1
|
Yu L, Lin F, Yu Y, Deng X, Shi X, Lu X, Lu Y, Wang D. Rehmannia glutinosa polysaccharides enhance intestinal immunity of mice through regulating the microbiota. Int J Biol Macromol 2024; 283:137878. [PMID: 39571844 DOI: 10.1016/j.ijbiomac.2024.137878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
The Rehmannia glutinosa polysaccharides (RGP) have various benefits such as enhancing immune cell activity, decreasing oxidative stress and delaying or inhibiting tumor occurrence. Although much research has been directed at understanding the role of RGP, its influence on gut immunity is largely understudied. Here, we aimed to dissect the immune-regulating effects of RGP in the mice intestines. In vivo experiments involving the oral administration of RGP to mice at dosages of 100, 200, and 400 mg/kg over seven consecutive days revealed that RGP therapy significantly increased the percentages of CD3+ T lymphocytes and CD19+ B lymphocytes in intestines and improved the integrity of the mucosal barrier. Moreover, RGP modified the gut microbiota composition by enhancing the abundance of beneficial bacteria like Lactobacillus and Akkermansia. Fecal microbiota transplantation (FMT) experiments further revealed that RGP modulated the host's intestinal immunological function by altering the gut microbiota composition. These findings indicate that RGP may control the immunological function of the intestines.
Collapse
Affiliation(s)
- Lin Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Fangzhu Lin
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yaming Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangwen Deng
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaofeng Shi
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xuanqi Lu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Lu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
2
|
Scarno G, Mazej J, Laffranchi M, Di Censo C, Mattiola I, Candelotti AM, Pietropaolo G, Stabile H, Fionda C, Peruzzi G, Brooks SR, Tsai WL, Mikami Y, Bernardini G, Gismondi A, Sozzani S, Di Santo JP, Vosshenrich CAJ, Diefenbach A, Gadina M, Santoni A, Sciumè G. Divergent roles for STAT4 in shaping differentiation of cytotoxic ILC1 and NK cells during gut inflammation. Proc Natl Acad Sci U S A 2023; 120:e2306761120. [PMID: 37756335 PMCID: PMC10556635 DOI: 10.1073/pnas.2306761120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/10/2023] [Indexed: 09/29/2023] Open
Abstract
Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1) require signal transducer and activator of transcription 4 (STAT4) to elicit rapid effector responses and protect against pathogens. By combining genetic and transcriptomic approaches, we uncovered divergent roles for STAT4 in regulating effector differentiation of these functionally related cell types. Stat4 deletion in Ncr1-expressing cells led to impaired NK cell terminal differentiation as well as to an unexpected increased generation of cytotoxic ILC1 during intestinal inflammation. Mechanistically, Stat4-deficient ILC1 exhibited upregulation of gene modules regulated by STAT5 in vivo and an aberrant effector differentiation upon in vitro stimulation with IL-2, used as a prototypical STAT5 activator. Moreover, STAT4 expression in NCR+ innate lymphocytes restrained gut inflammation in the dextran sulfate sodium-induced colitis model limiting pathogenic production of IL-13 from adaptive CD4+ T cells in the large intestine. Collectively, our data shed light on shared and distinctive mechanisms of STAT4-regulated transcriptional control in NK cells and ILC1 required for intestinal inflammatory responses.
Collapse
Affiliation(s)
- Gianluca Scarno
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
| | - Julija Mazej
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
| | - Mattia Laffranchi
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
| | - Chiara Di Censo
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
| | - Irene Mattiola
- Laboratory of Innate Immunity, Institute of Microbiology, Infectious Diseases and Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt–Universität zu Berlin, Campus Benjamin Franklin, Berlin12203, Germany
- Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, an Institute of the Leibniz Association, Berlin10117, Germany
| | - Arianna M. Candelotti
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
| | - Giuseppe Pietropaolo
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
| | - Helena Stabile
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
| | - Giovanna Peruzzi
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Rome00161, Italy
| | - Stephen R. Brooks
- Biodata Mining and Discovery Section, Office of Science and Technology, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD20892
| | - Wanxia Li Tsai
- Translational Immunology Section, Office of Science and Technology, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD20892
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo1608582, Japan
| | - Giovanni Bernardini
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
| | - Angela Gismondi
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
| | - Silvano Sozzani
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
- Istituti di Ricovero e Cura a Carattere Scientifico Neuromed, Isernia86077, Italy
| | - James P. Di Santo
- Innate Immunity Unit, Institut Pasteur, Université Paris Cité, INSERM U1223, Paris75724, France
| | | | - Andreas Diefenbach
- Laboratory of Innate Immunity, Institute of Microbiology, Infectious Diseases and Immunology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt–Universität zu Berlin, Campus Benjamin Franklin, Berlin12203, Germany
- Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, an Institute of the Leibniz Association, Berlin10117, Germany
| | - Massimo Gadina
- Translational Immunology Section, Office of Science and Technology, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD20892
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
- Istituti di Ricovero e Cura a Carattere Scientifico Neuromed, Isernia86077, Italy
| | - Giuseppe Sciumè
- Department of Molecular Medicine, Sapienza University of Rome, Rome00161, Italy
- Laboratory affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Rome00161, Italy
| |
Collapse
|
3
|
Zhang Y, Feng X, Chen J, Liu J, Wu J, Tan H, Mi Z, Rong P. Controversial role of ILC3s in intestinal diseases: A novelty perspective on immunotherapy. Front Immunol 2023; 14:1134636. [PMID: 37063879 PMCID: PMC10090672 DOI: 10.3389/fimmu.2023.1134636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
ILC3s have been identified as crucial immune regulators that play a role in maintaining host homeostasis and modulating the antitumor response. Emerging evidence supports the idea that LTi cells play an important role in initiating lymphoid tissue development, while other ILC3s can promote host defense and orchestrate adaptive immunity, mainly through the secretion of specific cytokines and crosstalk with other immune cells or tissues. Additionally, dysregulation of ILC3-mediated overexpression of cytokines, changes in subset abundance, and conversion toward other ILC subsets are closely linked with the occurrence of tumors and inflammatory diseases. Regulation of ILC3 cytokines, ILC conversion and LTi-induced TLSs may be a novel strategy for treating tumors and intestinal or extraintestinal inflammatory diseases. Herein, we discuss the development of ILCs, the biology of ILC3s, ILC plasticity, the correlation of ILC3s and adaptive immunity, crosstalk with the intestinal microenvironment, controversial roles of ILC3s in intestinal diseases and potential applications for treatment.
Collapse
Affiliation(s)
- Yunshu Zhang
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xuefei Feng
- Department of Government & Public Administration, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Juan Chen
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiahao Liu
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianmin Wu
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongpei Tan
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ze Mi
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Ze Mi, ; Pengfei Rong,
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Ze Mi, ; Pengfei Rong,
| |
Collapse
|
4
|
Sankova MV, Nikolenko VN, Sankov SV, Sinelnikov MY. SARS-CoV-2 and microbiome. AUTOIMMUNITY, COVID-19, POST-COVID19 SYNDROME AND COVID-19 VACCINATION 2023:279-337. [DOI: 10.1016/b978-0-443-18566-3.00023-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
5
|
Shi S, Ye L, Jin K, Xiao Z, Yu X, Wu W. Innate Lymphoid Cells: Emerging Players in Pancreatic Disease. Int J Mol Sci 2022; 23:3748. [PMID: 35409105 PMCID: PMC8998564 DOI: 10.3390/ijms23073748] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/19/2022] [Accepted: 03/27/2022] [Indexed: 02/07/2023] Open
Abstract
Common pancreatic diseases have caused significant economic and social burdens worldwide. The interstitial microenvironment is involved in and plays a crucial part in the occurrence and progression of pancreatic diseases. Innate lymphoid cells (ILCs), an innate population of immune cells which have only gradually entered our visual field in the last 10 years, play an important role in maintaining tissue homeostasis, regulating metabolism, and participating in regeneration and repair. Recent evidence indicates that ILCs in the pancreas, as well as in other tissues, are also key players in pancreatic disease and health. Herein, we examined the possible functions of different ILC subsets in common pancreatic diseases, including diabetes mellitus, pancreatitis and pancreatic cancer, and discussed the potential practical implications of the relevant findings for future further treatment of these pancreatic diseases.
Collapse
Affiliation(s)
- Saimeng Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (S.S.); (L.Y.); (K.J.); (Z.X.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Longyun Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (S.S.); (L.Y.); (K.J.); (Z.X.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (S.S.); (L.Y.); (K.J.); (Z.X.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Zhiwen Xiao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (S.S.); (L.Y.); (K.J.); (Z.X.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (S.S.); (L.Y.); (K.J.); (Z.X.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Weiding Wu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (S.S.); (L.Y.); (K.J.); (Z.X.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| |
Collapse
|
6
|
Sibilio A, Suñer C, Fernández-Alfara M, Martín J, Berenguer A, Calon A, Chanes V, Millanes-Romero A, Fernández-Miranda G, Batlle E, Fernández M, Méndez R. Immune translational control by CPEB4 regulates intestinal inflammation resolution and colorectal cancer development. iScience 2022; 25:103790. [PMID: 35243213 PMCID: PMC8859527 DOI: 10.1016/j.isci.2022.103790] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/05/2021] [Accepted: 01/12/2022] [Indexed: 12/05/2022] Open
Abstract
Upon tissue injury, cytokine expression reprogramming transiently remodels the inflammatory immune microenvironment to activate repair processes and subsequently return to homeostasis. However, chronic inflammation induces permanent changes in cytokine production which exacerbate tissue damage and may even favor tumor development. Here, we address the contribution of post-transcriptional regulation, by the RNA-binding protein CPEB4, to intestinal immune homeostasis and its role in inflammatory bowel diseases (IBD) and colorectal cancer (CRC) development. We found that intestinal damage induces CPEB4 expression in adaptive and innate immune cells, which is required for the translation of cytokine mRNA(s) such as the one encoding interleukin-22. Accordingly, CPEB4 is required for the development of gut-associated lymphoid tissues and to maintain intestinal immune homeostasis, mediating repair and remodeling after acute inflammatory tissue damage and promoting the resolution of intestinal inflammation. CPEB4 is chronically overexpressed in inflammatory cells in patients with IBD and in CRC, favoring tumor development. CPEB4 is overexpressed in Th17 and ILC3 cells upon intestinal barrier damage CPEB4 is required for Il-22 mRNA translation and IL-22 expression CPEB4 promotes tissue repair in acute transient inflammation In chronic inflammation CPEB4 exacerbates intestinal pathology and promotes tumor growth
Collapse
Affiliation(s)
- Annarita Sibilio
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Clara Suñer
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Marcos Fernández-Alfara
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Judit Martín
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Antonio Berenguer
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Alexandre Calon
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Veronica Chanes
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Alba Millanes-Romero
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Gonzalo Fernández-Miranda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | | | - Raúl Méndez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
7
|
Zheng M, Zhu J. Innate Lymphoid Cells and Intestinal Inflammatory Disorders. Int J Mol Sci 2022; 23:1856. [PMID: 35163778 PMCID: PMC8836863 DOI: 10.3390/ijms23031856] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 12/27/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a population of lymphoid cells that do not express T cell or B cell antigen-specific receptors. They are largely tissue-resident and enriched at mucosal sites to play a protective role against pathogens. ILCs mimic the functions of CD4 T helper (Th) subsets. Type 1 innate lymphoid cells (ILC1s) are defined by the expression of signature cytokine IFN-γ and the master transcription factor T-bet, involving in the type 1 immune response; ILC2s are characterized by the expression of signature cytokine IL-5/IL-13 and the master transcription factor GATA3, participating in the type 2 immune response; ILC3s are RORγt-expressing cells and are capable of producing IL-22 and IL-17 to maintain intestinal homeostasis. The discovery and investigation of ILCs over the past decades extends our knowledge beyond classical adaptive and innate immunology. In this review, we will focus on the roles of ILCs in intestinal inflammation and related disorders.
