Rehm
- *Department of Tumor Genetics and Immunogenetics and Department of Hematology, Oncology, and Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Martin Lipp
- *Department of Tumor Genetics and Immunogenetics and Department of Hematology, Oncology, and Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Uta E Höpken
- *Department of Tumor Genetics and Immunogenetics and Department of Hematology, Oncology, and Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
126
|
Laukens D, Brinkman BM, Raes J, De Vos M, Vandenabeele P. Heterogeneity of the gut microbiome in mice: guidelines for optimizing experimental design. FEMS Microbiol Rev 2015; 40:117-32. [PMID: 26323480 PMCID: PMC4703068 DOI: 10.1093/femsre/fuv036] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2015] [Indexed: 02/07/2023] Open
Abstract
Targeted manipulation of the gut flora is increasingly being recognized as a means to improve human health. Yet, the temporal dynamics and intra- and interindividual heterogeneity of the microbiome represent experimental limitations, especially in human cross-sectional studies. Therefore, rodent models represent an invaluable tool to study the host–microbiota interface. Progress in technical and computational tools to investigate the composition and function of the microbiome has opened a new era of research and we gradually begin to understand the parameters that influence variation of host-associated microbial communities. To isolate true effects from confounding factors, it is essential to include such parameters in model intervention studies. Also, explicit journal instructions to include essential information on animal experiments are mandatory. The purpose of this review is to summarize the factors that influence microbiota composition in mice and to provide guidelines to improve the reproducibility of animal experiments. Given the unmet need for standardizing the experimental work flow related to gut microbial research in animals, guidelines are required to isolate true effects from confounding factors.
Collapse
Affiliation(s)
- Debby Laukens
- Department of Gastroenterology, Ghent University, B-9000 Ghent, Belgium
| | - Brigitta M Brinkman
- Inflammation Research Center, VIB, B-9052 Ghent, Belgium Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Jeroen Raes
- Center for the Biology of Disease, VIB, B-3000 Leuven, Belgium Department Microbiology and Immunology, KU Leuven, B-3000 Leuven, Belgium
| | - Martine De Vos
- Department of Gastroenterology, Ghent University, B-9000 Ghent, Belgium
| | - Peter Vandenabeele
- Inflammation Research Center, VIB, B-9052 Ghent, Belgium Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium Methusalem Program, Ghent University, B-9000 Ghent, Belgium
| |
Collapse
|
127
|
Garidou L, Pomié C, Klopp P, Waget A, Charpentier J, Aloulou M, Giry A, Serino M, Stenman L, Lahtinen S, Dray C, Iacovoni JS, Courtney M, Collet X, Amar J, Servant F, Lelouvier B, Valet P, Eberl G, Fazilleau N, Douin-Echinard V, Heymes C, Burcelin R. The Gut Microbiota Regulates Intestinal CD4 T Cells Expressing RORγt and Controls Metabolic Disease. Cell Metab 2015; 22:100-12. [PMID: 26154056 DOI: 10.1016/j.cmet.2015.06.001] [Citation(s) in RCA: 213] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 03/31/2015] [Accepted: 06/02/2015] [Indexed: 02/06/2023]
Abstract
A high-fat diet (HFD) induces metabolic disease and low-grade metabolic inflammation in response to changes in the intestinal microbiota through as-yet-unknown mechanisms. Here, we show that a HFD-derived ileum microbiota is responsible for a decrease in Th17 cells of the lamina propria in axenic colonized mice. The HFD also changed the expression profiles of intestinal antigen-presenting cells and their ability to generate Th17 cells in vitro. Consistent with these data, the metabolic phenotype was mimicked in RORγt-deficient mice, which lack IL17 and IL22 function, and in the adoptive transfer experiment of T cells from RORγt-deficient mice into Rag1-deficient mice. We conclude that the microbiota of the ileum regulates Th17 cell homeostasis in the small intestine and determines the outcome of metabolic disease.
Collapse
Affiliation(s)
- Lucile Garidou
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France.
| | - Céline Pomié
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France
| | - Pascale Klopp
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France
| | - Aurélie Waget
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France
| | - Julie Charpentier
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France
| | - Meryem Aloulou
- Université Paul Sabatier, F-31432 Toulouse, France; Centre de Physiopathologie de Toulouse Purpan, INSERM U1043, F-31300 Toulouse, France; CNRS, UMR5282, F-31300 Toulouse, France
| | - Anaïs Giry
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France
| | - Matteo Serino
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France
| | - Lotta Stenman
- Danisco Sweeteners Oy Sokeritehtaantie 20 FI-02460 Kantvik, Finland
| | - Sampo Lahtinen
- Danisco Sweeteners Oy Sokeritehtaantie 20 FI-02460 Kantvik, Finland
| | - Cedric Dray
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France
| | - Jason S Iacovoni
- Plateau de Bioinformatique et Biostatistique, INSERM UMR1048, F-31432 Toulouse, France
| | - Michael Courtney
- Vaiomer SAS, 516 Rue Pierre et Marie Curie, F-31670 Labège, France
| | - Xavier Collet
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France
| | - Jacques Amar
- Université Paul Sabatier, F-31432 Toulouse, France; Hôpital Rangueil, Département Thérapeutique, F-31059 Toulouse, France
| | - Florence Servant
- Vaiomer SAS, 516 Rue Pierre et Marie Curie, F-31670 Labège, France
| | | | - Philippe Valet
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France
| | - Gérard Eberl
- Institut Pasteur, Unité de Développement des Tissus Lymphoïdes, F-75724 Paris, France
| | - Nicolas Fazilleau
- Université Paul Sabatier, F-31432 Toulouse, France; Centre de Physiopathologie de Toulouse Purpan, INSERM U1043, F-31300 Toulouse, France; CNRS, UMR5282, F-31300 Toulouse, France
| | - Victorine Douin-Echinard
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France
| | - Christophe Heymes
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France
| | - Rémy Burcelin
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048 F-31432 Toulouse, France; Université Paul Sabatier, F-31432 Toulouse, France.
