51
|
Hopkins EGD, Frankel G. Overview of the Effect of Citrobacter rodentium Infection on Host Metabolism and the Microbiota. Methods Mol Biol 2021; 2291:399-418. [PMID: 33704766 DOI: 10.1007/978-1-0716-1339-9_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Citrobacter rodentium is a natural enteric mouse pathogen that models human intestinal diseases, such as pathogenic E. coli infections, ulcerative colitis, and colon cancer. Upon reaching the monolayer of intestinal epithelial cells (IECs) lining the gut, a complex web of interactions between the host, the pathogen, and the microbiota ensues. A number of studies revealed surprisingly rapid changes in IEC bioenergetics upon infection, involving a switch from oxidative phosphorylation to aerobic glycolysis, leading to mucosal oxygenation and subsequent changes in microbiota composition. Microbiome studies have revealed a bloom in Enterobacteriaceae during C. rodentium infection in both resistant (i.e., C57BL/6) and susceptible (i.e., C3H/HeN) strains of mice concomitant with a depletion of butyrate-producing Clostridia. The emerging understanding that dysbiosis of cholesterol metabolism is induced by enteric infection further confirms the pivotal role immunometabolism plays in disease outcome. Inversely, the host and microbiota also impact upon the progression of infection, from the susceptibility of the distal colon to C. rodentium colonization to clearance of the pathogen, both via opsonization from the host adaptive immune system and out competition by the resident microbiota. Further complicating this compendium of interactions, C. rodentium exploits microbiota metabolites to fine-tune virulence gene expression and promote colonization. This chapter summarizes the current knowledge of the myriad of pathogen-host-microbiota interactions that occur during the progression of C. rodentium infection in mice and the broader implications of these findings on our understanding of enteric disease.
Collapse
Affiliation(s)
- Eve G D Hopkins
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London, UK
| | - Gad Frankel
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London, UK.
| |
Collapse
|
52
|
Abstract
Innate lymphoid cells (ILCs) are a large family of cells of the immune system that performs various functions in immune defense, inflammation, and tissue remodeling. As a part of the innate immune system, ILCs are a distinct form of lymphocytes different from T and B cells. ILCs can provide host defense against the source of infection and initiate the repair and remodeling processes to restore and maintain host body homeostasis. The number of patients with Crohn’s disease (CD) worldwide has continued to increase in recent years and this disease has brought sickness and death to many families. Numerous studies have found that ILCs also undergo a series of alternations during the development of CD and contribute to this disease. Despite this, the pathogenesis of CD is still not fully explained. So, we keep researching and exploring. In this review, we have closely linked the latest progress on ILCs and CD, and introduced, in detail, the specific roles of four different types of ILCs in CD. We also describe new progress in the pathogenesis of CD, with particular emphasis on the plasticity of ILC3s in this disease. These new studies and findings may provide new insights and breakthrough points for the treatment of CD.
Collapse
Affiliation(s)
- Ying Wu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
53
|
Transcriptional Regulation of Inflammasomes. Int J Mol Sci 2020; 21:ijms21218087. [PMID: 33138274 PMCID: PMC7663688 DOI: 10.3390/ijms21218087] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Inflammasomes are multimolecular complexes with potent inflammatory activity. As such, their activity is tightly regulated at the transcriptional and post-transcriptional levels. In this review, we present the transcriptional regulation of inflammasome genes from sensors (e.g., NLRP3) to substrates (e.g., IL-1β). Lineage-determining transcription factors shape inflammasome responses in different cell types with profound consequences on the responsiveness to inflammasome-activating stimuli. Pro-inflammatory signals (sterile or microbial) have a key transcriptional impact on inflammasome genes, which is largely mediated by NF-κB and that translates into higher antimicrobial immune responses. Furthermore, diverse intrinsic (e.g., circadian clock, metabolites) or extrinsic (e.g., xenobiotics) signals are integrated by signal-dependent transcription factors and chromatin structure changes to modulate transcriptionally inflammasome responses. Finally, anti-inflammatory signals (e.g., IL-10) counterbalance inflammasome genes induction to limit deleterious inflammation. Transcriptional regulations thus appear as the first line of inflammasome regulation to raise the defense level in front of stress and infections but also to limit excessive or chronic inflammation.
Collapse
|
54
|
Zhang Z, Zou J, Shi Z, Zhang B, Etienne-Mesmin L, Wang Y, Shi X, Shao F, Chassaing B, Gewirtz AT. IL-22-induced cell extrusion and IL-18-induced cell death prevent and cure rotavirus infection. Sci Immunol 2020; 5:eabd2876. [PMID: 33008915 PMCID: PMC7709835 DOI: 10.1126/sciimmunol.abd2876] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/10/2020] [Indexed: 12/22/2022]
Abstract
Bacterial flagellin can elicit production of TLR5-mediated IL-22 and NLRC4-mediated IL-18 cytokines that act in concert to cure and prevent rotavirus (RV) infection. This study investigated the mechanism by which these cytokines act to impede RV. Although IL-18 and IL-22 induce each other's expression, we found that IL-18 and IL-22 both impeded RV independently of one another and did so by distinct mechanisms that involved activation of their cognate receptors in intestinal epithelial cells (IEC). IL-22 drove IEC proliferation and migration toward villus tips, which resulted in increased extrusion of highly differentiated IEC that serve as the site of RV replication. In contrast, IL-18 induced cell death of RV-infected IEC thus directly interrupting the RV replication cycle, resulting in spewing of incompetent virus into the intestinal lumen and causing a rapid drop in the number of RV-infected IEC. Together, these actions resulted in rapid and complete expulsion of RV, even in hosts with severely compromised immune systems. These results suggest that a cocktail of IL-18 and IL-22 might be a means of treating viral infections that preferentially target short-lived epithelial cells.
Collapse
Affiliation(s)
- Zhan Zhang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences Georgia State University, Atlanta, GA 30303 USA
| | - Jun Zou
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences Georgia State University, Atlanta, GA 30303 USA
| | - Zhenda Shi
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences Georgia State University, Atlanta, GA 30303 USA
| | - Benyue Zhang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences Georgia State University, Atlanta, GA 30303 USA
| | - Lucie Etienne-Mesmin
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences Georgia State University, Atlanta, GA 30303 USA
- Université Clermont Auvergne, INRAe, UMR 454 MEDIS, F-63000 Clermont-Ferrand, France
| | - Yanling Wang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences Georgia State University, Atlanta, GA 30303 USA
| | - Xuyan Shi
- National Institute of Biological Sciences, Beijing 102206, China
| | - Feng Shao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Benoit Chassaing
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences Georgia State University, Atlanta, GA 30303 USA
- INSERM, U1016, team "Mucosal microbiota in chronic inflammatory diseases," Paris, France
- Université de Paris, Paris, France
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303 USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences Georgia State University, Atlanta, GA 30303 USA.