Collapse
Affiliation(s)
- Mingzhu Zheng
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Microbiology and Immunology, Southeast University, Nanjing 210009, China
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Műzes G, Bohusné Barta B, Sipos F. Colitis and Colorectal Carcinogenesis: The Focus on Isolated Lymphoid Follicles. Biomedicines 2022; 10:226. [PMID: 35203436 PMCID: PMC8869724 DOI: 10.3390/biomedicines10020226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/09/2022] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
Gut-associated lymphoid tissue is one of the most diverse and complex immune compartments in the human body. The subepithelial compartment of the gut consists of immune cells of innate and adaptive immunity, non-hematopoietic mesenchymal cells, and stem cells of different origins, and is organized into secondary (and even tertiary) lymphoid organs, such as Peyer's patches, cryptopatches, and isolated lymphoid follicles. The function of isolated lymphoid follicles is multifaceted; they play a role in the development and regeneration of the large intestine and the maintenance of (immune) homeostasis. Isolated lymphoid follicles are also extensively associated with the epithelium and its conventional and non-conventional immune cells; hence, they can also function as a starting point or maintainer of pathological processes such as inflammatory bowel diseases or colorectal carcinogenesis. These relationships can significantly affect both physiological and pathological processes of the intestines. We aim to provide an overview of the latest knowledge of isolated lymphoid follicles in colonic inflammation and colorectal carcinogenesis. Further studies of these lymphoid organs will likely lead to an extended understanding of how immune responses are initiated and controlled within the large intestine, along with the possibility of creating novel mucosal vaccinations and ways to treat inflammatory bowel disease or colorectal cancer.
Collapse
Affiliation(s)
| | | | - Ferenc Sipos
- Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary; (G.M.); (B.B.B.)
| |
Collapse
|
9
|
Pierre N, Salée C, Vieujean S, Bequet E, Merli AM, Siegmund B, Meuwis MA, Louis E. Review article: distinctions between ileal and colonic Crohn's disease: from physiology to pathology. Aliment Pharmacol Ther 2021; 54:779-791. [PMID: 34297423 DOI: 10.1111/apt.16536] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/15/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ileal and colonic Crohn's disease seem to be two separate entities. AIMS To describe the main physiological distinctions between the small and the large intestine and to analyse the differences between ileal and colonic Crohn's disease. METHODS The relevant literature was critically examined and synthesised. RESULTS The small and large intestine have fundamental distinctions (anatomy, cellular populations, immune defence, microbiota). The differences between ileal and colonic Crohn's disease are highlighted by a heterogeneous body of evidence including clinical features (natural history of the disease, efficacy of treatments, and monitoring), epidemiological data (smoking status, age, gender) and biological data (genetics, microbiota, immunity, mesenteric fat). However, the contribution of these factors to disease location remains poorly understood. CONCLUSION The classification of ileal and colonic Crohn's disease as distinct subphenotypes is well supported by the literature. Understanding of these differences could be exploited to develop more individualised patient care.
Collapse
Affiliation(s)
- Nicolas Pierre
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium
| | - Catherine Salée
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium
| | - Sophie Vieujean
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| | - Emeline Bequet
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Liège University Hospital, Liège, Belgium
| | - Angela-Maria Merli
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium
| | - Britta Siegmund
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Marie-Alice Meuwis
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| | - Edouard Louis
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| |
Collapse
|
10
|
Zheng M, Mao K, Fang D, Li D, Lyu J, Peng D, Chen X, Cannon N, Hu G, Han J, Zhao K, Chen W, Zhu J. B cell residency but not T cell-independent IgA switching in the gut requires innate lymphoid cells. Proc Natl Acad Sci U S A 2021; 118:e2106754118. [PMID: 34187897 PMCID: PMC8271577 DOI: 10.1073/pnas.2106754118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Immunoglobulin A (IgA)-producing plasma cells derived from conventional B cells in the gut play an important role in maintaining the homeostasis of gut flora. Both T cell-dependent and T cell-independent IgA class switching occurs in the lymphoid structures in the gut, whose formation depends on lymphoid tissue inducers (LTis), a subset of innate lymphoid cells (ILCs). However, our knowledge on the functions of non-LTi helper-like ILCs, the innate counter parts of CD4 T helper cells, in promoting IgA production is still limited. By cell adoptive transfer and utilizing a unique mouse strain, we demonstrated that the generation of IgA-producing plasma cells from B cells in the gut occurred efficiently in the absence of both T cells and helper-like ILCs and without engaging TGF-β signaling. Nevertheless, B cell recruitment and/or retention in the gut required functional NKp46-CCR6+ LTis. Therefore, while CCR6+ LTis contribute to the accumulation of B cells in the gut through inducing lymphoid structure formation, helper-like ILCs are not essential for the T cell-independent generation of IgA-producing plasma cells.
Collapse
Affiliation(s)
- Mingzhu Zheng
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Kairui Mao
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Difeng Fang
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Dan Li
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, People's Republic of China
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou 215004, People's Republic of China
| | - Jun Lyu
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, People's Republic of China
| | - Dingkang Peng
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
- The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Xi Chen
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Nikki Cannon
- Bioinformatics Core, West Virginia University, Morgantown, WV 26506
| | - Gangqing Hu
- Bioinformatics Core, West Virginia University, Morgantown, WV 26506
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
| | - Jiajia Han
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD 20892
| | - Keji Zhao
- Laboratory of Epigenome Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892
| | - Wanjun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD 20892
| | - Jinfang Zhu
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892;
| |
Collapse
|
11
|
Bao Q, Guo XX, Cao C, Li QY, Sun L, Ye XY, Li LY, Dong JC, Gao YF, Chen HX, Li CW. Presence of Tertiary Lymphoid Organ in Nasal Inverted Papilloma Is Correlated with Eosinophil Infiltration and Local Immunoglobulin Production. Int Arch Allergy Immunol 2020; 182:350-359. [PMID: 33207352 DOI: 10.1159/000510966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/18/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Nasal inverted papilloma (NIP) is a benign tumour with multiple inflammatory cell infiltration. Tertiary lymphoid organs (TLOs) support local antibody production and play important roles in airway inflammation. However, the evidence of TLOs and local immunoglobulins in NIP has not been reported yet. We investigated the presence of TLOs and immunoglobulins in NIP tissues and their association with the clinical-pathological characteristics of NIPs. METHODS We analyzed the occurrence and composition of TLOs and local immunoglobulins by immunohistochemistry and evaluated the lymph organogenesis associated genes and cytokines by quantitative qPCR and Luminex assays, respectively, in papilloma tissues from 84 NIP cases. RESULTS TLOs were present in 54% (45/84) of the NIP patients but not in control subjects. TLOs were composed of T cells, B cells, follicular dendritic cells, macrophages, and natural killer cells. Compared to NIP tissues without TLOs, tissues with TLOs showed significantly higher eosinophil infiltration levels (3.5-fold), elevation of lymphorganogenic genes (CXCL12, CXCL13, CCL20, CCL21, CD21L, and lymphotoxin alpha and beta), and increased Th17 (IL-21, IL-22, and GM-CSF) and Th2 (IL-5 and IL-13) cytokine production. Moreover, NIP with TLOs demonstrated a higher number of follicular T helper cells and immunoglobulin-producing plasma cells (CD138+ IgA+, CD138+ IgM+, CD138+ IgE+, and CD138+ IgG+) than those without TLOs, and these antibody-producing cells were positively correlated with the eosinophil number. CONCLUSION The high frequency of TLOs and excess local immunoglobulin production are associated with an eosinophilic and Th2 skew microenvironment in the NIP mucosa, which would contribute to an important immunopathogenic response during NIP pathogenesis.
Collapse
Affiliation(s)
- Qing Bao
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xue-Xue Guo
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chen Cao
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qian-Ying Li
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lin Sun
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Yan Ye
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Yue Li
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun-Chao Dong
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yi-Fang Gao
- Organ Transplantation Centre, Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - He-Xin Chen
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chun-Wei Li
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China,
| |
Collapse
|
12
|
The Immune Phenotype of Isolated Lymphoid Structures in Non-Tumorous Colon Mucosa Encrypts the Information on Pathobiology of Metastatic Colorectal Cancer. Cancers (Basel) 2020; 12:cancers12113117. [PMID: 33113874 PMCID: PMC7692185 DOI: 10.3390/cancers12113117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/22/2020] [Accepted: 10/19/2020] [Indexed: 01/26/2023] Open
Abstract
Simple Summary Today, the presence of well-organized functional structures of immune cells at tumor sites, known as ectopic lymphoid structures, and their strong association with patient survival have been reported in more than ten different cancer types. We aimed to investigate whether there is a link between the patient-specific characteristics of pre-formed isolated lymphoid structures in non-tumorous colon tissue and the disease pathobiology for patients with metastatic colorectal cancer. The study employed a powerful approach of quantitative tissue image cytometry to compare lymphoid structures of different anatomical locations within the same patients. We showed that the properties of isolated lymphoid structures in non-tumorous colon tissue predefine the immune phenotype of ectopic lymphoid structures at primary and metastatic sites. We discovered that B-cell-enriched and highly proliferative lymphoid structures are prognostic towards an improved clinical outcome. The knowledge gained from this study expands our understanding of tumor-immune interactions and draws particular attention to the anti-tumor immune response guided by isolated lymphoid structures outside of tumor tissue. Abstract The gut-associated lymphoid tissue represents an integral part of the immune system. Among the powerful players of the mucosa-associated lymphoid tissue are isolated lymphoid structures (ILSs), which as information centers, drive the local (and systemic) adaptive immune responses. Germinal center reactions, taking place within ILSs, involve the coordinated action of various immune cell types with a central role given to B cells. In the current study, we aimed at dissecting the impact of ILSs within non-tumorous colon tissue (NT) on the pathobiology of colorectal cancer (CRC) with metastasis in the liver (CRCLM). In particular, we focused on the immune phenotypes of ILSs and ectopic lymphoid structures (ELSs), built up at matching primary and metastatic tumor sites. We implemented an integrative analysis strategy on the basis of tissue image cytometry and clonality assessment to explore the immune phenotype of ILS/ELS at three tissue entities: NT, CRC, and CRCLM (69 specimens in total). Applying a panel of lineage markers used for immunostaining, we characterized and compared the anatomical features, the cellular composition, the activation, and proliferation status of ILSs and ELSs, and assessed the clinical relevance of staining-derived data sets. Our major discovery was that ILS characteristics at the NT site predefine the immune phenotype of ELSs at CRC and CRCLM. Thereby, B-cell-enriched (CD20) and highly proliferative (Ki67) ILSs and ELSs were found to be associated with improved clinical outcome in terms of survival and enabled patient stratification into risk groups. Moreover, the data revealed a linkage between B-cell clonality at the NT site and the metastatic characteristics of the tumor in the distant liver tissue. Consolidation of immunostaining-based findings with the results of compendium-wide transcriptomic analysis furthermore proposed CD27 as a novel marker of T follicular helper cells within lymphoid structures. Overall, the study nominates the ILS immune phenotype as a novel prognostic marker for patients with metastatic CRC.