| |
Collapse
|
128
|
Noort AR, van Zoest KPM, van Baarsen LG, Maracle CX, Helder B, Papazian N, Romera-Hernandez M, Tak PP, Cupedo T, Tas SW. Tertiary Lymphoid Structures in Rheumatoid Arthritis: NF-κB-Inducing Kinase-Positive Endothelial Cells as Central Players. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1935-43. [PMID: 25963989 DOI: 10.1016/j.ajpath.2015.03.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 03/20/2015] [Accepted: 03/24/2015] [Indexed: 11/25/2022]
Abstract
Tertiary lymphoid structures (TLSs) in chronic inflammation, including rheumatoid arthritis (RA) synovial tissue (ST), often contain high endothelial venules and follicular dendritic cells (FDCs). Endothelial cell (EC)-specific lymphotoxin β (LTβ) receptor signaling is critical for the formation of lymph nodes and high endothelial venules. FDCs arise from perivascular platelet-derived growth factor receptor β(+) precursor cells (preFDCs) that require specific group 3 innate lymphoid cells (ILC3s) and LTβ for their expansion. Previously, we showed that RA ST contains ECs that express NF-κB-inducing kinase (NIK), which is pivotal in LTβ-induced noncanonical NF-κB signaling. We studied the relation between NIK(+) ECs, (pre)FDCs, and ILC3s with respect to TLSs in RA ST. TLS(+) tissues exhibited a significantly increased expression of genes involved in noncanonical NF-κB signaling, including NIK, and immunohistochemical analysis revealed that NIK was almost exclusively expressed by ECs. ILC3s were present in human RA ST in very low numbers, but not differentially in TLS(+) tissues. In contrast, TLS(+) tissues contained significantly more NIK(+) ECs and perivascular platelet-derived growth factor receptor β(+) preFDCs, which correlated significantly with the quantity of FDCs. We established a strong link between NIK(+) ECs, (pre)FDCs, and the presence of TLSs, indicating that NIK(+) ECs may not only be important orchestrators of lymph node development but also contribute to the formation of TLSs in chronic inflammation.
Collapse
Affiliation(s)
- Ae R Noort
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Katinka P M van Zoest
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Lisa G van Baarsen
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Chrissta X Maracle
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Boy Helder
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Natalie Papazian
- Department of Hematology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Monica Romera-Hernandez
- Department of Hematology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Paul P Tak
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tom Cupedo
- Department of Hematology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Sander W Tas
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
129
|
Shen P, Fillatreau S. Antibody-independent functions of B cells: a focus on cytokines. Nat Rev Immunol 2015; 15:441-51. [PMID: 26065586 DOI: 10.1038/nri3857] [Citation(s) in RCA: 332] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cytokine production by B cells is important for multiple aspects of immunity. B cell-derived cytokines, including lymphotoxin, are essential for the ontogenesis, homeostasis and activation of secondary lymphoid organs, as well as for the development of tertiary lymphoid tissues at ectopic sites. Other B cell-derived cytokines, such as interleukin-6 (IL-6), interferon-γ and tumour necrosis factor, influence the development of effector and memory CD4(+) T cell responses. Finally, B cells can regulate inflammatory immune responses, primarily through their provision of IL-10 and IL-35. This Review summarizes these various roles of cytokine-producing B cells in immunity and discusses the rational for targeting these cells in the clinic.
Collapse
Affiliation(s)
- Ping Shen
- Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Chariteplatz 1, 10117 Berlin, Germany
| | - Simon Fillatreau
- Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Chariteplatz 1, 10117 Berlin, Germany
| |
Collapse
|
130
|
Donaldson DS, Bradford BM, Artis D, Mabbott NA. Reciprocal regulation of lymphoid tissue development in the large intestine by IL-25 and IL-23. Mucosal Immunol 2015; 8:582-95. [PMID: 25249168 PMCID: PMC4424384 DOI: 10.1038/mi.2014.90] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/29/2014] [Indexed: 02/04/2023]
Abstract
Isolated lymphoid follicles (ILFs) develop after birth in the small and large intestines (SI and LI) and represent a dynamic response of the gut immune system to the microbiota. Despite their similarities, ILF development in the SI and LI differs on a number of levels. We show that unlike ILF in the SI, the microbiota inhibits ILF development in the colon as conventionalization of germ-free mice reduced colonic ILFs. From this, we identified a novel mechanism regulating colonic ILF development through the action of interleukin (IL)-25 on IL-23 and its ability to modulate T regulatory cell (Treg) differentiation. Colonic ILF develop in the absence of a number of factors required for the development of their SI counterparts and can be specifically suppressed by factors other than IL-25. However, IL-23 is the only factor identified that specifically promotes colonic ILFs without affecting SI-ILF development. Both IL-23 and ILFs are associated with inflammatory bowel disease, suggesting that disruption to this pathway may have an important role in the breakdown of microbiota-immune homeostasis.
Collapse
Affiliation(s)
- D S Donaldson
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh, UK
| | - B M Bradford
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh, UK
| | - D Artis
- Department of Microbiology and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - N A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh, UK,
| |
Collapse
|
131
|
Foo SY, Zhang V, Lalwani A, Lynch JP, Zhuang A, Lam CE, Foster PS, King C, Steptoe RJ, Mazzone SB, Sly PD, Phipps S. Regulatory T cells prevent inducible BALT formation by dampening neutrophilic inflammation. THE JOURNAL OF IMMUNOLOGY 2015; 194:4567-76. [PMID: 25810394 DOI: 10.4049/jimmunol.1400909] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 02/16/2015] [Indexed: 11/19/2022]
Abstract
Inducible BALT (iBALT) can amplify pulmonary or systemic inflammatory responses to the benefit or detriment of the host. We took advantage of the age-dependent formation of iBALT to interrogate the underlying mechanisms that give rise to this ectopic, tertiary lymphoid organ. In this study, we show that the reduced propensity for weanling as compared with neonatal mice to form iBALT in response to acute LPS exposure is associated with greater regulatory T cell expansion in the mediastinal lymph nodes. Ab- or transgene-mediated depletion of regulatory T cells in weanling mice upregulated the expression of IL-17A and CXCL9 in the lungs, induced a tissue neutrophilia, and increased the frequency of iBALT to that observed in neonatal mice. Remarkably, neutrophil depletion in neonatal mice decreased the expression of the B cell active cytokines, a proliferation-inducing ligand and IL-21, and attenuated LPS-induced iBALT formation. Taken together, our data implicate a role for neutrophils in lymphoid neogenesis. Neutrophilic inflammation is a common feature of many autoimmune diseases in which iBALT are present and pathogenic, and hence the targeting of neutrophils or their byproducts may serve to ameliorate detrimental lymphoid neogenesis in a variety of disease contexts.