| |
Collapse
|
55
|
Castillo-Dela Cruz P, Wanek AG, Kumar P, An X, Elsegeiny W, Horne W, Fitch A, Burr AHP, Gopalakrishna KP, Chen K, Methé BA, Canna SW, Hand TW, Kolls JK. Intestinal IL-17R Signaling Constrains IL-18-Driven Liver Inflammation by the Regulation of Microbiome-Derived Products. Cell Rep 2020; 29:2270-2283.e7. [PMID: 31747600 PMCID: PMC6886715 DOI: 10.1016/j.celrep.2019.10.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 09/04/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022] Open
Abstract
Interleukin (IL)-17 signaling to the intestinal epithelium regulates the intestinal microbiome. Given the reported links between intestinal dysbiosis, bacterial translocation, and liver disease, we hypothesize that intestinal IL-17R signaling plays a critical role in mitigating hepatic inflammation. To test this, we study intestinal epithelium-specific IL-17RA-deficient mice in an immune-driven hepatitis model. At the naive state, these mice exhibit microbiome dysbiosis and increased translocation of bacterial products (CpG DNA), which drives liver IL-18 production. Upon disease induction, absence of enteric IL-17RA signaling exacerbates hepatitis and hepatocyte cell death. IL-18 is necessary for disease exacerbation and is associated with increased activated hepatic lymphocytes based on Ifng and Fasl expression. Thus, intestinal IL-17R regulates translocation of TLR9 ligands and constrains susceptibility to hepatitis. These data connect enteric Th17 signaling and the microbiome in hepatitis, with broader implications on the effects of impaired intestinal immunity and subsequent release of microbial products observed in other extra-intestinal pathologies. Castillo-dela Cruz et al. describe a unique protective role of intestinal IL-17RA in hepatitis. Disruption of intestinal IL-17RA signaling results in microbiome dysbiosis and translocation of bacterial products, specifically unmethylated CpG DNA, to the liver. This promotes IL-18 production and subsequent lymphocyte activation and cell death to exacerbate liver inflammation.
Collapse
Affiliation(s)
- Patricia Castillo-Dela Cruz
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Alanna G Wanek
- Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Pawan Kumar
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Xiaojing An
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Waleed Elsegeiny
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - William Horne
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Adam Fitch
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Ansen H P Burr
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Kathyayini P Gopalakrishna
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15213, USA
| | - Kong Chen
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Barbara A Methé
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Scott W Canna
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Timothy W Hand
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jay K Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
56
|
Couto MR, Gonçalves P, Magro F, Martel F. Microbiota-derived butyrate regulates intestinal inflammation: Focus on inflammatory bowel disease. Pharmacol Res 2020; 159:104947. [DOI: 10.1016/j.phrs.2020.104947] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/04/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
|
57
|
Zhou W, Sonnenberg GF. Activation and Suppression of Group 3 Innate Lymphoid Cells in the Gut. Trends Immunol 2020; 41:721-733. [PMID: 32646594 PMCID: PMC7395873 DOI: 10.1016/j.it.2020.06.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022]
Abstract
Group 3 innate lymphoid cells (ILC3s) have emerged as master regulators of intestinal health and tissue homeostasis in mammals. Through a diverse array of cytokines and cellular interactions, ILC3s crucially orchestrate lymphoid organogenesis, promote tissue protection or regeneration, facilitate antimicrobial responses, and directly regulate adaptive immunity. Further, translational studies have found that ILC3 responses are altered in the intestine of defined patient populations with chronic infectious, inflammatory, or metabolic diseases. Therefore, it is essential to broadly understand the signals that activate, suppress, or fine-tune ILC3s in the gut. Here, we discuss recent exciting advances in this field, integrate them into our current understanding of ILC3 biology, and highlight fundamental gaps in knowledge that require additional investigation.
Collapse
Affiliation(s)
- Wenqing Zhou
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gregory F Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
58
|
Wu N, Chen D, Sun H, Tan J, Zhang Y, Zhang T, Han Y, Liu H, Ouyang X, Yang XD, Niu X, Zhong J, Wang Z, Su B. MAP3K2 augments Th1 cell differentiation via IL-18 to promote T cell-mediated colitis. SCIENCE CHINA-LIFE SCIENCES 2020; 64:389-403. [PMID: 32737854 DOI: 10.1007/s11427-020-1720-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022]
Abstract
T cell-mediated immunity in the intestine is stringently controlled to ensure proper immunity against pathogenic microbes and to prevent autoimmunity, a known cause of inflammatory bowel disease. However, precisely how T cells regulate intestine immunity remains to be fully understood. In this study, we found that mitogen-activated protein kinase kinase kinase 2 (MAP3K2) is required for the CD4+ T cell-mediated inflammation in the intestine. Using a T cell transfer colitis model, we found that MAP3K2-deficient naïve CD4 T cells had a dramatically reduced ability to induce colitis compared to wild type T cells. In addition, significantly fewer IFN-γ- but more IL-17A-producing CD4+ T cells in the intestines of mice receiving MAP3K2-deficient T cells than in those from mice receiving wild type T cells was observed. Interestingly, under well-defined in vitro differentiation conditions, MAP3K2-deficient naïve T cells were not impaired in their ability to differentiate into Th1, Th17 and Treg. Furthermore, the MAP3K2-regulated colitis severity was mediated by Th1 but not Th17 cells in the intestine. At the molecular level, we showed that MAP3K2-mediated Th1 cell differentiation in the intestine was regulated by IL-18 and required specific JNK activation. Together, our study reveals a novel regulatory role of MAP3K2 in intestinal T cell immunity via the IL-18-MAP3K2-JNK axis and may provide a novel target for intervention in T cell-mediated colitis.
Collapse
Affiliation(s)
- Ningbo Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Dongping Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Hongxiang Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Jianmei Tan
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Yao Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Tianyu Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Yuheng Han
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Hongzhi Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Xinxing Ouyang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Xiao-Dong Yang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyin Niu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Jie Zhong
- Department of Gastroenterology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Zhengting Wang
- Department of Gastroenterology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China.
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China.
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
59
|
Prado MK, Fontanari C, Souza CO, Gardinassi LG, Zoccal KF, de Paula-Silva FW, Peti AP, Sorgi CA, Meirelles AF, Ramos SG, Alves-Filho JC, Faccioli LH. IL-22 Promotes IFN-γ-Mediated Immunity against Histoplasma capsulatum Infection. Biomolecules 2020; 10:E865. [PMID: 32517114 PMCID: PMC7356283 DOI: 10.3390/biom10060865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/28/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
Histoplasma capsulatum is the agent of histoplasmosis, one of the most frequent mycoses in the world. The infection initiates with fungal spore inhalation, transformation into yeasts in the lungs and establishment of a granulomatous disease, which is characterized by a Th1 response. The production of Th1 signature cytokines, such as IFN-γ, is crucial for yeast clearance from the lungs, and to prevent dissemination. Recently, it was demonstrated that IL-17, a Th17 signature cytokine, is also important for fungal control, particularly in the absence of Th1 response. IL-22 is another cytokine with multiple functions on host response and disease progression. However, little is known about the role of IL-22 during histoplasmosis. In this study, we demonstrated that absence of IL-22 affected the clearance of yeasts from the lungs and increased the spreading to the spleen. In addition, IL-22 deficient mice (Il22-/-) succumbed to infection, which correlated with reductions in the numbers of CD4+ IFN-γ+ T cells, reduced IFN-γ levels, and diminished nitric oxide synthase type 2 (NOS2) expression in the lungs. Importantly, treatment with rIFN-γ mitigated the susceptibility of Il22-/- mice to H. capsulatum infection. These data indicate that IL-22 is crucial for IFN-γ/NO production and resistance to experimental histoplasmosis.