Collapse
|
13
|
Sabihi M, Böttcher M, Pelczar P, Huber S. Microbiota-Dependent Effects of IL-22. Cells 2020; 9:E2205. [PMID: 33003458 PMCID: PMC7599675 DOI: 10.3390/cells9102205] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Cytokines are important contributors to immune responses against microbial and environmental threats and are of particular importance at epithelial barriers. These interfaces are continuously exposed to external factors and thus require immune components to both protect the host from pathogen invasion and to regulate overt inflammation. Recently, substantial efforts have been devoted to understanding how cytokines act on certain cells at barrier sites, and why the dysregulation of immune responses may lead to pathogenesis. In particular, the cytokine IL-22 is involved in preserving an intact epithelium, maintaining a balanced microbiota and a functioning defense system against external threats. However, a tight regulation of IL-22 is generally needed, since uncontrolled IL-22 production can lead to the progression of autoimmunity and cancer. Our aim in this review is to summarize novel findings on IL-22 and its interactions with specific microbial stimuli, and subsequently, to understand their contributions to the function of IL-22 and the clinical outcome. We particularly focus on understanding the detrimental effects of dysregulated control of IL-22 in certain disease contexts.
Collapse
Affiliation(s)
| | | | | | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; (M.S.); (M.B.); (P.P.)
| |
Collapse
|
14
|
Jiao Y, Wu L, Huntington ND, Zhang X. Crosstalk Between Gut Microbiota and Innate Immunity and Its Implication in Autoimmune Diseases. Front Immunol 2020; 11:282. [PMID: 32153586 PMCID: PMC7047319 DOI: 10.3389/fimmu.2020.00282] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
The emerging concept of microbiota contributing to local mucosal homeostasis has fueled investigation into its specific role in immunology. Gut microbiota is mostly responsible for maintaining the balance between host defense and immune tolerance. Dysbiosis of gut microbiota has been shown to be related to various alterations of the immune system. This review focuses on the reciprocal relationship between gut microbiota and innate immunity compartment, with emphasis on gut-associated lymphoid tissue, innate lymphoid cells, and phagocytes. From a clinical perspective, the review gives a possible explanation of how the “gut microbiota—innate immunity” axis might contribute to the pathogenesis of autoimmune diseases like rheumatoid arthritis, spondyloarthritis, and systemic lupus erythematosus.
Collapse
Affiliation(s)
- Yuhao Jiao
- The Ministry of Education Key Laboratory, Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,School of Medicine, Tsinghua University, Beijing, China
| | - Li Wu
- Institute for Immunology, Tsinghua University, Beijing, China.,Tsinghua-Peking Joint Centre for Life Sciences, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Nicholas D Huntington
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Xuan Zhang
- The Ministry of Education Key Laboratory, Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Immunology Centre, Medical Epigenetics Research Centre, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
15
|
Willard-Mack CL, Elmore SA, Hall WC, Harleman J, Kuper CF, Losco P, Rehg JE, Rühl-Fehlert C, Ward JM, Weinstock D, Bradley A, Hosokawa S, Pearse G, Mahler BW, Herbert RA, Keenan CM. Nonproliferative and Proliferative Lesions of the Rat and Mouse Hematolymphoid System. Toxicol Pathol 2020; 47:665-783. [PMID: 31526133 DOI: 10.1177/0192623319867053] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The INHAND Project (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions in Rats and Mice) is a joint initiative of the Societies of Toxicologic Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP), and North America (STP) to develop an internationally accepted nomenclature for proliferative and nonproliferative changes in rats and mice. The purpose of this publication is to provide a standardized nomenclature for classifying changes observed in the hematolymphoid organs, including the bone marrow, thymus, spleen, lymph nodes, mucosa-associated lymphoid tissues, and other lymphoid tissues (serosa-associated lymphoid clusters and tertiary lymphoid structures) with color photomicrographs illustrating examples of the lesions. Sources of material included histopathology databases from government, academia, and industrial laboratories throughout the world. Content includes spontaneous lesions as well as lesions induced by exposure to test materials. The nomenclature for these organs is divided into 3 terminologies: descriptive, conventional, and enhanced. Three terms are listed for each diagnosis. The rationale for this approach and guidance for its application to toxicologic pathology are described in detail below.
Collapse
Affiliation(s)
| | - Susan A Elmore
- Thymus subgroup lead.,National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | | - Johannes Harleman
- Lymph node subgroup lead.,Neoplasm subgroup leads.,Independent Consultant, Darmstadt, Germany
| | - C Frieke Kuper
- Associated lymphoid organs subgroup lead.,Independent Consultant, Utrecht, the Netherlands
| | - Patricia Losco
- General hematolymphoid subgroup lead.,Independent Consultant, West Chester, PA, USA
| | - Jerold E Rehg
- Spleen subgroup leads.,Neoplasm subgroup leads.,Saint Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Jerrold M Ward
- Spleen subgroup leads.,Neoplasm subgroup leads.,Global VetPathology, Montgomery Village, MD, USA
| | | | - Alys Bradley
- Charles River Laboratories, Tranent, Scotland, United Kingdom
| | - Satoru Hosokawa
- Eisai Co, Ltd, Drug Safety Research Laboratories, Ibaraki, Japan
| | | | - Beth W Mahler
- Experimental Pathology Laboratories, Research Triangle Park, NC, USA
| | - Ronald A Herbert
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | |
Collapse
|
16
|
Fan H, Wang A, Wang Y, Sun Y, Han J, Chen W, Wang S, Wu Y, Lu Y. Innate Lymphoid Cells: Regulators of Gut Barrier Function and Immune Homeostasis. J Immunol Res 2019; 2019:2525984. [PMID: 31930146 PMCID: PMC6942837 DOI: 10.1155/2019/2525984] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 09/17/2019] [Indexed: 02/08/2023] Open
Abstract
Innate lymphoid cells (ILCs), identified in the early years of this century as a new class of leukocyte family unlike the B or T lymphocytes, play a unique role bridging the innate and adaptive immune responses in mucosal immunity. Their origin, differentiation, and activation process and functions have caught global interest. Recently, accumulating evidence supports that ILCs are vital regulators for gastrointestinal mucosal homeostasis through interactions with other structural and stromal cells in gut epithelial barriers. This review will explore the functions of ILCs and other cells in maintaining gut homeostasis and feature the crosstalk between ILCs with other cells and potential pharmacotherapy targeting ILCs applicable in intestinal innate immunity.
Collapse
Affiliation(s)
- Hui Fan
- Jiangsu Key Laboratory for Efficacy and Safety Evaluation of Chinese Material Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Efficacy and Safety Evaluation of Chinese Material Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuan Wang
- Jiangsu Key Laboratory for Efficacy and Safety Evaluation of Chinese Material Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ye Sun
- Jiangsu Key Laboratory for Efficacy and Safety Evaluation of Chinese Material Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jing Han
- Jiangsu Key Laboratory for Efficacy and Safety Evaluation of Chinese Material Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wenxing Chen
- Jiangsu Key Laboratory for Efficacy and Safety Evaluation of Chinese Material Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shijun Wang
- Shandong Co-Innovation Center of TCM Formula, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Shandong 250035, China
| | - Yuanyuan Wu
- Jiangsu Key Laboratory for Efficacy and Safety Evaluation of Chinese Material Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yin Lu
- Jiangsu Key Laboratory for Efficacy and Safety Evaluation of Chinese Material Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
17
|
Zhang B, Gu J, Liu J, Huang B, Li J. Fecal Microbiota Taxonomic Shifts in Chinese Multiple Myeloma Patients Analyzed by Quantitative Polimerase Chain Reaction (QPCR) and 16S rRNA High-Throughput Sequencing. Med Sci Monit 2019; 25:8269-8280. [PMID: 31678982 PMCID: PMC6855177 DOI: 10.12659/msm.919988] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Increasing evidence has suggested that gut flora play an important role in tumor progression and prognosis. However, the relationship between fecal microbiota and hematologic malignancy requires further investigation. This study aimed to characterize the relationship of the fecal microbial community in multiple myeloma (MM) patients. Material/Methods A total of 40 MM patients and healthy controls (n=17) were retrospectively collected from the First Affiliated Hospital of Sun Yat-sen University between October 2018 and May 2019. The fecal samples were collected for 16S rRNA high-throughput sequencing for the fecal microbial community, as well as diversity and correlation analysis. Furthermore, 21 MM patients and their family members were selected for the matched pair analysis to confirm the fecal microbiota taxonomic changes by qRT-PCR assay. Results Diversity analysis showed that diversity measured by Shannon index was lower in MM patients compared with healthy controls. At the phylum level, higher abundances of Proteobacteria but lower abundances of Actinobacteria were identified in the MM group in comparison with the healthy control group. At the genus level, the proportion of Bacteroides, Faecalibacterium, and Roseburia was significantly higher in the MM group. The matched pair analysis showed that Pseudomonas aeruginosa and Faecalibacterium were significantly more abundant in the MM group. Further analysis on prognostic risk factors revealed that the Faecalibacterium prausnitzii level was significantly correlated with ISS stage. Conclusions Our study highlights the imbalanced composition and diversity of the gastrointestinal microbiome in MM patients, which could be further used as a potential biomarker for MM risk screening, therapeutic strategies, and prognostic prediction.