Collapse
Affiliation(s)
- Shen Yun Foo
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Vivian Zhang
- School of Biomedical Sciences, University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Amit Lalwani
- School of Biomedical Sciences, University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Jason P Lynch
- School of Biomedical Sciences, University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Aowen Zhuang
- School of Biomedical Sciences, University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Chuan En Lam
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Paul S Foster
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Cecile King
- Department of Immunology, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Raymond J Steptoe
- Diamantina Institute, University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Stuart B Mazzone
- School of Biomedical Sciences, University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Peter D Sly
- Queensland Children's Medical Research Institute, University of Queensland, Herston, Queensland 4006, Australia; and Australian Infectious Diseases Research Centre, University of Queensland, St. Lucia, Queensland 4006, Australia
| | - Simon Phipps
- School of Biomedical Sciences, University of Queensland, St. Lucia, Queensland 4072, Australia; Australian Infectious Diseases Research Centre, University of Queensland, St. Lucia, Queensland 4006, Australia
| |
Collapse
|
132
|
Buckley CD, Barone F, Nayar S, Bénézech C, Caamaño J. Stromal Cells in Chronic Inflammation and Tertiary Lymphoid Organ Formation. Annu Rev Immunol 2015; 33:715-45. [DOI: 10.1146/annurev-immunol-032713-120252] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Christopher D. Buckley
- Rheumatology Research Group, Center for Translational Inflammation Research, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham B15 2WD, United Kingdom
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| | - Francesca Barone
- Rheumatology Research Group, Center for Translational Inflammation Research, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham B15 2WD, United Kingdom
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| | - Saba Nayar
- Rheumatology Research Group, Center for Translational Inflammation Research, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham B15 2WD, United Kingdom
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| | - Cecile Bénézech
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| | - Jorge Caamaño
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| |
Collapse
|
133
|
Macho-Fernandez E, Koroleva EP, Spencer CM, Tighe M, Torrado E, Cooper AM, Fu YX, Tumanov AV. Lymphotoxin beta receptor signaling limits mucosal damage through driving IL-23 production by epithelial cells. Mucosal Immunol 2015; 8:403-13. [PMID: 25183367 PMCID: PMC4364000 DOI: 10.1038/mi.2014.78] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/26/2014] [Indexed: 02/04/2023]
Abstract
The immune mechanisms regulating epithelial cell repair after injury remain poorly defined. We demonstrate here that lymphotoxin beta receptor (LTβR) signaling in intestinal epithelial cells promotes self-repair after mucosal damage. Using a conditional gene-targeted approach, we demonstrate that LTβR signaling in intestinal epithelial cells is essential for epithelial interleukin-23 (IL-23) production and protection against epithelial injury. We further show that epithelial-derived IL-23 promotes mucosal wound healing by inducing the IL-22-mediated proliferation and survival of epithelial cells and mucus production. Additionally, we identified CD4(-)CCR6(+)T-bet(-) RAR-related orphan receptor gamma t (RORγt)(+) lymphoid tissue inducer cells as the main producers of protective IL-22 after epithelial damage. Thus, our results reveal a novel role for LTβR signaling in epithelial cells in the regulation of intestinal epithelial cell homeostasis to limit mucosal damage.
Collapse
Affiliation(s)
| | | | - C M Spencer
- Trudeau Institute, Saranac Lake, New York, USA
| | - M Tighe
- Trudeau Institute, Saranac Lake, New York, USA
| | - E Torrado
- Trudeau Institute, Saranac Lake, New York, USA
| | - A M Cooper
- Trudeau Institute, Saranac Lake, New York, USA
| | - Y-X Fu
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - A V Tumanov
- Trudeau Institute, Saranac Lake, New York, USA
| |
Collapse
|
134
|
Slack E, Balmer ML, Macpherson AJ. B cells as a critical node in the microbiota-host immune system network. Immunol Rev 2015; 260:50-66. [PMID: 24942681 DOI: 10.1111/imr.12179] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mutualism with our intestinal microbiota is a prerequisite for healthy existence. This requires physical separation of the majority of the microbiota from the host (by secreted antimicrobials, mucus, and the intestinal epithelium) and active immune control of the low numbers of microbes that overcome these physical and chemical barriers, even in healthy individuals. In this review, we address how B-cell responses to members of the intestinal microbiota form a robust network with mucus, epithelial integrity, follicular helper T cells, innate immunity, and gut-associated lymphoid tissues to maintain host-microbiota mutualism.