Collapse
Affiliation(s)
- Morgana K.B. Prado
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil; (M.K.B.P.); (C.F.); (C.O.S.S.); (L.G.G.); (K.F.Z.); (F.W.G.P.-S.); (A.P.F.P.); (C.A.S.); (A.F.G.M.)
- Programa de Pós-Graduação em Imunologia Básica e Aplicada da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Caroline Fontanari
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil; (M.K.B.P.); (C.F.); (C.O.S.S.); (L.G.G.); (K.F.Z.); (F.W.G.P.-S.); (A.P.F.P.); (C.A.S.); (A.F.G.M.)
| | - Camila O.S. Souza
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil; (M.K.B.P.); (C.F.); (C.O.S.S.); (L.G.G.); (K.F.Z.); (F.W.G.P.-S.); (A.P.F.P.); (C.A.S.); (A.F.G.M.)
- Programa de Pós-Graduação em Imunologia Básica e Aplicada da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Luiz G. Gardinassi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil; (M.K.B.P.); (C.F.); (C.O.S.S.); (L.G.G.); (K.F.Z.); (F.W.G.P.-S.); (A.P.F.P.); (C.A.S.); (A.F.G.M.)
| | - Karina F. Zoccal
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil; (M.K.B.P.); (C.F.); (C.O.S.S.); (L.G.G.); (K.F.Z.); (F.W.G.P.-S.); (A.P.F.P.); (C.A.S.); (A.F.G.M.)
| | - Francisco W.G. de Paula-Silva
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil; (M.K.B.P.); (C.F.); (C.O.S.S.); (L.G.G.); (K.F.Z.); (F.W.G.P.-S.); (A.P.F.P.); (C.A.S.); (A.F.G.M.)
| | - Ana P.F. Peti
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil; (M.K.B.P.); (C.F.); (C.O.S.S.); (L.G.G.); (K.F.Z.); (F.W.G.P.-S.); (A.P.F.P.); (C.A.S.); (A.F.G.M.)
| | - Carlos A. Sorgi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil; (M.K.B.P.); (C.F.); (C.O.S.S.); (L.G.G.); (K.F.Z.); (F.W.G.P.-S.); (A.P.F.P.); (C.A.S.); (A.F.G.M.)
| | - Alyne F.G. Meirelles
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil; (M.K.B.P.); (C.F.); (C.O.S.S.); (L.G.G.); (K.F.Z.); (F.W.G.P.-S.); (A.P.F.P.); (C.A.S.); (A.F.G.M.)
| | - Simone G. Ramos
- Departamento de Patologia e Medicina Legal da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil;
| | - José C. Alves-Filho
- Departamento de Farmacologia da Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil;
| | - Lúcia H. Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil; (M.K.B.P.); (C.F.); (C.O.S.S.); (L.G.G.); (K.F.Z.); (F.W.G.P.-S.); (A.P.F.P.); (C.A.S.); (A.F.G.M.)
| |
Collapse
|
60
|
Park J, Hunter CA. The role of macrophages in protective and pathological responses to Toxoplasma gondii. Parasite Immunol 2020; 42:e12712. [PMID: 32187690 DOI: 10.1111/pim.12712] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/12/2020] [Accepted: 02/24/2020] [Indexed: 02/06/2023]
Abstract
The ability of Toxoplasma gondii to cause clinical disease in immune-competent and immune-deficient hosts coupled with its ease of use in vitro and availability of murine models has led to its use as a model organism to study how the immune system controls an intracellular infection. This article reviews the studies that established the role of the cytokine IFN-γ in the activation of macrophages to control T gondii and the events that lead to the mobilization and expansion of macrophage populations and their ability to limit parasite replication. Macrophages also have pro-inflammatory functions that promote protective NK and T-cell activities as well as regulatory properties that facilitate the resolution of inflammation. Nevertheless, while macrophages are important in determining the outcome of infection, T gondii has evolved mechanisms to subvert macrophage activation and can utilize their migratory activities to promote dissemination and these two properties underlie the ability of this parasite to persist and cause disease.
Collapse
Affiliation(s)
- Jeongho Park
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA.,Kangwon National University College of Veterinary Medicine and Institute of Veterinary Science, Chuncheon, Korea
| | | |
Collapse
|
61
|
Keir ME, Yi T, Lu TT, Ghilardi N. The role of IL-22 in intestinal health and disease. J Exp Med 2020; 217:e20192195. [PMID: 32997932 PMCID: PMC7062536 DOI: 10.1084/jem.20192195] [Citation(s) in RCA: 223] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/24/2022] Open
Abstract
The cytokine interleukin-22 (IL-22) is a critical regulator of epithelial homeostasis. It has been implicated in multiple aspects of epithelial barrier function, including regulation of epithelial cell growth and permeability, production of mucus and antimicrobial proteins (AMPs), and complement production. In this review, we focus specifically on the role of IL-22 in the intestinal epithelium. We summarize recent advances in our understanding of how IL-22 regulates homeostasis and host defense, and we discuss the IL-22 pathway as a therapeutic target in diseases of the intestine, including inflammatory bowel disease (IBD), graft-versus-host disease (GVHD), and cancer.
Collapse
Affiliation(s)
- Mary E. Keir
- Biomarker Discovery, Genentech, South San Francisco, CA
| | - Tangsheng Yi
- Department of Immunology, Genentech, South San Francisco, CA
| | - Timothy T. Lu
- Early Clinical Development, Genentech, South San Francisco, CA
| | | |
Collapse
|
62
|
Emerging Roles for Interleukin-18 in the Gastrointestinal Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1240:59-72. [PMID: 32060888 DOI: 10.1007/978-3-030-38315-2_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Interleukin (IL)-18, a member of the IL-1 family of cytokines, has emerged as a key regulator of mucosal homeostasis within the gastrointestinal tract. Like other members of this family, IL-18 is secreted as an inactive protein and is processed into its active form by caspase-1, although other contributors to precursor processing are emerging.Numerous studies have evaluated the role of IL-18 within the gastrointestinal tract using genetic or complementary pharmacological tools and have revealed multiple roles in tumorigenesis. Most striking among these are the divergent roles for IL-18 in colon and gastric cancers. Here, we review our current understanding of IL-18 biology and how this applies to colorectal and gastric cancers.
Collapse
|
63
|
Mühl H, Bachmann M. IL-18/IL-18BP and IL-22/IL-22BP: Two interrelated couples with therapeutic potential. Cell Signal 2019; 63:109388. [PMID: 31401146 DOI: 10.1016/j.cellsig.2019.109388] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023]
Abstract
Interleukin (IL)-18 and IL-22 are key components of cytokine networks that play a decisive role in (pathological) inflammation, host defense, and tissue regeneration. Tight regulation of cytokine-driven signaling, inflammation, and immunoactivation is supposed to enable nullification of a given deleterious trigger without mediating overwhelming collateral tissue damage or even activating a cancerous face of regeneration. In fact, feedback regulation by specific cytokine opponents is regarded as a major means by which the immune system is kept in balance. Herein, we shine a light on the interplay between IL-18 and IL-22 and their opponents IL-18 binding protein (IL-18BP) and IL-22BP in order to provide integrated information on their biology, pathophysiological significance, and prospect as targets and/or instruments of therapeutic intervention.