Collapse
Affiliation(s)
- Bingqing Zhang
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Jingli Gu
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Junru Liu
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Beihui Huang
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Juan Li
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
18
|
Abdellatif AM, Sarvetnick NE. Current understanding of the role of gut dysbiosis in type 1 diabetes. J Diabetes 2019; 11:632-644. [PMID: 30864231 DOI: 10.1111/1753-0407.12915] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/13/2019] [Accepted: 03/11/2019] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disorder that results from destruction of the insulin-producing pancreatic β-cells. The disease mainly affects juveniles. Changes in the composition of the gut microbiota (dysbiosis) and changes in the properties of the gut barrier have been documented in T1D subjects. Because these factors affect immune system functions, they are likely to play a role in disease pathogenesis. However, their exact role is currently not fully understood and is under intensive investigation. In this article we discuss recent advancements depicting the role of intestinal dysbiosis on immunity and autoimmunity in T1D. We also discuss therapies aimed at maintaining a healthy gut barrier as prevention strategies for T1D.
Collapse
Affiliation(s)
- Ahmed M Abdellatif
- Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, Nebraska
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Nora E Sarvetnick
- Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, Nebraska
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
19
|
Basmaciyan L, Bon F, Paradis T, Lapaquette P, Dalle F. " Candida Albicans Interactions With The Host: Crossing The Intestinal Epithelial Barrier". Tissue Barriers 2019; 7:1612661. [PMID: 31189436 PMCID: PMC6619947 DOI: 10.1080/21688370.2019.1612661] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 02/08/2023] Open
Abstract
Formerly a commensal organism of the mucosal surfaces of most healthy individuals, Candida albicans is an opportunistic pathogen that causes infections ranging from superficial to the more life-threatening disseminated infections, especially in the ever-growing population of vulnerable patients in the hospital setting. In these situations, the fungus takes advantage of its host following a disturbance in the host defense system and/or the mucosal microbiota. Overwhelming evidence suggests that the gastrointestinal tract is the main source of disseminated C. albicans infections. Major risk factors for disseminated candidiasis include damage to the mucosal intestinal barrier, immune dysfunction, and dysbiosis of the resident microbiota. A better understanding of C. albicans' interaction with the intestinal epithelial barrier will be useful for designing future therapies to avoid systemic candidiasis. In this review, we provide an overview of the current knowledge regarding the mechanisms of pathogenicity that allow the fungus to reach and translocate the gut barrier.
Collapse
Affiliation(s)
- Louise Basmaciyan
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Fabienne Bon
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Tracy Paradis
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Pierre Lapaquette
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Frédéric Dalle
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| |
Collapse
|
20
|
Kuo SH, Wu MS, Yeh KH, Lin CW, Hsu PN, Chen LT, Cheng AL. Novel Insights of Lymphomagenesis of Helicobacter pylori-Dependent Gastric Mucosa-Associated Lymphoid Tissue Lymphoma. Cancers (Basel) 2019; 11:547. [PMID: 30999581 PMCID: PMC6520890 DOI: 10.3390/cancers11040547] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/09/2019] [Accepted: 04/12/2019] [Indexed: 02/06/2023] Open
Abstract
Gastric mucosa-associated lymphoid tissue (MALT) lymphoma is the most common subtype of gastric lymphoma. Most gastric MALT lymphomas are characterized by their association with the Helicobacter pylori (HP) infection and are cured by first-line HP eradication therapy (HPE). Several studies have been conducted to investigate why most gastric MALT lymphomas remain localized, are dependent on HP infection, and show HP-specific intratumoral T-cells (e.g., CD40-mediated signaling, T-helper-2 (Th2)-type cytokines, chemokines, costimulatory molecules, and FOXP3+ regulatory T-cells) and their communication with B-cells. Furthermore, the reason why the antigen stimuli of these intratumoral T-cells with tonic B-cell receptor signaling promote lymphomagenesis of gastric MALT lymphoma has also been investigated. In addition to the aforementioned mechanisms, it has been demonstrated that the translocated HP cytotoxin-associated gene A (CagA) can promote B-cell proliferation through the activation of Src homology-2 domain-containing phosphatase (SHP-2) phosphorylation-dependent signaling, extracellular-signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (MAPK), B-cell lymphoma (Bcl)-2, and Bcl-xL. Furthermore, the expression of CagA and these CagA-signaling molecules is closely associated with the HP-dependence of gastric MALT lymphomas (completely respond to first-line HPE). In this article, we summarize evidence of the classical theory of HP-reactive T-cells and the new paradigm of direct interaction between HP and B-cells that contributes to the HP-dependent lymphomagenesis of gastric MALT lymphomas. Although the role of first-line HPE in the treatment of HP-negative gastric MALT lymphoma remains uncertain, several case series suggest that a proportion of HP-negative gastric MALT lymphomas remains antibiotic-responsive and is cured by HPE. Considering the complicated interaction between microbiomes and the genome/epigenome, further studies on the precise mechanisms of HP- and other bacteria-directed lymphomagenesis in antibiotic-responsive gastric MALT lymphomas are warranted.
Collapse
Affiliation(s)
- Sung-Hsin Kuo
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
- Cancer Research Center, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei 106, Taiwan.
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Kun-Huei Yeh
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
- Cancer Research Center, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei 106, Taiwan.
| | - Chung-Wu Lin
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Ping-Ning Hsu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, National Cheng-Kung University Hospital, Tainan 704, Taiwan.
| | - Ann-Lii Cheng
- Cancer Research Center, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei 106, Taiwan.
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| |
Collapse
|
21
|
Senda T, Dogra P, Granot T, Furuhashi K, Snyder ME, Carpenter DJ, Szabo PA, Thapa P, Miron M, Farber DL. Microanatomical dissection of human intestinal T-cell immunity reveals site-specific changes in gut-associated lymphoid tissues over life. Mucosal Immunol 2019; 12:378-389. [PMID: 30523311 PMCID: PMC6375790 DOI: 10.1038/s41385-018-0110-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/30/2018] [Indexed: 02/04/2023]
Abstract
Defining adaptive immunity with the complex structures of the human gastrointestinal (GI) tract over life is essential for understanding immune responses to ingested antigens, commensal and pathogenic microorganisms, and dysfunctions in disease. We present here an analysis of lymphocyte localization and T cell subset composition across the human GI tract including mucosal sites (jejunum, ileum, colon), gut-associated lymphoid tissues (isolated lymphoid follicles (ILFs), Peyer's patches (PPs), appendix), and mesenteric lymph nodes (MLNs) from a total of 68 donors spanning eight decades of life. In pediatric donors, ILFs and PP containing naïve T cells and regulatory T cells (Tregs) are prevalent in the jejunum and ileum, respectively; these decline in frequency with age, contrasting stable frequencies of ILFs and T cell subsets in the colon. In the mucosa, tissue resident memory T cells develop during childhood, and persist in high frequencies into advanced ages, while T cell composition changes with age in GALT and MLN. These spatial and temporal features of human intestinal T cell immunity define signatures that can be used to train predictive machine learning algorithms. Our findings demonstrate an anatomic basis for age-associated alterations in immune responses, and establish a quantitative baseline for intestinal immunity to define disease pathologies.
Collapse
Affiliation(s)
- Takashi Senda
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Surgery, Columbia University Medical Center, New York, NY, 10032, USA
| | - Pranay Dogra
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Tomer Granot
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
- Compugen, San Francisco, CA, USA
| | - Kazuhiro Furuhashi
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Mark E Snyder
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Dustin J Carpenter
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Surgery, Columbia University Medical Center, New York, NY, 10032, USA
| | - Peter A Szabo
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Puspa Thapa
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Michelle Miron
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA.
- Department of Surgery, Columbia University Medical Center, New York, NY, 10032, USA.
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
22
|
Casanova-Acebes M, Nicolás-Ávila JA, Li JL, García-Silva S, Balachander A, Rubio-Ponce A, Weiss LA, Adrover JM, Burrows K, A-González N, Ballesteros I, Devi S, Quintana JA, Crainiciuc G, Leiva M, Gunzer M, Weber C, Nagasawa T, Soehnlein O, Merad M, Mortha A, Ng LG, Peinado H, Hidalgo A. Neutrophils instruct homeostatic and pathological states in naive tissues. J Exp Med 2018; 215:2778-2795. [PMID: 30282719 PMCID: PMC6219739 DOI: 10.1084/jem.20181468] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/26/2018] [Accepted: 09/13/2018] [Indexed: 12/31/2022] Open
Abstract
Immune protection relies on the capacity of neutrophils to infiltrate challenged tissues. Naive tissues, in contrast, are believed to remain free of these cells and protected from their toxic cargo. Here, we show that neutrophils are endowed with the capacity to infiltrate multiple tissues in the steady-state, a process that follows tissue-specific dynamics. By focusing in two particular tissues, the intestine and the lungs, we find that neutrophils infiltrating the intestine are engulfed by resident macrophages, resulting in repression of Il23 transcription, reduced G-CSF in plasma, and reinforced activity of distant bone marrow niches. In contrast, diurnal accumulation of neutrophils within the pulmonary vasculature influenced circadian transcription in the lungs. Neutrophil-influenced transcripts in this organ were associated with carcinogenesis and migration. Consistently, we found that neutrophils dictated the diurnal patterns of lung invasion by melanoma cells. Homeostatic infiltration of tissues unveils a facet of neutrophil biology that supports organ function, but can also instigate pathological states.
Collapse
Affiliation(s)
- Maria Casanova-Acebes
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - José A Nicolás-Ávila
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Jackson LiangYao Li
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Singapore Immunology Nework (SIgN), A*STAR, Biopolis, Singapore
| | - Susana García-Silva
- Department of Molecular Oncology, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | | | - Andrea Rubio-Ponce
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Linnea A Weiss
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - José M Adrover
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Kyle Burrows
- Department of Immunology, University of Toronto, Canada
| | - Noelia A-González
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ivan Ballesteros
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Sapna Devi
- Singapore Immunology Nework (SIgN), A*STAR, Biopolis, Singapore
| | - Juan A Quintana
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Georgiana Crainiciuc
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Magdalena Leiva
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Matthias Gunzer
- University Duisburg-Essen, University Hospital, Institute for Experimental Immunology and Imaging, Essen, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University, Munich, Germany.,Dept. of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University, Munich, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Miriam Merad
- Tisch Cancer Institute and Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Canada
| | - Lai Guan Ng
- Singapore Immunology Nework (SIgN), A*STAR, Biopolis, Singapore
| | - Hector Peinado
- Department of Molecular Oncology, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Andrés Hidalgo
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain .,Institute for Cardiovascular Prevention, Ludwig-Maximilians University, Munich, Germany
| |
Collapse
|
23
|
Porter RJ, Andrews C, Brice DP, Durum SK, McLean MH. Can We Target Endogenous Anti-inflammatory Responses as a Therapeutic Strategy for Inflammatory Bowel Disease? Inflamm Bowel Dis 2018; 24:2123-2134. [PMID: 30020451 PMCID: PMC6140439 DOI: 10.1093/ibd/izy230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel disease (IBD) describes chronic relapsing remitting inflammation of the gastrointestinal tract including ulcerative colitis and Crohn's disease. The prevalence of IBD is rising across the globe. Despite a growing therapeutic arsenal, current medical treatments are not universally effective, do not induce lasting remission in all, or are accompanied by short- and long-term adverse effects. Therefore, there is a clinical need for novel therapeutic strategies for IBD. Current treatments for IBD mainly manipulate the immune system for therapeutic gain by inhibiting pro-inflammatory activity. There is a robust endogenous immunoregulatory capacity within the repertoire of both innate and adaptive immune responses. An alternative treatment strategy for IBD is to hijack and bolster this endogenous capability for therapeutic gain. This review explores this hypothesis and presents current evidence for this therapeutic direction in immune cell function, cytokine biology, and alternative mechanisms of immunoregulation such as microRNA, oligonucleotides, and the endocannabinoid system.