Collapse
Affiliation(s)
- Emma Slack
- Institute for Microbiology, ETH Zürich, Zurich, Switzerland
| | | | | |
Collapse
|
135
|
Kök A, Hocqueloux L, Hocini H, Carrière M, Lefrou L, Guguin A, Tisserand P, Bonnabau H, Avettand-Fenoel V, Prazuck T, Katsahian S, Gaulard P, Thiébaut R, Lévy Y, Hüe S. Early initiation of combined antiretroviral therapy preserves immune function in the gut of HIV-infected patients. Mucosal Immunol 2015; 8:127-40. [PMID: 24985081 DOI: 10.1038/mi.2014.50] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 05/15/2014] [Indexed: 02/04/2023]
Abstract
Massive loss of lamina propria CD4(+) T cells, changes in the lymphatic architecture, and altered intestinal epithelial barrier leading to microbial translocation are the common features of HIV-1 infection and are not fully restored under combined antiretroviral therapy (cART). To better understand determinants of gut mucosal restoration, we have performed phenotypic and gene expression analyses of the gut from HIV-infected patients, naive or treated with cART initiated either at the early phase of the primary infection or later during the chronic phase. We found a depletion of T helper type 22 (Th22) and interleukin-17-producing cells in naive patients. These populations, except Th22 cells, were not restored under cART. Regulatory T cells/Th17 ratio was significantly increased in HIV-infected patients and was inversely correlated to the restoration of CD4(+) T cells but not to gut HIV DNA levels. Gene profile analysis of gut mucosal distinguished two groups of patients, which fitted with the timing of cART initiation. In their majority early, but not later treated patients, exhibited conserved intestinal lymphoid structure, epithelial barrier integrity and dendritic cell maturation pathways. Our data demonstrate that early initiation of cART helps to preserve and/or restore lymphoid gut mucosal homeostasis and provide a rationale for initiating cART during the acute phase of HIV infection.
Collapse
Affiliation(s)
- A Kök
- 1] INSERM U955, Team 16, Créteil, France [2] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France [3] Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - L Hocqueloux
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, Orléans, France
| | - H Hocini
- 1] INSERM U955, Team 16, Créteil, France [2] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | | | - L Lefrou
- Service d'Hépato-Gastro-Entérologie, CHR d'Orléans-La Source, Orléans, France
| | - A Guguin
- 1] INSERM U955, Team 16, Créteil, France [2] Plateforme de Cytométrie en flux, IMRB, UFR de Médecine, Créteil, France
| | - P Tisserand
- 1] INSERM U955, Team 16, Créteil, France [2] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - H Bonnabau
- 1] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France [2] INSERM U897 - INRIA SISTM - Univ. Bordeaux Segalen ISPED, Bordeaux, France
| | - V Avettand-Fenoel
- AP-HP, CHU Necker-Enfants Malades, Laboratoire de Virologie, Paris, France
| | - T Prazuck
- Service des Maladies Infectieuses et Tropicales, CHR d'Orléans-La Source, Orléans, France
| | - S Katsahian
- Assistance publique -Hôpitaux de Paris (AP-HP)-Hôpital Henri Mondor-Université Paris-Est Créteil, Val-de Marne, France
| | - P Gaulard
- 1] Université Paris Est Créteil, Faculté de Médecine, Créteil, France [2] INSERM U955, Team 9, Créteil, France [3] Département de Pathologie, Groupe Hospitalier Henri Mondor, AP-HP, Créteil, France
| | - R Thiébaut
- 1] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France [2] INSERM U897 - INRIA SISTM - Univ. Bordeaux Segalen ISPED, Bordeaux, France
| | - Y Lévy
- 1] INSERM U955, Team 16, Créteil, France [2] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France [3] Université Paris Est Créteil, Faculté de Médecine, Créteil, France [4] Service d'Immunologie Clinique, Groupe Hospitalier Henri Mondor, AP-HP, Créteil, France
| | - S Hüe
- 1] INSERM U955, Team 16, Créteil, France [2] Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France [3] Université Paris Est Créteil, Faculté de Médecine, Créteil, France [4] Service d'Immunologie Biologique, Groupe Hospitalier Henri Mondor, AP-HP, Créteil, France
| |
Collapse
|
136
|
Understanding host-adherent-invasive Escherichia coli interaction in Crohn's disease: opening up new therapeutic strategies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:567929. [PMID: 25580435 PMCID: PMC4279263 DOI: 10.1155/2014/567929] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/16/2014] [Accepted: 09/16/2014] [Indexed: 02/07/2023]
Abstract
A trillion of microorganisms colonize the mammalian intestine. Most of them have coevolved with the host in a symbiotic relationship and some of them have developed strategies to promote their replication in the presence of competing microbiota. Recent evidence suggests that perturbation of the microbial community favors the emergence of opportunistic pathogens, in particular adherent-invasive Escherichia coli (AIEC) that can increase incidence and severity of gut inflammation in the context of Crohn's disease (CD). This review will report the importance of AIEC as triggers of intestinal inflammation, focusing on their impact on epithelial barrier function and stimulation of mucosal inflammation. Beyond manipulation of immune response, restoration of gut microbiota as a new treatment option for CD patients will be discussed.
Collapse
|
137
|
Cao S, Feehley TJ, Nagler CR. The role of commensal bacteria in the regulation of sensitization to food allergens. FEBS Lett 2014; 588:4258-66. [PMID: 24791655 PMCID: PMC4216641 DOI: 10.1016/j.febslet.2014.04.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 04/24/2014] [Accepted: 04/24/2014] [Indexed: 12/23/2022]
Abstract
The prevalence of life-threatening anaphylactic responses to food is rising at an alarming rate. The emerging role of the gut microbiota in regulating food allergen sensitization may help explain this trend. The mechanisms by which commensal bacteria influence sensitization to dietary antigens are only beginning to be explored. We have found that a population of mucosa-associated commensal anaerobes prevents food allergen sensitization by promoting an IL-22-dependent barrier protective immune response that limits the access of food allergens to the systemic circulation. This early response is followed by an adaptive immune response mediated in part by an expansion of Foxp3(+) Tregs that fortifies the tolerogenic milieu needed to maintain non-responsiveness to food. Bacterial metabolites, such as short-chain fatty acids, may contribute to the process through their ability to promote Foxp3(+) Treg differentiation. This work suggests that environmentally induced alterations of the gut microbiota offset the regulatory signals conferred by protective bacterial species to promote aberrant responses to food. Our research presents exciting new possibilities for preventing and treating food allergies based on interventions that modulate the composition of the gut microbiota.