Collapse
Affiliation(s)
- Heiko Mühl
- pharmazentrum frankfurt/ZAFES, University Hospital Goethe University Frankfurt am Main, Theodor-Stern- Kai 7, 60590 Frankfurt am Main, Germany.
| | - Malte Bachmann
- pharmazentrum frankfurt/ZAFES, University Hospital Goethe University Frankfurt am Main, Theodor-Stern- Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
64
|
Winsor N, Krustev C, Bruce J, Philpott DJ, Girardin SE. Canonical and noncanonical inflammasomes in intestinal epithelial cells. Cell Microbiol 2019; 21:e13079. [PMID: 31265745 DOI: 10.1111/cmi.13079] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022]
Abstract
Inflammasomes are cytosolic, multimeric protein complexes capable of activating pro-inflammatory cytokines such as IL-1β and IL-18, which play a key role in host defence. Inflammasome components are highly expressed in the intestinal epithelium. In recent years, studies have begun to demonstrate that epithelial-intrinsic inflammasomes play a critical role in regulating epithelial homeostasis, both by defending the epithelium from pathogenic insult and through the regulation of the mucosal environment. However, the majority of research regarding inflammasome activation has focused on professional immune cells, such as macrophages. Here, we present an overview of the current understanding of inflammasome function in epithelial cells and at mucosal surfaces and, in particular, in the intestine.
Collapse
Affiliation(s)
- Nathaniel Winsor
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Christian Krustev
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jessica Bruce
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Stephen E Girardin
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
65
|
Induction of Inflammatory Macrophages in the Gut and Extra-Gut Tissues by Colitis-Mediated Escherichia coli. iScience 2019; 21:474-489. [PMID: 31707260 PMCID: PMC6849333 DOI: 10.1016/j.isci.2019.10.046] [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: 04/01/2019] [Revised: 09/13/2019] [Accepted: 10/23/2019] [Indexed: 12/02/2022] Open
Abstract
Inflammatory macrophages play a critical role in gut and extra-gut inflammatory disorders, which may be promoted through the dysbiosis of gut microbiota. However, it is poorly understood how gut microbiota affect inflammatory macrophages. Here, we found that increased Escherichia coli (E. coli) in inflamed colon may induce inflammatory macrophages in gut and extra-gut tissues. These E. coli are different from other commensal and pathogenic E. coli in genomic components and also in ability to induce inflammatory responses. Dominant E. coli from colitic tissues induce gut inflammatory macrophages through a regulating network consisted of IL-18, IFN-γ, IL-12, and IL-22 in gut tissues. These E. coli also directly activate macrophages. Cytosolic inflammasome components PCKδ, NLRC4, caspase8, and caspase1/11 are involved in E. coli-mediated activation in both gut epithelial cells and macrophages. These disclose a novel mechanism for how dysbiosis of gut microbiota in colitis cause inflammatory macrophages related to multiple diseases. Increased commensal E. coli in colitis induce inflammatory macrophages Colitic E. coli are different from other commensal and pathogenic E. coli Gut inflammatory macrophages by E. coli need IL-18, IFN-γ, IL-12, and IL-22 PCKδ, NLRC4, caspase8, and caspase1/11 are required for E. coli-mediated activation
Collapse
|
66
|
Wang Q, Li J, Yu TS, Liu Y, Li K, Liu S, Liu Y, Feng Q, Zhang L, Li GS, Shao LL, Peng J, Hou M, Liu XG. Disrupted balance of CD4 + T-cell subsets in bone marrow of patients with primary immune thrombocytopenia. Int J Biol Sci 2019; 15:2798-2814. [PMID: 31853219 PMCID: PMC6909963 DOI: 10.7150/ijbs.33779] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 10/05/2019] [Indexed: 12/18/2022] Open
Abstract
Disequilibrium of CD4+ T-cell subpopulations in peripheral blood (PB) of patients with primary immune thrombocytopenia (ITP) has been well established, whereas the profile of CD4+ T-cell subpopulations in bone marrow (BM) remains elusive. In the present study, the frequencies of T helper 22 (Th22), Th17, Th1, Th2, follicular T helper (Tfh) cells and regulatory T cells (Tregs) as well as their effector cytokines in BM and PB from active ITP patients and healthy controls (HCs) were determined. Results showed that the frequencies of Th22, Th17, Th1, and Tfh cells were significantly higher, but Treg number was remarkably lower in BM from ITP patients than from HCs. In the ITP group, it was notable that the numbers of BM Th22, Th17, Th1, Th2, and Tfh cells were significantly elevated compared with the matched PB counterparts, while Treg number in BM was considerably reduced compared with that in PB. In consistence with the BM Th subset pattern, plasma levels of interleukin (IL)-22, IL-17A, and interferon (INF)-γ in BM from ITP patients were significantly increased compared with that from HCs. Therefore, the balance of CD4+ T-cell subsets was disrupted in both BM and PB of ITP patients, suggesting that this might play important roles in the pathophysiological process of ITP.
Collapse
Affiliation(s)
- Qian Wang
- Department of Hematology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Jinan, P. R. China.,Department of Clinical Laboratory, Qilu Hospital, Shandong University (Qingdao), 758 Hefei Road, Qingdao, P. R. China
| | - Juan Li
- Department of Clinical Laboratory, Qilu Hospital, Shandong University (Qingdao), 758 Hefei Road, Qingdao, P. R. China
| | - Tian-Shu Yu
- Department of Hematology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Jinan, P. R. China
| | - Yu Liu
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, 3501 Daxue Road, Jinan, P. R. China
| | - Kai Li
- Department of Radiotherapy, Zhangqiu People's Hospital, 1920 Huiquan Road, Jinan, P. R. China
| | - Shuang Liu
- Department of Hematology, Taian Central Hospital, Taian, P. R. China
| | - Yang Liu
- Department of Hematology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Jinan, P. R. China
| | - Qi Feng
- Department of Hematology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Jinan, P. R. China
| | - Lei Zhang
- Department of Orthopedics, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Guo-Sheng Li
- Department of Hematology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Jinan, P. R. China
| | - Lin-Lin Shao
- Department of Hematology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Jinan, P. R. China
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Jinan, P. R. China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Jinan, P. R. China.,Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Jinan, China
| | - Xin-Guang Liu
- Department of Hematology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Jinan, P. R. China
| |
Collapse
|
67
|
Borghi M, Pariano M, Solito V, Puccetti M, Bellet MM, Stincardini C, Renga G, Vacca C, Sellitto F, Mosci P, Brancorsini S, Romani L, Costantini C. Targeting the Aryl Hydrocarbon Receptor With Indole-3-Aldehyde Protects From Vulvovaginal Candidiasis via the IL-22-IL-18 Cross-Talk. Front Immunol 2019; 10:2364. [PMID: 31681274 PMCID: PMC6798081 DOI: 10.3389/fimmu.2019.02364] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/20/2019] [Indexed: 12/21/2022] Open
Abstract
Vulvovaginal candidiasis (VVC) is a common mucosal infection caused by Candida spp., most frequently by Candida albicans, which may become recurrent and severely impacting the quality of life of susceptible women. Although it is increasingly being recognized that mucosal damage is mediated by an exaggerated inflammatory response, current therapeutic approaches are only based on antifungals that may relieve the symptomatology, but fail to definitely prevent recurrences. The unrestrained activation of the NLRP3 inflammasome with continuous production of IL-1β and recruitment of neutrophils is recognized as a pathogenic factor in VVC. We have previously shown that IL-22 is required to dampen pathogenic inflammasome activation in VVC via the NLRC4/IL-1Ra axis. However, IL-22 also regulates IL-18, a product of the inflammasome activity that regulates IL-22 expression. Here we describe a cross-regulatory circuit between IL-18 and IL-22 in murine VVC that is therapeutically druggable. We found that IL-18 production was dependent on IL-22 and NLRC4, and that IL-18, in turn, contributes to IL-22 activity. Like in IL-22 deficiency, IL-18 deficiency was associated with an increased susceptibility to VVC and unbalanced Th17/Treg response, suggesting that IL-18 can regulate both the innate and the adaptive responses to the fungus. Administration of the microbial metabolite indole-3-aldehyde, known to stimulate the production of IL-22 via the aryl hydrocarbon receptor (AhR), promoted IL-18 expression and protection against Candida infection. Should low levels of IL-18 be demonstrated in the vaginal fluids of women with recurrent VVC, targeting the AhR/IL-22/IL-18 pathway could be exploited for future therapeutic approaches in VVC. This study suggests that a deeper understanding of the mechanisms regulating inflammasome activity may lead to the identification of novel targets for intervention in VVC.