Collapse
Affiliation(s)
- Ross John Porter
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, United Kingdom
| | - Caroline Andrews
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | - Daniel Paul Brice
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, United Kingdom
| | - Scott Kenneth Durum
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| | - Mairi Hall McLean
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, United Kingdom,Address correspondence to: Mairi H. McLean, Institute of Medical Sciences, Foresterhill, Aberdeen, Scotland, UK, AB25 2ZD. E-mail:
| |
Collapse
|
24
|
Ueno A, Jeffery L, Kobayashi T, Hibi T, Ghosh S, Jijon H. Th17 plasticity and its relevance to inflammatory bowel disease. J Autoimmun 2018; 87:38-49. [DOI: 10.1016/j.jaut.2017.12.004] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 12/03/2017] [Indexed: 02/08/2023]
|
25
|
Zeng Q, He X, Puthiyakunnon S, Xiao H, Gong Z, Boddu S, Chen L, Tian H, Huang SH, Cao H. Probiotic Mixture Golden Bifido Prevents Neonatal Escherichia coli K1 Translocation via Enhancing Intestinal Defense. Front Microbiol 2017; 8:1798. [PMID: 28979247 PMCID: PMC5611410 DOI: 10.3389/fmicb.2017.01798] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/05/2017] [Indexed: 12/15/2022] Open
Abstract
Escherichia coli (E. coli) K1 sepsis and meningitis is a severe infection characterized by high mortality in neonates. Successful colonization and translocation across the intestinal mucosa have been regarded as the critical steps for E. coli K1 sepsis and meningitis. We recently reported that the probiotic mixture, Golden Bifido (containing live Lactobacillus bulgaricus, Bifidobacterium, and Streptococcus thermophilus, LBS) has a preventive role against neonatal E. coli K1 bacteremia and meningitis. However, the interaction between the neonatal gut barrier, probiotics and E. coli K1 is still not elucidated. The present study aims to investigate how LBS exerts its protective effects on neonatal gut barrier during E. coli K1 infection. The beneficial effects of LBS were explored in vitro and in vivo using human colon carcinoma cell lines HT-29 and rat model of neonatal E. coli K1 infection, respectively. Our results showed that stimulation with E. coli K1 was able to cause intestinal barrier dysfunction, which were reflected by E. coli K1-induced intestinal damage and apoptosis of intestinal epithelial cells, reduction of mucin, immunoglobulin A (IgA) and tight junction proteins expression, as well as increase in intestinal permeability, all these changes facilitate E. coli K1 intestinal translocation. However, these changes were alleviated when HT-29 cells were treated with LBS before E. coli K1 infection. Furthermore, we found that LBS-treated neonatal rats (without E. coli K1 infection) have showed higher production of mucin, ZO-1, IgA, Ki67 in intestinal mucosa as well as lower intestinal permeability than that of non-treated rats, indicating that LBS could accelerate the development of neonatal intestinal defense. Taken together, our results suggest that enhancement of the neonatal intestinal defense to fight against E. coli K1 translocation could be the potential mechanism to elucidate how LBS confers a protective effect against neonatal E. coli K1 bacteremia and meningitis. This indirect mechanism makes LBS exert preventive effect on most of gut-derived pathogenic infections rather than only E. coli.
Collapse
Affiliation(s)
- Qing Zeng
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Xiaolong He
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Santhosh Puthiyakunnon
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Hansen Xiao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Zelong Gong
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Swapna Boddu
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Lecheng Chen
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| | - Huiwen Tian
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China.,The First School of Clinical Medicine, Southern Medical UniversityGuangzhou, China
| | - Sheng-He Huang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China.,Saban Research Institute, Children's Hospital Los Angeles, University of Southern California, Los AngelesCA, United States
| | - Hong Cao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical UniversityGuangzhou, China
| |
Collapse
|
26
|
Li S, Heller JJ, Bostick JW, Lee A, Schjerven H, Kastner P, Chan S, Chen ZE, Zhou L. Ikaros Inhibits Group 3 Innate Lymphoid Cell Development and Function by Suppressing the Aryl Hydrocarbon Receptor Pathway. Immunity 2017; 45:185-97. [PMID: 27438771 DOI: 10.1016/j.immuni.2016.06.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 04/27/2016] [Accepted: 05/10/2016] [Indexed: 02/08/2023]
Abstract
Group 3 innate lymphoid cells (ILC3s) expressing the transcription factor (TF) RORγt are important for the defense and homeostasis of host intestinal tissues. The zinc finger TF Ikaros, encoded by Ikzf1, is essential for the development of RORγt(+) fetal lymphoid tissue inducer (LTi) cells and lymphoid organogenesis, but its role in postnatal ILC3s is unknown. Here, we show that small-intestinal ILC3s had lower Ikaros expression than ILC precursors and other ILC subsets. Ikaros inhibited ILC3s in a cell-intrinsic manner through zinc-finger-dependent inhibition of transcriptional activity of the aryl hydrocarbon receptor, a key regulator of ILC3 maintenance and function. Ablation of Ikzf1 in RORγt(+) ILC3s resulted in increased expansion and cytokine production of intestinal ILC3s and protection against infection and colitis. Therefore, in contrast to being required for LTi development, Ikaros inhibits postnatal ILC3 development and function to regulate gut immune responses at steady state and in disease.
Collapse
Affiliation(s)
- Shiyang Li
- 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 Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Jennifer J Heller
- 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
| | - 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
| | - Aileen Lee
- 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
| | - Hilde Schjerven
- Department of Laboratory Medicine, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Philippe Kastner
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Susan Chan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U964, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Zongming E Chen
- Department of Laboratory Medicine in Geisinger Health System, 100 N. Academy Avenue, MC 19-20, Danville, PA 17822, 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; Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA.
| |
Collapse
|
27
|
Moingeon P, Mascarell L. Differences and similarities between sublingual immunotherapy of allergy and oral tolerance. Semin Immunol 2017; 30:52-60. [PMID: 28760498 DOI: 10.1016/j.smim.2017.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/13/2017] [Indexed: 12/27/2022]
Abstract
Allergen immunotherapy is the only treatment altering the natural course of IgE-mediated allergies. Whereas the subcutaneous route for immunotherapy (SCIT) has been historically considered as a reference, we discuss herein the relative advantages of the sublingual and oral routes as alternatives to SCIT in order to elicit allergen-specific tolerance. The buccal and gut immune systems are similarly organized to favor immune tolerance to antigens/allergens, due to the presence of tolerogenic dendritic cells and macrophages promoting the differentiation of CD4+ regulatory T cells. Sublingual immunotherapy (SLIT) is now established as a valid treatment option, with clinical efficacy demonstrated in allergic rhinoconjunctivitis (to either grass, tree, weed pollens or mite allergens) and encouraging results obtained in the management of mild/moderate allergic asthma. While still exploratory, oral immunotherapy (OIT) has shown promising results in the desensitization of patients with food allergies. We review at both biological and clinical levels the perspectives currently pursued for those two mucosal routes.
Collapse
Affiliation(s)
- Philippe Moingeon
- Research Department, Stallergenes Greer, 6 rue Alexis de Tocqueville, 92160 Antony, France.
| | - Laurent Mascarell
- Research Department, Stallergenes Greer, 6 rue Alexis de Tocqueville, 92160 Antony, France
| |
Collapse
|
28
|
Jinnohara T, Kanaya T, Hase K, Sakakibara S, Kato T, Tachibana N, Sasaki T, Hashimoto Y, Sato T, Watarai H, Kunisawa J, Shibata N, Williams IR, Kiyono H, Ohno H. IL-22BP dictates characteristics of Peyer's patch follicle-associated epithelium for antigen uptake. J Exp Med 2017; 214:1607-1618. [PMID: 28512157 PMCID: PMC5460992 DOI: 10.1084/jem.20160770] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 02/15/2017] [Accepted: 04/04/2017] [Indexed: 12/19/2022] Open
Abstract
Interleukin-22 (IL-22) acts protectively and harmfully on intestinal tissue depending on the situation; therefore, IL-22 signaling needs to be tightly regulated. IL-22 binding protein (IL-22BP) binds IL-22 to inhibit IL-22 signaling. It is expressed in intestinal and lymphoid tissues, although its precise distribution and roles have remained unclear. In this study, we show that IL-22BP is highly expressed by CD11b+CD8α- dendritic cells in the subepithelial dome region of Peyer's patches (PPs). We found that IL-22BP blocks IL-22 signaling in the follicle-associated epithelium (FAE) covering PPs, indicating that IL-22BP plays a role in regulating the characteristics of the FAE. As expected, FAE of IL-22BP-deficient (Il22ra2-/-) mice exhibited altered properties such as the enhanced expression of mucus and antimicrobial proteins as well as prominent fucosylation, which are normally suppressed in FAE. Additionally, Il22ra2-/- mice exhibited the decreased uptake of bacterial antigens into PPs without affecting M cell function. Our present study thus demonstrates that IL-22BP promotes bacterial uptake into PPs by influencing FAE gene expression and function.