Collapse
Affiliation(s)
- Severine Cao
- Department of Pathology and Committee on Immunology, The University of Chicago, 924 East 57th Street, JFK R120, Chicago, IL 60637, United States
| | - Taylor J Feehley
- Department of Pathology and Committee on Immunology, The University of Chicago, 924 East 57th Street, JFK R120, Chicago, IL 60637, United States
| | - Cathryn R Nagler
- Department of Pathology and Committee on Immunology, The University of Chicago, 924 East 57th Street, JFK R120, Chicago, IL 60637, United States.
| |
Collapse
|
138
|
Kabat AM, Srinivasan N, Maloy KJ. Modulation of immune development and function by intestinal microbiota. Trends Immunol 2014; 35:507-17. [PMID: 25172617 DOI: 10.1016/j.it.2014.07.010] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/18/2014] [Accepted: 07/30/2014] [Indexed: 12/12/2022]
Abstract
The immune system must constantly monitor the gastrointestinal tract for the presence of pathogens while tolerating trillions of commensal microbiota. It is clear that intestinal microbiota actively modulate the immune system to maintain a mutually beneficial relation, but the mechanisms that maintain homeostasis are not fully understood. Recent advances have begun to shed light on the cellular and molecular factors involved, revealing that a range of microbiota derivatives can influence host immune functions by targeting various cell types, including intestinal epithelial cells, mononuclear phagocytes, innate lymphoid cells, and B and T lymphocytes. Here, we review these findings, highlighting open questions and important challenges to overcome in translating this knowledge into new therapies for intestinal and systemic immune disorders.
Collapse
Affiliation(s)
- Agnieszka M Kabat
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Naren Srinivasan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK; Immunobiology Laboratory, Cancer Research UK, London Research Institute, London, UK
| | - Kevin J Maloy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|
139
|
Lécuyer E, Rakotobe S, Lengliné-Garnier H, Lebreton C, Picard M, Juste C, Fritzen R, Eberl G, McCoy KD, Macpherson AJ, Reynaud CA, Cerf-Bensussan N, Gaboriau-Routhiau V. Segmented filamentous bacterium uses secondary and tertiary lymphoid tissues to induce gut IgA and specific T helper 17 cell responses. Immunity 2014; 40:608-20. [PMID: 24745335 DOI: 10.1016/j.immuni.2014.03.009] [Citation(s) in RCA: 243] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 01/29/2014] [Indexed: 12/12/2022]
Abstract
Segmented filamentous bacterium (SFB) is a symbiont that drives postnatal maturation of gut adaptive immune responses. In contrast to nonpathogenic E. coli, SFB stimulated vigorous development of Peyer's patches germinal centers but paradoxically induced only a low frequency of specific immunoglobulin A (IgA)-secreting cells with delayed accumulation of somatic mutations. Moreover, blocking Peyer's patch development abolished IgA responses to E. coli, but not to SFB. Indeed, SFB stimulated the postnatal development of isolated lymphoid follicles and tertiary lymphoid tissue, which substituted for Peyer's patches as inductive sites for intestinal IgA and SFB-specific T helper 17 (Th17) cell responses. Strikingly, in mice depleted of gut organized lymphoid tissue, SFB still induced a substantial but nonspecific intestinal Th17 cell response. These results demonstrate that SFB has the remarkable capacity to induce and stimulate multiple types of intestinal lymphoid tissues that cooperate to generate potent IgA and Th17 cell responses displaying only limited target specificity.
Collapse
Affiliation(s)
- Emelyne Lécuyer
- INSERM UMR1163, Laboratory of Intestinal Immunity; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Sabine Rakotobe
- INSERM UMR1163, Laboratory of Intestinal Immunity; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; INRA Micalis UMR1319, 78350 Jouy-en-Josas, France
| | - Hélène Lengliné-Garnier
- INSERM UMR1163, Laboratory of Intestinal Immunity; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; AP-HP, Department of Pediatric Gastroenterology, Hôpital Necker, 75015 Paris, France
| | - Corinne Lebreton
- INSERM UMR1163, Laboratory of Intestinal Immunity; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Marion Picard
- INSERM UMR1163, Laboratory of Intestinal Immunity; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | | | - Rémi Fritzen
- Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; INSERM UMR 1151, Institut Necker-Enfants Malades, Université Paris Descartes- Sorbonne Paris Cité, 75014 Paris, France
| | - Gérard Eberl
- Institut Pasteur, Lymphoid Tissue Development Unit, 75015 Paris, France
| | - Kathy D McCoy
- Maurice Müller Laboratories, Universitätsklinik für Viszerale Chirurgie und Medizin (UVCM), University of Bern, 3008 Bern, Switzerland
| | - Andrew J Macpherson
- Maurice Müller Laboratories, Universitätsklinik für Viszerale Chirurgie und Medizin (UVCM), University of Bern, 3008 Bern, Switzerland
| | - Claude-Agnès Reynaud
- Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; INSERM UMR 1151, Institut Necker-Enfants Malades, Université Paris Descartes- Sorbonne Paris Cité, 75014 Paris, France
| | - Nadine Cerf-Bensussan
- INSERM UMR1163, Laboratory of Intestinal Immunity; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France.
| | - Valérie Gaboriau-Routhiau
- INSERM UMR1163, Laboratory of Intestinal Immunity; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; INRA Micalis UMR1319, 78350 Jouy-en-Josas, France.
| |
Collapse
|
140
|
Pitzalis C, Jones GW, Bombardieri M, Jones SA. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat Rev Immunol 2014; 14:447-62. [PMID: 24948366 DOI: 10.1038/nri3700] [Citation(s) in RCA: 487] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ectopic lymphoid-like structures often develop at sites of inflammation where they influence the course of infection, autoimmune disease, cancer and transplant rejection. These lymphoid aggregates range from tight clusters of B cells and T cells to highly organized structures that comprise functional germinal centres. Although the mechanisms governing ectopic lymphoid neogenesis in human pathology remain poorly defined, the presence of ectopic lymphoid-like structures within inflamed tissues has been linked to both protective and deleterious outcomes in patients. In this Review, we discuss investigations in both experimental model systems and patient cohorts to provide a perspective on the formation and functions of ectopic lymphoid-like structures in human pathology, with particular reference to the clinical implications and the potential for therapeutic targeting.