Collapse
MESH Headings
- Animals
- Basic Helix-Loop-Helix Transcription Factors/agonists
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/immunology
- Candida albicans/immunology
- Candidiasis, Vulvovaginal/drug therapy
- Candidiasis, Vulvovaginal/genetics
- Candidiasis, Vulvovaginal/immunology
- Candidiasis, Vulvovaginal/pathology
- Female
- Indoles/pharmacology
- Inflammasomes/genetics
- Inflammasomes/immunology
- Interleukin-18/genetics
- Interleukin-18/immunology
- Interleukins/genetics
- Interleukins/immunology
- Mice
- Mice, Knockout
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/immunology
- Receptors, Aryl Hydrocarbon/agonists
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/immunology
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Th17 Cells/immunology
- Th17 Cells/pathology
- Interleukin-22
Collapse
Affiliation(s)
- Monica Borghi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Valentina Solito
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Marina M. Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Carmine Vacca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Federica Sellitto
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Paolo Mosci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | | | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
68
|
Di Luccia B, Gilfillan S, Cella M, Colonna M, Huang SCC. ILC3s integrate glycolysis and mitochondrial production of reactive oxygen species to fulfill activation demands. J Exp Med 2019; 216:2231-2241. [PMID: 31296736 PMCID: PMC6781001 DOI: 10.1084/jem.20180549] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/12/2019] [Accepted: 06/19/2019] [Indexed: 12/29/2022] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are the innate counterparts of Th17 that require the transcription factor RORγt for development and contribute to the defense against pathogens through IL-22 and IL-17 secretion. Proliferation and effector functions of Th17 require a specific mTOR-dependent metabolic program that utilizes high-rate glycolysis, while mitochondrial lipid oxidation and production of reactive oxygen species (mROS) support alternative T reg cell differentiation. Whether ILC3s employ a specific metabolic program is not known. Here, we find that ILC3s rely on mTOR complex 1 (mTORC1) for proliferation and production of IL-22 and IL-17A after in vitro activation and Citrobacter rodentium infection. mTORC1 induces activation of HIF1α, which reprograms ILC3 metabolism toward glycolysis and sustained expression of RORγt. However, in contrast to Th17, ILC3 activation requires mROS production; rather than inducing an alternative regulatory fate as it does in CD4 T cells, mROS stabilizes HIF1α and RORγt in ILC3s and thereby promotes their activation. We conclude that ILC3 activation relies on a metabolic program that integrates glycolysis with mROS production.
Collapse
Affiliation(s)
- Blanda Di Luccia
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Marina Cella
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Stanley Ching-Cheng Huang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| |
Collapse
|
69
|
Citrobacter rodentium-host-microbiota interactions: immunity, bioenergetics and metabolism. Nat Rev Microbiol 2019; 17:701-715. [PMID: 31541196 DOI: 10.1038/s41579-019-0252-z] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2019] [Indexed: 12/26/2022]
Abstract
Citrobacter rodentium is an extracellular enteric mouse-specific pathogen used to model infections with human pathogenic Escherichia coli and inflammatory bowel disease. C. rodentium injects type III secretion system effectors into intestinal epithelial cells (IECs) to target inflammatory, metabolic and cell survival pathways and establish infection. While the host responds to infection by activating innate and adaptive immune signalling, required for clearance, the IECs respond by rapidly shifting bioenergetics to aerobic glycolysis, which leads to oxygenation of the epithelium, an instant expansion of mucosal-associated commensal Enterobacteriaceae and a decline of obligate anaerobes. Moreover, infected IECs reprogramme intracellular metabolic pathways, characterized by simultaneous activation of cholesterol biogenesis, import and efflux, leading to increased serum and faecal cholesterol levels. In this Review we summarize recent advances highlighting the intimate relationship between C. rodentium pathogenesis, metabolism and the gut microbiota.
Collapse
|
70
|
Šefcová M, Levkut M, Bobíková K, Karaffová V, Revajová V, Maruščáková IC, Levkutová M, Ševčíková Z, Herich R, Levkut M. Cytokine response after stimulation of culture cells by zinc and probiotic strain. In Vitro Cell Dev Biol Anim 2019; 55:830-837. [PMID: 31520371 DOI: 10.1007/s11626-019-00401-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/08/2019] [Indexed: 01/06/2023]
Abstract
Intestinal porcine epithelial cells were used for an in vitro analysis of mRNA expression levels of inflammatory cytokines (IL-8, IL-18) and transcriptional factors (MyD88 and NF-κβ). Cells were exposed to inorganic and organic zinc sources (in two different concentrations-50 μmol/L and 100 μmol/L) alone or combined with Lactobacillus reuteri B6/1, which was also applied individually. The total exposure time was 4 h. Quantitative reverse transcriptase PCR was used to determine expression levels of the aforementioned parameters. In general, upregulation was observed; however, a decrease of some mRNA's abundance was also determined. Differences in expression were analysed statistically using ANOVA and Tukey analyses. High relative expression was shown for IL-8, IL-18 and MyD88 in groups treated with 100 μmol/L of inorganic sources of zinc (ZnSO4) (p < 0.05), while groups treated with the organic form did not exhibit significant changes in expression. Also, 50 μmol/L of either zinc source did not significantly modify the transcriptional profile of the cytokines and transcription factors, showing that even inorganic sources, at lower concentrations, do not elicit a significant inflammatory reaction. In summary, supplementation of organic zinc source (Gly-Zn chelate) ensures that IL-8, IL-18, MyD88 and NF-κβ expression levels are not positively regulated. In contrast, inorganic sources of zinc (ZnSO4) could induce an inflammatory reaction. However, this response could be dampened if L. reuteri B6/1 is administered, showing the helpful aspect of using probiotics to modulate an inflammatory response. Conclusively, the use Gly-Zn chelate appears as an optimal alternative for Zn administration that does not compromise normal intestinal homeostasis.