Collapse
Affiliation(s)
- Toshi Jinnohara
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, Institute of Physical and Chemical Research, Yokohama 230-0045, Japan
- Department of Medical Life Science, Division of Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Takashi Kanaya
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, Institute of Physical and Chemical Research, Yokohama 230-0045, Japan
- Department of Medical Life Science, Division of Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
- Division of Mucosal Barriology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Sayuri Sakakibara
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, Institute of Physical and Chemical Research, Yokohama 230-0045, Japan
| | - Tamotsu Kato
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, Institute of Physical and Chemical Research, Yokohama 230-0045, Japan
| | - Naoko Tachibana
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, Institute of Physical and Chemical Research, Yokohama 230-0045, Japan
| | - Takaharu Sasaki
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, Institute of Physical and Chemical Research, Yokohama 230-0045, Japan
| | - Yusuke Hashimoto
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, Institute of Physical and Chemical Research, Yokohama 230-0045, Japan
- Department of Medical Life Science, Division of Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Toshiro Sato
- Department of Gastroenterology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hiroshi Watarai
- Division of Stem Cell Cellomics, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Jun Kunisawa
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Naoko Shibata
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Ifor R Williams
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Tokyo 102-0076, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, Institute of Physical and Chemical Research, Yokohama 230-0045, Japan
- Department of Medical Life Science, Division of Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| |
Collapse
|
29
|
Mora-Velandia LM, Castro-Escamilla O, Méndez AG, Aguilar-Flores C, Velázquez-Avila M, Tussié-Luna MI, Téllez-Sosa J, Maldonado-García C, Jurado-Santacruz F, Ferat-Osorio E, Martínez-Barnetche J, Pelayo R, Bonifaz LC. A Human Lin - CD123 + CD127 low Population Endowed with ILC Features and Migratory Capabilities Contributes to Immunopathological Hallmarks of Psoriasis. Front Immunol 2017; 8:176. [PMID: 28303135 PMCID: PMC5332395 DOI: 10.3389/fimmu.2017.00176] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/07/2017] [Indexed: 02/06/2023] Open
Abstract
Innate lymphoid cells (ILC) are members of a heterogeneous family with a lymphoid origin that mimics the T helper (Th) cytokine profile. ILC are involved in early effector cytokine-mediated responses during infections in peripheral tissues. ILC also play an important role in chronic skin inflammatory diseases, including psoriasis. Although classical ILC express CD127, it has been recently reported that the presence of non-classical CD127- ILC populations and an early ILC precursor (EILP) CD127low. ILC development has predominately been investigated in mouse models. However, in humans, different transcription factors have been described for ILC identification. NFIL3 (nuclear factor, IL-3 regulated) is crucial for ILC development in response to IL-7. CD123 (IL-3Rα) is usually used to exclude basophils during ILC identification, however, it is unknown if in response to IL-3, NFIL3 could be relevant to induce ILC features in Lin- CD123+ populations in addition, is also unknown whether peripheral blood (PB) population with ILC features may have skin-homing potential to participate in skin inflammatory chronic diseases. Here, we report a Lin- CD123+ CD127low CD7+ CLA+ population that share some phenotypic properties with basophils, but expresses several transcription factors for ILC commitment such as inhibitor of DNA binding 2 (Id2), NFIL3, promyelocytic leukemia zinc finger (PLZF), thymocyte selection-associated high-mobility group box protein (TOX), and T cell factor-1 (TCF-1). In addition, this population expresses different ILC markers: CD132, CD90, CD161, α4 integrin, c-Kit, CRTH2, AhR, and IL-23R. IL-3 prevents apoptosis and increases their NFIL3, TOX, and PLZF expression. In PB, the CD123+ CD127low population is predominantly a conspicuous population that expresses T-bet and RORγt. The Lin- CD123+ CD127low population in PB has a limited Th type cytokine expression and highly expresses IL-8. The Lin- CD123+ CD127low population expresses skin-homing receptors (cutaneous lymphocyte antigen and CXCR4) and transmigrates through endothelial cells in response to SDF-1. An equivalent Lin- CD123low population was identified in control skin, which shows a broader phenotypic diversity and cytokine production, including IL-22 and IL-17. Remarkably, the CD123low population in the lesion and non-lesion skin of psoriasis patients expresses IL-17 and IL-22. Our findings suggest the identification of an alternative Lin- CD123+ CD127low population with ILC features endowed with migratory capabilities that might contribute to immunopathological hallmarks of psoriasis.
Collapse
Affiliation(s)
- Luz María Mora-Velandia
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico; Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Octavio Castro-Escamilla
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico; Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Andrés González Méndez
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social , Mexico City , Mexico
| | - Cristina Aguilar-Flores
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social , Mexico City , Mexico
| | - Martha Velázquez-Avila
- Unidad de Investigación en Enfermedades Oncológicas, Hospital de Oncología, Centro Médico Nacional 'Siglo XXI' , Mexico City , Mexico
| | - María Isabel Tussié-Luna
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico; Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México "Federico Gómez", Mexico City, Mexico
| | - Juan Téllez-Sosa
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública , Cuernavaca, Morelos , Mexico
| | - César Maldonado-García
- Centro Dermatológico "Dr. Ladislao de la Pascua", Secretaria de Salud de la Ciudad de México , Mexico City , Mexico
| | - Fermín Jurado-Santacruz
- Centro Dermatológico "Dr. Ladislao de la Pascua", Secretaria de Salud de la Ciudad de México , Mexico City , Mexico
| | - Eduardo Ferat-Osorio
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social , Mexico City , Mexico
| | - Jesus Martínez-Barnetche
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública , Cuernavaca, Morelos , Mexico
| | - Rosana Pelayo
- Unidad de Investigación en Enfermedades Oncológicas, Hospital de Oncología, Centro Médico Nacional 'Siglo XXI' , Mexico City , Mexico
| | - Laura C Bonifaz
- Unidad de Investigación Médica en Inmunoquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social , Mexico City , Mexico
| |
Collapse
|
30
|
Kuper CF, Wijnands MVW, Zander SAL. Mucosa-Associated Lymphoid Tissues. IMMUNOPATHOLOGY IN TOXICOLOGY AND DRUG DEVELOPMENT 2017. [DOI: 10.1007/978-3-319-47385-7_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
31
|
Tafalla C, Leal E, Yamaguchi T, Fischer U. T cell immunity in the teleost digestive tract. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 64:167-177. [PMID: 26905634 DOI: 10.1016/j.dci.2016.02.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 02/10/2016] [Accepted: 02/16/2016] [Indexed: 06/05/2023]
Abstract
Fish (along with cyclostomes) constitute the most ancient animal group in which an acquired immune system is present. As in higher vertebrates, both B and T lymphocytes cooperate in implementing an adequate response. Although there is still a debate on whether fish possess a true gut associated lymphoid tissue (GALT), the presence of diffuse B and T lymphocytes throughout all mucosal surfaces has been demonstrated in a wide variety of fish species. The lack of antibodies against T lymphocyte markers has hampered the performance of functional assays in both systemic and mucosal compartments. However, most components associated with T lymphocyte function have been identified in fish through extensive genomic research, suggesting similar functionalities for fish and mammalian T lymphocytes. Thus, the aim of this review is to briefly summarize what is known in teleost concerning the characteristics and functionalities of the different T cell subsets, to then focus on what is known to date regarding their presence and role in the gastrointestinal tract, through either direct functional assays or indirectly by conclusions drawn from transcriptomic analysis.
Collapse
Affiliation(s)
- Carolina Tafalla
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, Madrid, Spain.
| | - Esther Leal
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, Madrid, Spain
| | - Takuya Yamaguchi
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Uwe Fischer
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| |
Collapse
|
32
|
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: 87] [Impact Index Per Article: 9.7] [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
|
33
|
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: 3.1] [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
|
34
|
Zhang R, Zhu W, Mao S. High-concentrate feeding upregulates the expression of inflammation-related genes in the ruminal epithelium of dairy cattle. J Anim Sci Biotechnol 2016; 7:42. [PMID: 27478614 PMCID: PMC4966727 DOI: 10.1186/s40104-016-0100-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 07/12/2016] [Indexed: 12/14/2022] Open
Abstract
Background The objective of this study was to characterize the mRNA expression profile related to rumen epithelial inflammation through the in vivo and in vitro experiments. In the in vivo experiment, rumen papillae were collected from four dairy cows adapted to either a 40 % (LC) or 70 % (HC) concentrate feeds for microarray analysis. Results Results showed that 245 differentially expressed genes (DEGs) were detected in the cows fed the HC relative to the LC diet. The DEGs were first annotated, and results revealed that the expression of inflammation-related genes, including IL-1β, IL-2, IL-22, CCL19, CCL8, CX3CR1, CXCL6, INHBE, LEPR, PRL, and TNFRSF9 found in the cytokine-cytokine receptor pathway were up-regulated in the HC-fed cows, indicating local inflammation in the rumen epithelium was triggered. The expression of IL-1β, IL-2, and IL-6 was further validated by qRT-PCR. To demonstrate whether there were relationships between cytokine mRNA expression and ruminal factors (pH and LPS), the isolated ruminal epithelial cells were cultured in vitro. Results showed that the mRNA expression of IL-1β, IL-2, IL-6, and IL-8 increased after the LPS treatment, while low-pH treatment elevated the mRNA expression of TNF-α, suggesting that low-pH coupled with higher levels of LPS in rumen of cows fed the HC may be mainly responsible for the triggered local ruminal inflammation. Conclusions Our results indicate that ruminal local inflammation response might be triggered during HC feeding, and these findings also enhance the knowledge of rumen epithelial adaptation to HC at the molecular level. Electronic supplementary material The online version of this article (doi:10.1186/s40104-016-0100-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ruiyang Zhang
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 Peoples Republic of China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 Peoples Republic of China
| | - Shengyong Mao
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 Peoples Republic of China
| |
Collapse
|
35
|
Hwang JY, Randall TD, Silva-Sanchez A. Inducible Bronchus-Associated Lymphoid Tissue: Taming Inflammation in the Lung. Front Immunol 2016; 7:258. [PMID: 27446088 PMCID: PMC4928648 DOI: 10.3389/fimmu.2016.00258] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/17/2016] [Indexed: 01/09/2023] Open
Abstract
Following pulmonary inflammation, leukocytes that infiltrate the lung often assemble into structures known as inducible Bronchus-Associated Lymphoid Tissue (iBALT). Like conventional lymphoid organs, areas of iBALT have segregated B and T cell areas, specialized stromal cells, high endothelial venules, and lymphatic vessels. After inflammation is resolved, iBALT is maintained for months, independently of inflammation. Once iBALT is formed, it participates in immune responses to pulmonary antigens, including those that are unrelated to the iBALT-initiating antigen, and often alters the clinical course of disease. However, the mechanisms that govern immune responses in iBALT and determine how iBALT impacts local and systemic immunity are poorly understood. Here, we review our current understanding of iBALT formation and discuss how iBALT participates in pulmonary immunity.