Collapse
Affiliation(s)
- Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Gareth W Jones
- Cardiff Institute for Infection and Immunity, The School of Medicine, Cardiff University, The Tenovus Building, Heath Campus, Cardiff CF14 4XN, Wales, UK
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Simon A Jones
- Cardiff Institute for Infection and Immunity, The School of Medicine, Cardiff University, The Tenovus Building, Heath Campus, Cardiff CF14 4XN, Wales, UK
| |
Collapse
|
141
|
Parlato M, Yeretssian G. NOD-like receptors in intestinal homeostasis and epithelial tissue repair. Int J Mol Sci 2014; 15:9594-627. [PMID: 24886810 PMCID: PMC4100112 DOI: 10.3390/ijms15069594] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/16/2014] [Accepted: 05/20/2014] [Indexed: 12/13/2022] Open
Abstract
The intestinal epithelium constitutes a dynamic physical barrier segregating the luminal content from the underlying mucosal tissue. Following injury, the epithelial integrity is restored by rapid migration of intestinal epithelial cells (IECs) across the denuded area in a process known as wound healing. Hence, through a sequence of events involving restitution, proliferation and differentiation of IECs the gap is resealed and homeostasis reestablished. Relapsing damage followed by healing of the inflamed mucosa is a hallmark of several intestinal disorders including inflammatory bowel diseases (IBD). While several regulatory peptides, growth factors and cytokines stimulate restitution of the epithelial layer after injury, recent evidence in the field underscores the contribution of innate immunity in controlling this process. In particular, nucleotide-binding and oligomerization domain-like receptors (NLRs) play critical roles in sensing the commensal microbiota, maintaining homeostasis, and regulating intestinal inflammation. Here, we review the process of intestinal epithelial tissue repair and we specifically focus on the impact of NLR-mediated signaling mechanisms involved in governing epithelial wound healing during disease.
Collapse
Affiliation(s)
- Marianna Parlato
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Garabet Yeretssian
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
142
|
Randall TD, Mebius RE. The development and function of mucosal lymphoid tissues: a balancing act with micro-organisms. Mucosal Immunol 2014; 7:455-66. [PMID: 24569801 DOI: 10.1038/mi.2014.11] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/24/2014] [Indexed: 02/06/2023]
Abstract
Mucosal surfaces are constantly exposed to environmental antigens, colonized by commensal organisms and used by pathogens as points of entry. As a result, the immune system has devoted the bulk of its resources to mucosal sites to maintain symbiosis with commensal organisms, prevent pathogen entry, and avoid unnecessary inflammatory responses to innocuous antigens. These functions are facilitated by a variety of mucosal lymphoid organs that develop during embryogenesis in the absence of microbial stimulation as well as ectopic lymphoid tissues that develop in adults following microbial exposure or inflammation. Each of these lymphoid organs samples antigens from different mucosal sites and contributes to immune homeostasis, commensal containment, and immunity to pathogens. Here we discuss the mechanisms, mostly based on mouse studies, that control the development of mucosal lymphoid organs and how the various lymphoid tissues cooperate to maintain the integrity of the mucosal barrier.
Collapse
Affiliation(s)
- T D Randall
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham Alabama, USA
| | - R E Mebius
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
143
|
Xiao F, Yu Q, Li J, Johansson MEV, Singh AK, Xia W, Riederer B, Engelhardt R, Montrose M, Soleimani M, Tian DA, Xu G, Hansson GC, Seidler U. Slc26a3 deficiency is associated with loss of colonic HCO3 (-) secretion, absence of a firm mucus layer and barrier impairment in mice. Acta Physiol (Oxf) 2014; 211:161-75. [PMID: 24373192 DOI: 10.1111/apha.12220] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 11/08/2013] [Accepted: 12/17/2013] [Indexed: 12/11/2022]
Abstract
AIM Downregulated in adenoma (DRA, Slc26a3) is a member of the solute carrier family 26 (SLC26), family of anion transporters, which is mutated in familial chloride-losing diarrhoea (CLD). Besides Cl(-) -rich diarrhoea, CLD patients also have a higher-than-average incidence of intestinal inflammation. In a search for potential explanations for this clinical finding, we investigated colonic electrolyte transport, the mucus layer and susceptibility against dextran sodium sulphate (DSS)-induced colitis in Slc26a3(-/-) mice. METHODS HCO3 (-) secretory (JHCO3 (-) ) and fluid absorptive rates were measured by single-pass perfusion in vivo and in isolated mid-distal colonic mucosa in Ussing chambers in vitro. Colonocyte intracellular pH (pHi ) was assessed fluorometrically, the mucus layer by immunohistochemistry and colitis susceptibility by the addition of DSS to the drinking water. RESULTS HCO3 (-) secretory (JHCO3- ) and fluid absorptive rates were strongly reduced in Slc26a3(-/-) mice compared to wild-type (WT) littermates. Despite an increase in sodium/hydrogen exchanger 3 (NHE3) mRNA and protein expression, and intact acid-activation of NHE3, the high colonocyte pH in Slc26a3(-/-) mice prevented Na(+) /H(+) exchange-mediated fluid absorption in vivo. Mucin 2 (MUC2) immunohistochemistry revealed the absence of a firm mucus layer, implying that alkaline secretion and/or an absorptive flux may be necessary for optimal mucus gel formation. Slc26a3(-/-) mice were highly susceptible to DSS damage. CONCLUSIONS Deletion of DRA results in severely reduced colonic HCO3 (-) secretory rate, a loss of colonic fluid absorption, a lack of a firmly adherent mucus layer and a severely reduced colonic mucosal resistance to DSS damage. These data provide potential pathophysiological explanations for the increased susceptibility of CLD patients to intestinal inflammation.