Collapse
Affiliation(s)
- Miroslava Šefcová
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic.
| | - Martin Levkut
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Katarína Bobíková
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Viera Karaffová
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Viera Revajová
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Ivana Cingeľová Maruščáková
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Mária Levkutová
- Department of Epizootiology and Parasitology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Zuzana Ševčíková
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Róbert Herich
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
| | - Mikuláš Levkut
- Department of Pathological Anatomy and Pathological Physiology, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181, Košice, Slovak Republic
- Institute of Neuroimmunology, Slovak Academy of Science, Dúbravská cesta 9, 84510, Bratislava, Slovak Republic
| |
Collapse
|
71
|
Goto Y. Epithelial Cells as a Transmitter of Signals From Commensal Bacteria and Host Immune Cells. Front Immunol 2019; 10:2057. [PMID: 31555282 PMCID: PMC6724641 DOI: 10.3389/fimmu.2019.02057] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022] Open
Abstract
Intestinal epithelial cells (IECs) are non-hematopoietic cells that form a physical barrier against external antigens. Recent studies indicate that IECs have pleiotropic functions in the regulation of luminal microbiota and the host immune system. IECs produce various immune modulatory cytokines and chemokines in response to commensal bacteria and contribute to developing the intestinal immune system. In contrast, IECs receive cytokine signals from immune cells and produce various immunological factors against luminal bacteria. This bidirectional function of IECs is critical to regulate homeostasis of microbiota and the host immune system. Disruption of the epithelial barrier leads to detrimental host diseases such as inflammatory bowel disease, colonic cancer, and pathogenic infection. This review provides an overview of the functions and physiology of IECs and highlights their bidirectional functions against luminal bacteria and immune cells, which contribute to maintaining gut homeostasis.
Collapse
Affiliation(s)
- Yoshiyuki Goto
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan.,Division of Mucosal Symbiosis, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
72
|
Volk JK, Nyström EEL, van der Post S, Abad BM, Schroeder BO, Johansson Å, Svensson F, Jäverfelt S, Johansson MEV, Hansson GC, Birchenough GMH. The Nlrp6 inflammasome is not required for baseline colonic inner mucus layer formation or function. J Exp Med 2019; 216:2602-2618. [PMID: 31420376 PMCID: PMC6829596 DOI: 10.1084/jem.20190679] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/06/2019] [Accepted: 07/22/2019] [Indexed: 12/26/2022] Open
Abstract
It is thought that the Nlrp6 inflammasome regulates formation of the inner mucus layer (IML) barrier that prohibits contact between the microbiota and colonic epithelial cells. Using microbiota-controlled mice and combined ex vivo/in vivo IML analytical approaches, Volk et al. delineate the relative roles of the inflammasome and microbiota in shaping IML formation and function. The inner mucus layer (IML) is a critical barrier that protects the colonic epithelium from luminal threats and inflammatory bowel disease. Innate immune signaling is thought to regulate IML formation via goblet cell Nlrp6 inflammasome activity that controls secretion of the mucus structural component Muc2. We report that isolated colonic goblet cells express components of several inflammasomes; however, analysis of IML properties in multiple inflammasome-deficient mice, including littermate-controlled Nlrp6−/−, detect a functional IML barrier in all strains. Analysis of mice lacking inflammasome substrate cytokines identifies a defective IML in Il18−/− mice, but this phenotype is ultimately traced to a microbiota-driven, Il18-independent effect. Analysis of phenotypic transfer between IML-deficient and IML-intact mice finds that the Bacteroidales family S24-7 (Muribaculaceae) and genus Adlercrutzia consistently positively covary with IML barrier function. Together, our results demonstrate that baseline IML formation and function is independent of inflammasome activity and highlights the role of the microbiota in determining IML barrier function.
Collapse
Affiliation(s)
- Joana K Volk
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Elisabeth E L Nyström
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Sjoerd van der Post
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Beatriz M Abad
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Bjoern O Schroeder
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Åsa Johansson
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Frida Svensson
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Sofia Jäverfelt
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Malin E V Johansson
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Gunnar C Hansson
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - George M H Birchenough
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
73
|
Castellanos JG, Longman RS. The balance of power: innate lymphoid cells in tissue inflammation and repair. J Clin Invest 2019; 129:2640-2650. [PMID: 31180335 DOI: 10.1172/jci124617] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Over the last ten years, immunologists have recognized the central importance of an emerging group of innate lymphoid cells (ILCs) in health and disease. Characterization of these cells has provided a molecular definition of ILCs and their tissue-specific functions. Although the lineage-defining transcription factors, cytokine production, and nomenclature parallel those of T helper cells, ILCs do not require adaptive immune programming. Both environmental and host-derived signals shape the function of these evolutionarily ancient cells, which provide pathogen protection and promote tissue restoration. As such, ILCs function as a double-edged sword, balancing the inflammatory and reparative responses that arise during injury and disease. This Review highlights our recent understanding of tissue-resident ILCs and the signals that regulate their contribution to inflammation and tissue repair in health and disease.
Collapse
|
74
|
McEntee CP, Finlay CM, Lavelle EC. Divergent Roles for the IL-1 Family in Gastrointestinal Homeostasis and Inflammation. Front Immunol 2019; 10:1266. [PMID: 31231388 PMCID: PMC6568214 DOI: 10.3389/fimmu.2019.01266] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/17/2019] [Indexed: 12/11/2022] Open
Abstract
Inflammatory disorders of the gastro-intestinal tract are a major cause of morbidity and significant burden from a health and economic perspective in industrialized countries. While the incidence of such conditions has a strong environmental component, in particular dietary composition, epidemiological studies have identified specific hereditary mutations which result in disequilibrium between pro- and anti-inflammatory factors. The IL-1 super-family of cytokines and receptors is highly pleiotropic and plays a fundamental role in the pathogenesis of several auto-inflammatory conditions including rheumatoid arthritis, multiple sclerosis and psoriasis. However, the role of this super-family in the etiology of inflammatory bowel diseases remains incompletely resolved despite extensive research. Herein, we highlight the currently accepted paradigms as they pertain to specific IL-1 family members and focus on some recently described non-classical roles for these pathways in the gastrointestinal tract. Finally, we address some of the shortcomings and sources of variance in the field which to date have yielded several conflicting results from similar studies and discuss the potential effect of these factors on data interpretation.