Collapse
Affiliation(s)
- Ji Young Hwang
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham , Birmingham, AL , USA
| | - Troy D Randall
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham , Birmingham, AL , USA
| | - Aaron Silva-Sanchez
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham , Birmingham, AL , USA
| |
Collapse
|
36
|
Kathania M, Khare P, Zeng M, Cantarel B, Zhang H, Ueno H, Venuprasad K. Itch inhibits IL-17-mediated colon inflammation and tumorigenesis by ROR-γt ubiquitination. Nat Immunol 2016; 17:997-1004. [PMID: 27322655 DOI: 10.1038/ni.3488] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/05/2016] [Indexed: 02/06/2023]
Abstract
Dysregulated expression of interleukin 17 (IL-17) in the colonic mucosa is associated with colonic inflammation and cancer. However, the cell-intrinsic molecular mechanisms by which IL-17 expression is regulated remain unclear. We found that deficiency in the ubiquitin ligase Itch led to spontaneous colitis and increased susceptibility to colon cancer. Itch deficiency in the TH17 subset of helper T cells, innate lymphoid cells and γδ T cells resulted in the production of elevated amounts of IL-17 in the colonic mucosa. Mechanistically, Itch bound to the transcription factor ROR-γt and targeted ROR-γt for ubiquitination. Inhibition or genetic inactivation of ROR-γt attenuated IL-17 expression and reduced spontaneous colonic inflammation in Itch(-/-) mice. Thus, we have identified a previously unknown role for Itch in regulating IL-17-mediated colonic inflammation and carcinogenesis.
Collapse
Affiliation(s)
- Mahesh Kathania
- Baylor Institute for Immunology Research, Baylor Research Institute, Dallas, Texas, USA
| | - Prashant Khare
- Baylor Institute for Immunology Research, Baylor Research Institute, Dallas, Texas, USA
| | - Minghui Zeng
- Baylor Institute for Immunology Research, Baylor Research Institute, Dallas, Texas, USA
| | - Brandi Cantarel
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Haiying Zhang
- Department of Pathology, Baylor University Medical Center, Dallas, Texas, USA
| | - Hideki Ueno
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - K Venuprasad
- Baylor Institute for Immunology Research, Baylor Research Institute, Dallas, Texas, USA
| |
Collapse
|
37
|
Roulis M, Bongers G, Armaka M, Salviano T, He Z, Singh A, Seidler U, Becker C, Demengeot J, Furtado GC, Lira SA, Kollias G. Host and microbiota interactions are critical for development of murine Crohn's-like ileitis. Mucosal Immunol 2016; 9:787-97. [PMID: 26487367 PMCID: PMC5027991 DOI: 10.1038/mi.2015.102] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 08/31/2015] [Indexed: 02/04/2023]
Abstract
Deregulation of host-microbiota interactions in the gut is a pivotal characteristic of Crohn's disease. It remains unclear, however, whether commensals and/or the dysbiotic microbiota associated with pathology in humans are causally involved in Crohn's pathogenesis. Here, we show that Crohn's-like ileitis in Tnf(ΔARE/+) mice is microbiota-dependent. Germ-free Tnf(ΔARE/+) mice are disease-free and the microbiota and its innate recognition through Myd88 are indispensable for tumor necrosis factor (TNF) overexpression and disease initiation in this model. The epithelium of diseased mice shows no major defects in mucus barrier and paracellular permeability. However, Tnf(ΔARE/+) ileitis associates with the reduction of lysozyme-expressing Paneth cells, mediated by adaptive immune effectors. Furthermore, we show that established but not early ileitis in Tnf(ΔARE/+) mice involves defective expression of antimicrobials and dysbiosis, characterized by Firmicutes expansion, including epithelial-attaching segmented filamentous bacteria, and decreased abundance of Bacteroidetes. Microbiota modulation by antibiotic treatment at an early disease stage rescues ileitis. Our results suggest that the indigenous microbiota is sufficient to drive TNF overexpression and Crohn's ileitis in the genetically susceptible Tnf(ΔARE/+) hosts, whereas dysbiosis in this model results from disease-associated alterations including loss of lysozyme-expressing Paneth cells.
Collapse
Affiliation(s)
- M Roulis
- Institute of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Vari 16672, Greece
| | - G Bongers
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - M Armaka
- Institute of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Vari 16672, Greece
| | - T Salviano
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Z He
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - A Singh
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - U Seidler
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - C Becker
- Department of Medicine 1, Universitätsklinikum der Friedrich-Alexander-Universität, Erlangen, Germany
| | - J Demengeot
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - GC Furtado
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - SA Lira
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA,Address correspondence to: Sergio Lira, Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, Box 1630, New York, NY 10029-6574. Phone: 1-212-659-9404; Fax: 1-212-849-2525; and George Kollias, Biomedical Sciences Research Center “Alexander Fleming”, 34, Al. Fleming Street, 16672 Vari, Greece. Phone: +302109656507; Fax: +302109656563; and Department of Physiology, Medical School, National & Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27, Greece. Phone: +302107462507; Fax: +30210-7462571;
| | - G Kollias
- Institute of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Vari 16672, Greece,Second address: Department of Physiology, Medical School, National & Kapodistrian University of Athens, Athens 11527, Greece,Address correspondence to: Sergio Lira, Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, Box 1630, New York, NY 10029-6574. Phone: 1-212-659-9404; Fax: 1-212-849-2525; and George Kollias, Biomedical Sciences Research Center “Alexander Fleming”, 34, Al. Fleming Street, 16672 Vari, Greece. Phone: +302109656507; Fax: +302109656563; and Department of Physiology, Medical School, National & Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27, Greece. Phone: +302107462507; Fax: +30210-7462571;
| |
Collapse
|
38
|
Olsen L, Åkesson CP, Aleksandersen M, Boysen P, Press CM, Drouet F, Storset AK, Espenes A. NCR1+ cells appear early in GALT development of the ovine foetus and acquire a c-kit+ phenotype towards the end of gestation. Vet Immunol Immunopathol 2016; 169:79-84. [PMID: 26827843 DOI: 10.1016/j.vetimm.2015.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 11/17/2022]
Abstract
The amount, distribution and phenotype of ovine NCR1+ cells were investigated during developing GALT from day 70 of gestation. Antibodies against CD3 and CD79 were used to identify the compartments of GALT, and the localization of NCR1+ cells were correlated within these structures. Markers CD34 and c-kit, in addition to Ki67, were used to investigate possible origin and the stage of development of the NCR1+ cells. NCR1+ cells were present as single cells in the subepithelial tissue as early as 70 days of gestation, and were predominantly present in the T cell rich IFAs and domes as these intestinal wall compartments developed. While NCR1+ cells proliferated more intensively at mid-gestation (70-104 days), the number of NCR1+ cells also expressing c-kit, increased at the end of gestation. In conclusion, NCR1+ cells appeared early in T cell areas of the gut and displayed a phenotype consistent with intermediate stages of cNK cells and/or a subpopulation of ILC22.
Collapse
Affiliation(s)
- Line Olsen
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| | - Caroline Piercey Åkesson
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| | - Mona Aleksandersen
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| | - Preben Boysen
- Department of Food Safety & Infection Biology, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway.
| | - Charles McL Press
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| | - Françoise Drouet
- Institut National de la Recherche Agronomique, UMR1282, Infectiologie et Santé Publique, Laboratoire Apicomplexes et Immunité Muqueuse, Nouzilly, France.
| | - Anne K Storset
- Department of Food Safety & Infection Biology, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway.
| | - Arild Espenes
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| |
Collapse
|
39
|
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: 0.3] [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
|
40
|
Tian Z, van Velkinburgh JC, Wu Y, Ni B. Innate lymphoid cells involve in tumorigenesis. Int J Cancer 2016; 138:22-9. [PMID: 25604320 DOI: 10.1002/ijc.29443] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/14/2015] [Indexed: 12/17/2022]
Abstract
Innate lymphoid cells (ILCs) promptly initiate cytokine responses to pathogen exposure in the mucosa and mucosal-associated lymphoid tissues. ILCs were recently categorized as being of the lymphoid lineage and have been classified into three groups. ILCs play important roles in immunity against pathogens, and an anti-tumor immune-related function was recently demonstrated. In this review we discuss whether and how ILCs involve in the tumorigenesis, providing new insights into the mechanisms underlying the particular functions of ILCs as well as the potential targets for tumor intervention.
Collapse
Affiliation(s)
- Zhiqiang Tian
- Institute of Immunology PLA, Third Military Medical University, Chongqing, China
| | | | - Yuzhang Wu
- Institute of Immunology PLA, Third Military Medical University, Chongqing, China
| | - Bing Ni
- Institute of Immunology PLA, Third Military Medical University, Chongqing, China
| |
Collapse
|
41
|
Xu H, Wang X, Lackner AA, Veazey RS. Type 3 innate lymphoid cell depletion is mediated by TLRs in lymphoid tissues of simian immunodeficiency virus-infected macaques. FASEB J 2015; 29:5072-80. [PMID: 26283536 PMCID: PMC4653054 DOI: 10.1096/fj.15-276477] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/13/2015] [Indexed: 12/22/2022]
Abstract
Innate lymphoid cells (ILCs) type 3, also known as lymphoid tissue inducer cells, plays a major role in both the development and remodeling of organized lymphoid tissues and the maintenance of adaptive immune responses. HIV/simian immunodeficiency virus (SIV) infection causes breakdown of intestinal barriers resulting in microbial translocation, leading to systemic immune activation and disease progression. However, the effects of HIV/SIV infection on ILC3 are unknown. Here, we analyzed ILC3 from mucosal and systemic lymphoid tissues in chronically SIV-infected macaques and uninfected controls. ILC3 cells were defined and identified in macaque lymphoid tissues as non-T, non-B (lineage-negative), c-Kit(+)IL-7Rα(+) (CD117(+)CD127(+)) cells. These ILC3 cells highly expressed CD90 (∼ 63%) and aryl hydrocarbon receptor and produced IL-17 (∼ 63%), IL-22 (∼ 36%), and TNF-α (∼ 72%) but did not coexpress CD4 or NK cell markers. The intestinal ILC3 cell loss correlated with the reduction of total CD4(+) T cells and T helper (Th)17 and Th22 cells in the gut during SIV infection (P < 0.001). Notably, ILC3 could be induced to undergo apoptosis by microbial products through the TLR2 (lipoteichoic acid) and/or TLR4 (LPS) pathway. These findings indicated that persistent microbial translocation may result in loss of ILC3 in lymphoid tissues in SIV-infected macaques, further contributing to the HIV-induced impairment of gut-associated lymphoid tissue structure and function, especially in mucosal tissues.