Collapse
Affiliation(s)
- F. Xiao
- Department of Gastroenterology; Hannover Medical School; Hannover Germany
- Department of Gastroenterology; Tongji Hospital; Huazhong University of Science & Technology; Wuhan China
| | - Q. Yu
- Department of Gastroenterology; Hannover Medical School; Hannover Germany
- Department of Gastroenterology; Tongji Hospital; Huazhong University of Science & Technology; Wuhan China
| | - J. Li
- Department of Gastroenterology; Hannover Medical School; Hannover Germany
- Department of Nephrology; Tongji Hospital; Huazhong University of Science & Technology; Wuhan China
| | - M. E. V. Johansson
- Department of Medical Biochemistry; University of Gothenburg; Gothenburg Sweden
| | - A. K. Singh
- Department of Gastroenterology; Hannover Medical School; Hannover Germany
| | - W. Xia
- Department of Gastroenterology; Hannover Medical School; Hannover Germany
- School of Medicine; Key Lab of Combined Multiorgan Transplantation; The First Affiliated Hospital; Zhejiang University; Hangzhou China
| | - B. Riederer
- Department of Gastroenterology; Hannover Medical School; Hannover Germany
| | - R. Engelhardt
- Department of Gastroenterology; Hannover Medical School; Hannover Germany
| | - M. Montrose
- Center on Genetics of Transport and Epithelial Biology; University of Cincinnati; Cincinnati OH USA
| | - M. Soleimani
- Center on Genetics of Transport and Epithelial Biology; University of Cincinnati; Cincinnati OH USA
| | - D. A Tian
- Department of Gastroenterology; Tongji Hospital; Huazhong University of Science & Technology; Wuhan China
| | - G. Xu
- Department of Nephrology; Tongji Hospital; Huazhong University of Science & Technology; Wuhan China
| | - G. C. Hansson
- Department of Medical Biochemistry; University of Gothenburg; Gothenburg Sweden
| | - U. Seidler
- Department of Gastroenterology; Hannover Medical School; Hannover Germany
| |
Collapse
|
144
|
TNFα-dependent development of lymphoid tissue in the absence of RORγt⁺ lymphoid tissue inducer cells. Mucosal Immunol 2014; 7:602-14. [PMID: 24129162 PMCID: PMC4264842 DOI: 10.1038/mi.2013.79] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 08/28/2013] [Accepted: 09/09/2013] [Indexed: 02/04/2023]
Abstract
Lymphoid tissue often forms within sites of chronic inflammation. Here we report that expression of the proinflammatory cytokine tumor necrosis factor α (TNFα) drives development of lymphoid tissue in the intestine. Formation of this ectopic lymphoid tissue was not dependent on the presence of canonical RORgt(+) lymphoid tissue-inducer (LTi) cells, because animals expressing increased levels of TNFα but lacking RORgt(+) LTi cells (TNF/Rorc(gt)(-/-) mice) developed lymphoid tissue in inflamed areas. Unexpectedly, such animals developed several lymph nodes (LNs) that were structurally and functionally similar to those of wild-type animals. TNFα production by F4/80(+) myeloid cells present within the anlagen was important for the activation of stromal cells during the late stages of embryogenesis and for the activation of an organogenic program that allowed the development of LNs. Our results show that lymphoid tissue organogenesis can occur in the absence of LTi cells and suggest that interactions between TNFα-expressing myeloid cells and stromal cells have an important role in secondary lymphoid organ formation.
Collapse
|
145
|
Upadhyay V, Fu YX. Lymphotoxin organizes contributions to host defense and metabolic illness from innate lymphoid cells. Cytokine Growth Factor Rev 2014; 25:227-33. [PMID: 24411493 PMCID: PMC3999173 DOI: 10.1016/j.cytogfr.2013.12.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/15/2013] [Indexed: 01/02/2023]
Abstract
The lymphotoxin (LT)-pathway is a unique constituent branch of the Tumor Necrosis Superfamily (TNFSF). Use of LT is a critical mechanism by which fetal innate lymphoid cells regulate lymphoid organogenesis. Within recent years, adult innate lymphoid cells have been discovered to utilize this same pathway to regulate IL-22 and IL-23 production for host defense. Notably, genetic studies have linked polymorphisms in the genes encoding LTα to several phenotypes contributing to metabolic syndrome. The role of the LT-pathway may lay the foundation for a bridge between host immune response, microbiota, and metabolic syndrome. The contribution of the LT-pathway to innate lymphoid cell function and metabolic syndrome will be visited in this review.
Collapse
Affiliation(s)
- Vaibhav Upadhyay
- Committee on Immunology, University of Chicago, United States; Department of Pathology, University of Chicago, United States
| | - Yang-Xin Fu
- Committee on Immunology, University of Chicago, United States; Department of Pathology, University of Chicago, United States.
| |
Collapse
|
146
|
Lu TT, Browning JL. Role of the Lymphotoxin/LIGHT System in the Development and Maintenance of Reticular Networks and Vasculature in Lymphoid Tissues. Front Immunol 2014; 5:47. [PMID: 24575096 PMCID: PMC3920476 DOI: 10.3389/fimmu.2014.00047] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/27/2014] [Indexed: 01/08/2023] Open
Abstract
Lymphoid organs are meeting zones where lymphocytes come together and encounter antigens present in the blood and lymph or as delivered by cells migrating from the draining tissue bed. The exquisite efficiency of this process relies heavily on highly specialized anatomy to direct and position the various players. Gated entry and exit control access to these theaters and reticular networks and associated chemokines guide cells into the proper sections. Lymphoid tissues are remarkably plastic, being able to expand dramatically and then involute upon resolution of the danger. All of the reticular scaffolds and vascular and lymphatic components adapt accordingly. As such, the lymph node (LN) is a wonderful example of a physiologic remodeling process and is potentially a guide to study such elements in pathological settings such as fibrosis, chronic infection, and tumor metastasis. The lymphotoxin/LIGHT axis delivers critical differentiation signals that direct and hone differentiation of both reticular networks and the vasculature. Considerable progress has been made recently in understanding the mesenchymal differentiation pathways leading to these specialized networks and in the remodeling that occurs in reactive LNs. In this article, we will review some new advances in the area in terms of developmental, differentiation, and maintenance events mediated by this axis.