Collapse
Affiliation(s)
- Craig P McEntee
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Conor M Finlay
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Faculty of Biology, Medicine and Health, Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
75
|
Wang S, El-Fahmawi A, Christian DA, Fang Q, Radaelli E, Chen L, Sullivan MC, Misic AM, Ellringer JA, Zhu XQ, Winter SE, Hunter CA, Beiting DP. Infection-Induced Intestinal Dysbiosis Is Mediated by Macrophage Activation and Nitrate Production. mBio 2019; 10:e00935-19. [PMID: 31138751 PMCID: PMC6538788 DOI: 10.1128/mbio.00935-19] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 04/24/2019] [Indexed: 01/08/2023] Open
Abstract
Oral infection of C57BL/6J mice with Toxoplasma gondii results in a marked bacterial dysbiosis and the development of severe pathology in the distal small intestine that is dependent on CD4+ T cells and interferon gamma (IFN-γ). This dysbiosis and bacterial translocation contribute to the development of ileal pathology, but the factors that support the bloom of bacterial pathobionts are unclear. The use of microbial community profiling and shotgun metagenomics revealed that Toxoplasma infection induces a dysbiosis dominated by Enterobacteriaceae and an increased potential for nitrate respiration. In vivo experiments using bacterial metabolic mutants revealed that during this infection, host-derived nitrate supports the expansion of Enterobacteriaceae in the ileum via nitrate respiration. Additional experiments with infected mice indicate that the IFN-γ/STAT1/iNOS axis, while essential for parasite control, also supplies a pool of nitrate that serves as a source for anaerobic respiration and supports overgrowth of Enterobacteriaceae Together, these data reveal a trade-off in intestinal immunity after oral infection of C57BL/6J mice with T. gondii, in which inducible nitric oxide synthase (iNOS) is required for parasite control, while this host enzyme is responsible for specific modification of the composition of the microbiome that contributes to pathology.IMPORTANCEToxoplasma gondii is a protozoan parasite and a leading cause of foodborne illness. Infection is initiated when the parasite invades the intestinal epithelium, and in many host species, this leads to intense inflammation and a dramatic disruption of the normal microbial ecosystem that resides in the healthy gut (the so-called microbiome). One characteristic change in the microbiome during infection with Toxoplasma-as well as numerous other pathogens-is the overgrowth of Escherichia coli or similar bacteria and a breakdown of commensal containment leading to seeding of peripheral organs with gut bacteria and subsequent sepsis. Our findings provide one clear explanation for how this process is regulated, thereby improving our understanding of the relationship between parasite infection, inflammation, and disease. Furthermore, our results could serve as the basis for the development of novel therapeutics to reduce the potential for harmful bacteria to bloom in the gut during infection.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Ayah El-Fahmawi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Qun Fang
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Longfei Chen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Megan C Sullivan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ana M Misic
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jodi A Ellringer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Sebastian E Winter
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
76
|
Williams MA, O'Callaghan A, Corr SC. IL-33 and IL-18 in Inflammatory Bowel Disease Etiology and Microbial Interactions. Front Immunol 2019; 10:1091. [PMID: 31139196 PMCID: PMC6527769 DOI: 10.3389/fimmu.2019.01091] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/29/2019] [Indexed: 12/19/2022] Open
Abstract
The IL-1 cytokines are a newly expanded family, with each of its 11 members playing an important role in health and disease. Typically acting as pro- or anti-inflammatory mediators of first-line innate immunity, their production is particularly important in the context of mucosal defenses, through handling breach of the delicate epithelial barrier and mediating a local immune response to invading pathogens. Mucosal immunity is often aberrantly orchestrated in intestinal diseases, such as Inflammatory Bowel Disease (IBD). Various studies have pointed to IL-1 cytokines as being important players in IBD with context-dependent roles, either through promoting auto-inflammatory mechanisms, or alleviating disease through protection against breach of pathogens across the epithelial barrier. This mini-review will succinctly examine the role of IL-1 family members in IBD, with a special focus on the recently described IL-33 as well as IL-18, and will explore the disease models within which these cytokines have been studied. Furthermore, we will examine the evidence of interplay of these cytokines with the gut microbiota, with hopes of summarizing our current knowledge of these family members and their potential for unraveling novel molecular mechanisms of IBD pathology.
Collapse
Affiliation(s)
- Michelle A Williams
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventative Medicine, Trinity College Dublin, Dublin, Ireland
| | - Amy O'Callaghan
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventative Medicine, Trinity College Dublin, Dublin, Ireland
| | - Sinéad C Corr
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventative Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
77
|
Ouyang W, O'Garra A. IL-10 Family Cytokines IL-10 and IL-22: from Basic Science to Clinical Translation. Immunity 2019; 50:871-891. [PMID: 30995504 DOI: 10.1016/j.immuni.2019.03.020] [Citation(s) in RCA: 600] [Impact Index Per Article: 120.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/01/2019] [Accepted: 03/21/2019] [Indexed: 12/12/2022]
Abstract
Cytokines are among the most important effector and messenger molecules in the immune system. They profoundly participate in immune responses during infection and inflammation, protecting against or contributing to diseases such as allergy, autoimmunity, and cancer. Manipulating cytokine pathways, therefore, is one of the most effective strategies to treat various diseases. IL-10 family cytokines exert essential functions to maintain tissue homeostasis during infection and inflammation through restriction of excessive inflammatory responses, upregulation of innate immunity, and promotion of tissue repairing mechanisms. Their important functions in diseases are supported by data from many preclinical models, human genetic studies, and clinical interventions. Despite significant efforts, however, there is still no clinically approved therapy through manipulating IL-10 family cytokines. Here, we summarize the recent progress in understanding the biology of this family of cytokines, suggesting more specific strategies to maneuver these cytokines for the effective treatment of inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Wenjun Ouyang
- Department of Inflammation and Oncology Research, Amgen, South San Francisco, CA 94080, USA.
| | - Anne O'Garra
- Laboratory of Immunoregulation and Infection, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
78
|
Heimesaat MM, Dunay IR, Bereswill S. Comprehensive Kinetic Survey of Intestinal, Extra-Intestinal and Systemic Sequelae of Murine Ileitis Following Peroral Low-Dose Toxoplasma gondii Infection. Front Cell Infect Microbiol 2019; 9:98. [PMID: 31032232 PMCID: PMC6474322 DOI: 10.3389/fcimb.2019.00098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023] Open
Abstract
We have recently shown that following peroral low-dose Toxoplasma gondii infection susceptible mice develop subacute ileitis within 10 days. Data regarding long-term intestinal and extra-intestinal sequelae of infection are scarce, however. We therefore challenged conventional C57BL/6 mice with one cyst of T. gondii ME49 strain by gavage and performed a comprehensive immunopathological survey 10, 36, and 57 days later. As early as 10 days post-infection, mice were suffering from subacute ileitis as indicated by mild-to-moderate histopathological changes of the ileal mucosa. Furthermore, numbers of apoptotic and proliferating/regenerating epithelial cells as well as of T and B lymphocytes in the mucosa and lamina propria of the ileum were highest at day 10 post-infection, but declined thereafter, and were accompanied by enhanced pro-inflammatory mediator secretion in ileum, colon and mesenteric lymph nodes that was most pronounced during the early phase of infection. In addition, subacute ileitis was accompanied by distinct shifts in the commensal gut microbiota composition in the small intestines. Remarkably, immunopathological sequelae of T. gondii infection were not restricted to the intestines, but could also be observed in extra-intestinal tissues including the liver, kidneys, lungs, heart and strikingly, in systemic compartments that were most prominent at day 10 post-infection. We conclude that the here provided long-term kinetic survey of immunopathological sequalae following peroral low-dose T. gondii infection provides valuable corner stones for a better understanding of the complex interactions within the triangle relationship of (parasitic) pathogens, the host immunity and the commensal gut microbiota during intestinal inflammation. The low-dose T. gondii infection model may be applied as valuable gut inflammation model in future pre-clinical studies in order to test potential treatment options for intestinal inflammatory conditions in humans.