Collapse
Affiliation(s)
- Huanbin Xu
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA
| | - Xiaolei Wang
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA
| | - Andrew A Lackner
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA
| | - Ronald S Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, USA
| |
Collapse
|
42
|
Kisseleva EP. Innate immunity underlies symbiotic relationships. BIOCHEMISTRY (MOSCOW) 2015; 79:1273-85. [PMID: 25716721 DOI: 10.1134/s0006297914120013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Here, the modern data regarding interactions between normal microbiota and barrier tissues in plants, humans and animals are reviewed. The main homeostatic mechanisms responsible for interactions between epithelium and innate immune cells with symbiotic bacteria are described. A key step in this process is recognition of soluble microbial products by ligation to pattern-recognition receptors expressed on the host cells. As a result, epithelial cells secrete mucus, antibacterial peptides and immunoregulatory molecules. The main outcomes from immunological reactions towards symbiotic bacteria involve development of conditions for formation and maintenance of microbial biocenosis as well as providing safety for the host. Also, it is considered important to preserve and transfer beneficial bacteria to progeny.
Collapse
Affiliation(s)
- E P Kisseleva
- Institute of Experimental Medicine, Russian Academy of Medical Sciences, St. Petersburg, 197376, Russia.
| |
Collapse
|
43
|
CCL2 Promotes Colorectal Carcinogenesis by Enhancing Polymorphonuclear Myeloid-Derived Suppressor Cell Population and Function. Cell Rep 2015; 12:244-57. [PMID: 26146082 PMCID: PMC4620029 DOI: 10.1016/j.celrep.2015.06.024] [Citation(s) in RCA: 293] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/16/2015] [Accepted: 06/06/2015] [Indexed: 12/12/2022] Open
Abstract
Our study reveals a non-canonical role for CCL2 in modulating non-macrophage, myeloid-derived suppressor cells (MDSCs) and shaping a tumor-permissive microenvironment during colon cancer development. We found that intratumoral CCL2 levels increased in patients with colitis-associated colorectal cancer (CRC), adenocarcinomas, and adenomas. Deletion of CCL2 blocked progression from dysplasia to adenocarcinoma and reduced the number of colonic MDSCs in a spontaneous mouse model of colitis-associated CRC. In a transplantable mouse model of adenocarcinoma and an APC-driven adenoma model, CCL2 fostered MDSC accumulation in evolving colonic tumors and enhanced polymorphonuclear (PMN)-MDSC immunosuppressive features. Mechanistically, CCL2 regulated T cell suppression of PMN-MDSCs in a STAT3-mediated manner. Furthermore, CCL2 neutralization decreased tumor numbers and MDSC accumulation and function. Collectively, our experiments support that perturbing CCL2 and targeting MDSCs may afford therapeutic opportunities for colon cancer interception and prevention.
Collapse
|
44
|
Miranda PJ, Delgobo M, Marino GF, Paludo KS, da Silva Baptista M, de Souza Pinto SE. The Oral Tolerance as a Complex Network Phenomenon. PLoS One 2015; 10:e0130762. [PMID: 26115356 PMCID: PMC4483238 DOI: 10.1371/journal.pone.0130762] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/23/2015] [Indexed: 11/18/2022] Open
Abstract
The phenomenon of oral tolerance refers to a local and systemic state of tolerance induced in the gut after its exposure to innocuous antigens. Recent findings have shown the interrelationship between cellular and molecular components of oral tolerance, but its representation through a network of interactions has not been investigated. Our work aims at identifying the causal relationship of each element in an oral tolerance network, and also to propose a phenomenological model that's capable of predicting the stochastic behavior of this network when under manipulation. We compared the changes of a "healthy" network caused by "knock-outs" (KOs) in two approaches: an analytical approach by the Perron Frobenius theory; and a computational approach, which we describe within this work in order to find numerical results for the model. Both approaches have shown the most relevant immunological components for this phenomena, that happens to corroborate the empirical results from animal models. Besides explain in a intelligible fashion how the components interacts in a complex manner, we also managed to describe and quantify the importance of KOs that hasn't been empirically tested.
Collapse
Affiliation(s)
| | - Murilo Delgobo
- Department of Biology, State University of Ponta Grossa, Ponta Grossa, Paraná, Brazil
| | - Giovani Favero Marino
- Department of Biology, State University of Ponta Grossa, Ponta Grossa, Paraná, Brazil
| | - Kátia Sabrina Paludo
- Department of Structural Biology, Molecular and Genetics, State University of Ponta Grossa, Ponta Grossa, Paraná, Brazil
| | - Murilo da Silva Baptista
- Institute for Complex Systems and Mathematical Biology, SUPA, University of Aberdeen, Aberdeen, United Kingdom
| | | |
Collapse
|
45
|
Chmiela M, Gajewski A, Rudnicka K. Helicobacter pylori vs coronary heart disease - searching for connections. World J Cardiol 2015; 7:187-203. [PMID: 25914788 PMCID: PMC4404374 DOI: 10.4330/wjc.v7.i4.187] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/16/2015] [Accepted: 02/09/2015] [Indexed: 02/07/2023] Open
Abstract
In this review, we discussed the findings and concepts underlying the potential role of Helicobacter pylori (H. pylori) infections in the initiation, development or persistence of atherosclerosis and coronary heart disease (CHD). This Gram-negative bacterium was described by Marshall and Warren in 1984. The majority of infected subjects carries and transmits H. pylori with no symptoms; however, in some individuals these bacteria may cause peptic ulcers, and even gastric cancers. The widespread prevalence of H. pylori infections and the fact that frequently they remain asymptomatic may suggest that, similarly to intestinal microflora, H. pylori may deliver antigens that stimulate not only local, but also systemic inflammatory response. Recently, possible association between H. pylori infection and extragastric disorders has been suggested. Knowledge on the etiology of atherosclerosis together with current findings in the area of H. pylori infections constitute the background for the newly proposed hypothesis that those two processes may be related. Many research studies confirm the indirect association between the prevalence of H. pylori and the occurrence of CHD. According to majority of findings the involvement of H. pylori in this process is based on the chronic inflammation which might facilitate the CHD-related pathologies. It needs to be elucidated, if the infection initiates or just accelerates the formation of atheromatous plaque.
Collapse
|
46
|
Schey R, Danzer C, Mattner J. Perturbations of mucosal homeostasis through interactions of intestinal microbes with myeloid cells. Immunobiology 2015; 220:227-235. [PMID: 25466587 PMCID: PMC4273735 DOI: 10.1016/j.imbio.2014.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 11/20/2014] [Accepted: 11/21/2014] [Indexed: 12/28/2022]
Abstract
Mucosal surfaces represent the largest areas of interactions of the host with its environment. Subsequently, the mucosal immune system has evolved complex strategies to maintain the integrity of the host by inducing protective immune responses against pathogenic and tolerance against dietary and commensal microbial antigens within the broad range of molecules the intestinal epithelium is exposed to. Among many other specialized cell subsets, myeloid cell populations - due to their strategic location in the subepithelial lamina propria - are the first ones to scavenge and process these intestinal antigens and to send consecutive signals to other immune and non-immune cell subsets. Thus, myeloid cell populations represent attractive targets for clinical intervention in chronic inflammatory bowel diseases (IBDs) such as ulcerative colitis (UC) and Crohn's disease (CD) as they initiate and modulate inflammatory or regulatory immune response and shape the intestinal T cell pool. Here, we discuss the interactions of the intestinal microbiota with dendritic cell and macrophage populations and review in this context the literature on four promising candidate molecules that are critical for the induction and maintenance of intestinal homeostasis on the one hand, but also for the initiation and propagation of chronic intestinal inflammation on the other.
Collapse
Affiliation(s)
- Regina Schey
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| | - Claudia Danzer
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA.
| |
Collapse
|
47
|
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: 3.8] [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
|
48
|
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: 48] [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
|
49
|
Dishaw LJ, Cannon JP, Litman GW, Parker W. Immune-directed support of rich microbial communities in the gut has ancient roots. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 47:36-51. [PMID: 24984114 PMCID: PMC4146740 DOI: 10.1016/j.dci.2014.06.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 05/30/2014] [Accepted: 06/21/2014] [Indexed: 05/12/2023]
Abstract
The animal gut serves as a primary location for the complex host-microbe interplay that is essential for homeostasis and may also reflect the types of ancient selective pressures that spawned the emergence of immunity in metazoans. In this review, we present a phylogenetic survey of gut host-microbe interactions and suggest that host defense systems arose not only to protect tissue directly from pathogenic attack but also to actively support growth of specific communities of mutualists. This functional dichotomy resulted in the evolution of immune systems much more tuned for harmonious existence with microbes than previously thought, existing as dynamic but primarily cooperative entities in the present day. We further present the protochordate Ciona intestinalis as a promising model for studying gut host-bacterial dialogue. The taxonomic position, gut physiology and experimental tractability of Ciona offer unique advantages in dissecting host-microbe interplay and can complement studies in other model systems.
Collapse
Affiliation(s)
- Larry J Dishaw
- Department of Pediatrics, University of South Florida Morsani College of Medicine, USF/ACH Children's Research Institute, 140 7th Avenue South, St. Petersburg, FL 33701, USA.
| | - John P Cannon
- Department of Pediatrics, University of South Florida Morsani College of Medicine, USF/ACH Children's Research Institute, 140 7th Avenue South, St. Petersburg, FL 33701, USA
| | - Gary W Litman
- Department of Pediatrics, University of South Florida Morsani College of Medicine, USF/ACH Children's Research Institute, 140 7th Avenue South, St. Petersburg, FL 33701, USA; Department of Molecular Genetics, All Children's Hospital-Johns Hopkins Medicine, 501 6th Avenue South, St. Petersburg, FL 33701, USA
| | - William Parker
- Department of Surgery, Duke University Medical Center, Box 2605, Durham, NC 27710, USA
| |
Collapse
|
50
|
NK cells in mucosal defense against infection. BIOMED RESEARCH INTERNATIONAL 2014; 2014:413982. [PMID: 25197644 PMCID: PMC4150440 DOI: 10.1155/2014/413982] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 07/31/2014] [Indexed: 01/06/2023]
Abstract
Conventional natural killer cells (NK cells) provide continual surveillance for cancer and rapid responses to infection. They develop in the bone marrow, emerge as either NK precursor cells, immature, or mature cells, and disperse throughout the body. In the periphery NK cells provide critical defense against pathogens and cancer and are noted to develop features of adaptive immune responses. In the tightly regulated and dynamic mucosal tissues, they set up residency via unknown mechanisms and from sources that are yet to be defined. Once resident, they appear to have the ability to functionally mature dependent on the mucosal tissue microenvironment. Mucosal NK cells play a pivotal role in early protection through their cytolytic function and IFNγ production against bacteria, fungi, viruses, and parasitic infections. This review presents what is known about NK cell development and phenotypes of mucosal tissue resident conventional NK cells. The question of how they come to reside in their tissues and published data on their function against pathogens during mucosal infection are discussed. Dissecting major questions highlighted in this review will be important to the further understanding of NK cell homing and functional diversity and improve rational design of NK cell based therapies against mucosal infection.
Collapse
|