Collapse
Affiliation(s)
- Theresa T Lu
- Autoimmunity and Inflammation Program and Pediatric Rheumatology, Hospital for Special Surgery , New York, NY , USA ; Department of Microbiology and Immunology, Weill Cornell Medical College , New York, NY , USA
| | - Jeffrey L Browning
- Department of Microbiology and Section of Rheumatology, Boston University School of Medicine , Boston, MA , USA
| |
Collapse
|
147
|
Bellinger DL, Lorton D. Autonomic regulation of cellular immune function. Auton Neurosci 2014; 182:15-41. [PMID: 24685093 DOI: 10.1016/j.autneu.2014.01.006] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 01/17/2014] [Indexed: 12/21/2022]
Abstract
The nervous system and the immune system (IS) are two integrative systems that work together to detect threats and provide host defense, and to maintain/restore homeostasis. Cross-talk between the nervous system and the IS is vital for health and well-being. One of the major neural pathways responsible for regulating host defense against injury and foreign antigens and pathogens is the sympathetic nervous system (SNS). Stimulation of adrenergic receptors (ARs) on immune cells regulates immune cell development, survival, proliferative capacity, circulation, trafficking for immune surveillance and recruitment, and directs the cell surface expression of molecules and cytokine production important for cell-to-cell interactions necessary for a coordinated immune response. Finally, AR stimulation of effector immune cells regulates the activational state of immune cells and modulates their functional capacity. This review focuses on our current understanding of the role of the SNS in regulating host defense and immune homeostasis. SNS regulation of IS functioning is a critical link to the development and exacerbation of chronic immune-mediated diseases. However, there are many mechanisms that need to be further unraveled in order to develop sound treatment strategies that act on neural-immune interaction to resolve or prevent chronic inflammatory diseases, and to improve health and quality of life.
Collapse
Affiliation(s)
- Denise L Bellinger
- Department of Pathology and Human Anatomy, Loma Linda University, School of Medicine, Loma Linda, CA, 92350, USA.
| | - Dianne Lorton
- College of Arts and Sciences, Kent State University and the Kent Summa Initiative for Clinical and Translational Research, Summa Health System, Akron, OH 44304, USA
| |
Collapse
|
148
|
Kumar V. Innate lymphoid cells: New paradigm in immunology of inflammation. Immunol Lett 2014; 157:23-37. [DOI: 10.1016/j.imlet.2013.11.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 10/20/2013] [Accepted: 11/04/2013] [Indexed: 12/27/2022]
|
149
|
Uo M, Hisamatsu T, Miyoshi J, Kaito D, Yoneno K, Kitazume MT, Mori M, Sugita A, Koganei K, Matsuoka K, Kanai T, Hibi T. Mucosal CXCR4+ IgG plasma cells contribute to the pathogenesis of human ulcerative colitis through FcγR-mediated CD14 macrophage activation. Gut 2013; 62:1734-44. [PMID: 23013725 DOI: 10.1136/gutjnl-2012-303063] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Chronic inflammation characterised by IgG-producing plasma cell infiltration of colonic mucosa is a histological hallmark of ulcerative colitis (UC); however, whether its function is pathogenic or protective remains unclear. OBJECTIVE To explore the contribution of intestinal IgG plasma cells to UC pathogenesis. METHODS We isolated lamina propria mononuclear cells (LPMCs) from intestinal mucosa of UC patients and analysed the characteristics of intestinal plasma cells (expression profiles of differentiation molecules and chemokine receptors). We investigated the involvement of IgG-immune complex (IC)-Fc gamma receptor (FcγR) signalling in intestinal inflammation by examining the cytokine production by LPMCs in response to IgG-IC stimulation. RESULTS IgG plasma cells that were markedly increased in number in the inflamed mucosa of UC patients showed a distinct expression profile (CD19(+)CD27(low), CCR10(low)CXCR4(high)) compared with IgA plasma cells (CD19(+/-)CD27(high), CCR10(high)CXCR4(-/low)). In vitro IgG-IC stimulation activated intestinal CD14 macrophages that were increased in number in the inflamed mucosa of UC patients via FcγRI and FcγRII, and induced the extensive production of pro-inflammatory cytokines such as tumour necrosis factor (TNF) and interleukin-1β (IL-1β), comparable to the effect of commensal bacteria stimulation. Co-stimulation with IgG-IC and commensal bacteria increased TNF and IL-1β production more than stimulation with the latter alone. Furthermore, IgG-IC notably up-regulated the expression of TL1A, whereas commensal bacteria specifically induced IL-23. CONCLUSIONS Collectively, these results demonstrate a novel aspect of UC pathogenesis in which unique IgG plasma cells infiltrate the inflamed mucosa via CXCR4, and critically influence UC pathogenesis by exacerbating mucosal inflammation through the activation of 'pathogenic' intestinal CD14 macrophages via IgG-IC-FcγR signalling.
Collapse
Affiliation(s)
- Michihide Uo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Thaiss CA, Levy M, Suez J, Elinav E. The interplay between the innate immune system and the microbiota. Curr Opin Immunol 2013; 26:41-8. [PMID: 24556399 DOI: 10.1016/j.coi.2013.10.016] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 10/21/2013] [Accepted: 10/28/2013] [Indexed: 01/03/2023]
Abstract
The human gastrointestinal tract harbors one of the highest densities of microorganisms on earth, called the microbiota. In fact, the number of microbial cells in the intestine outnumbers the amount of human cells of the entire organism by a factor of 10. As such, a human being is more and more perceived as a super-organism consisting of a eukaryotic and a prokaryotic part. The compartment mediating the communication between both parts is the innate immune system and its various microbe-sensing pattern-recognition receptors. Co-evolution of the microbiota with the innate immune system has resulted in elaborate interdependency and feedback mechanisms by which both systems control mutual homeostasis. Here, we review the most important innate immune-microbiota interdependencies known to date. While microbial sensing by pattern-recognition receptors is required for stable microbial composition, the presence of the microbiota, in turn, is necessary for proper development and function of the immune system.
Collapse
Affiliation(s)
- Christoph A Thaiss
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Maayan Levy
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Jotham Suez
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|