Collapse
Affiliation(s)
- Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ildiko R Dunay
- Medical Faculty, Institute of Inflammation and Neurodegeneration, University Hospital Magdeburg, Magdeburg, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
79
|
Intestinal Epithelial Cells and the Microbiome Undergo Swift Reprogramming at the Inception of Colonic Citrobacter rodentium Infection. mBio 2019; 10:mBio.00062-19. [PMID: 30940698 PMCID: PMC6445932 DOI: 10.1128/mbio.00062-19] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mouse pathogen C. rodentium is a widely used model for colonic infection and has been a major tool in fundamental discoveries in the fields of bacterial pathogenesis and mucosal immunology. Despite extensive studies probing acute C. rodentium infection, our understanding of the early stages preceding the infection climax remains relatively undetailed. To this end, we apply a multiomics approach to resolve temporal changes to the host and microbiome during early infection. Unexpectedly, we found immediate and dramatic responses occurring on the day of colonic infection, both in the host intestinal epithelial cells and in the microbiome. Our study suggests changes in cholesterol and carbon metabolism in epithelial cells are instantly induced upon pathogen detection in the colon, corresponding with a shift to primarily facultative anaerobes constituting the microbiome. This study contributes to our knowledge of disease pathogenesis and mechanisms of barrier regulation, which is required for development of novel therapeutics targeting the intestinal epithelium. We used the mouse attaching and effacing (A/E) pathogen Citrobacter rodentium, which models the human A/E pathogens enteropathogenic Escherichia coli and enterohemorrhagic E. coli (EPEC and EHEC), to temporally resolve intestinal epithelial cell (IEC) responses and changes to the microbiome during in vivo infection. We found the host to be unresponsive during the first 3 days postinfection (DPI), when C. rodentium resides in the caecum. In contrast, at 4 DPI, the day of colonic colonization, despite only sporadic adhesion to the apex of the crypt, we observed robust upregulation of cell cycle and DNA repair processes, which were associated with expansion of the crypt Ki67-positive replicative zone, and downregulation of multiple metabolic processes (including the tricarboxylic acid [TCA] cycle and oxidative phosphorylation). Moreover, we observed dramatic depletion of goblet and deep crypt secretory cells and an atypical regulation of cholesterol homeostasis in IECs during early infection, with simultaneous upregulation of cholesterol biogenesis (e.g., 3-hydroxy-3-methylglutaryl–coenzyme A reductase [Hmgcr]), import (e.g., low-density lipoprotein receptor [Ldlr]), and efflux (e.g., AbcA1). We also detected interleukin 22 (IL-22) responses in IECs (e.g., Reg3γ) on the day of colonic colonization, which occurred concomitantly with a bloom of commensal Enterobacteriaceae on the mucosal surface. These results unravel a new paradigm in host-pathogen-microbiome interactions, showing for the first time that sensing a small number of pathogenic bacteria triggers swift intrinsic changes to the IEC composition and function, in tandem with significant changes to the mucosa-associated microbiome, which parallel innate immune responses.
Collapse
|
80
|
Valatas V, Kolios G, Bamias G. TL1A (TNFSF15) and DR3 (TNFRSF25): A Co-stimulatory System of Cytokines With Diverse Functions in Gut Mucosal Immunity. Front Immunol 2019; 10:583. [PMID: 30972074 PMCID: PMC6445966 DOI: 10.3389/fimmu.2019.00583] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 03/04/2019] [Indexed: 12/11/2022] Open
Abstract
TL1A and its functional receptor DR3 are members of the TNF/TNFR superfamilies of proteins. Binding of APC-derived TL1A to lymphocytic DR3 provides co-stimulatory signals for activated lymphocytes. DR3 signaling affects the proliferative activity of and cytokine production by effector lymphocytes, but also critically influences the development and suppressive function of regulatory T-cells. DR3 was also found to be highly expressed by innate lymphoid cells (ILCS), which respond to stimulation by TL1A. Several recent studies with transgenic and knockout mice as well as neutralizing or agonistic antibodies for these two proteins, have clearly shown that TL1A/DR3 are important mediators of several chronic immunological disorders, including Inflammatory Bowel Disease (IBD). TL1A and DR3 are abundantly localized at inflamed intestinal areas of patients with IBD and mice with experimental ileitis or colitis and actively participate in the immunological pathways that underlie mucosal homeostasis and intestinal inflammation. DR3 signaling has demonstrated a dichotomous role in mucosal immunity. On the one hand, during acute mucosal injury it exerts protective functions by ameliorating the severity of acute inflammatory responses and facilitating tissue repair. On the other hand, it critically participates in the pro-inflammatory pathways that underlie chronic inflammatory responses, such as those that take place in IBD. These effects are mediated through modulation of the relative mucosal abundance and function of Th1, Th2, Th17, Th9, and Treg lymphocytes, but also of all types of ILCs. Recently, an important role was demonstrated for TL1A/DR3 as potential mediators of intestinal fibrosis that is associated with the presence of gut inflammation. These accumulating data have raised the possibility that TL1A/DR3 pathways may represent a valid therapeutic target for chronic immunological diseases. Nevertheless, applicability of such a therapeutic approach will greatly rely on the net result of TL1A/DR3 manipulation on the various cell populations that will be affected by this approach.
Collapse
Affiliation(s)
- Vassilis Valatas
- Gastroenterology and Hepatology Research Laboratory, Medical School, University of Crete, Heraklion, Greece
| | - George Kolios
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Giorgos Bamias
- GI-unit, National & Kapodistrian University of Athens, Third Department of Internal Medicine, Sotiria Hospital, Athens, Greece
| |
Collapse
|
81
|
Interleukin-18 in Health and Disease. Int J Mol Sci 2019; 20:ijms20030649. [PMID: 30717382 PMCID: PMC6387150 DOI: 10.3390/ijms20030649] [Citation(s) in RCA: 322] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-18 was originally discovered as a factor that enhanced IFN-γ production from anti-CD3-stimulated Th1 cells, especially in the presence of IL-12. Upon stimulation with Ag plus IL-12, naïve T cells develop into IL-18 receptor (IL-18R) expressing Th1 cells, which increase IFN-γ production in response to IL-18 stimulation. Therefore, IL-12 is a commitment factor that induces the development of Th1 cells. In contrast, IL-18 is a proinflammatory cytokine that facilitates type 1 responses. However, IL-18 without IL-12 but with IL-2, stimulates NK cells, CD4+ NKT cells, and established Th1 cells, to produce IL-3, IL-9, and IL-13. Furthermore, together with IL-3, IL-18 stimulates mast cells and basophils to produce IL-4, IL-13, and chemical mediators such as histamine. Therefore, IL-18 is a cytokine that stimulates various cell types and has pleiotropic functions. IL-18 is a member of the IL-1 family of cytokines. IL-18 demonstrates a unique function by binding to a specific receptor expressed on various types of cells. In this review article, we will focus on the unique features of IL-18 in health and disease in experimental animals and humans.
Collapse
|
82
|
Abstract
Redox signalling in the gastrointestinal mucosa is held in an intricate balance. Potent microbicidal mechanisms can be used by infiltrating immune cells, such as neutrophils, to protect compromised mucosae from microbial infection through the generation of reactive oxygen species. Unchecked, collateral damage to the surrounding tissue from neutrophil-derived reactive oxygen species can be detrimental; thus, maintenance and restitution of a breached intestinal mucosal barrier are paramount to host survival. Redox reactions and redox signalling have been studied for decades with a primary focus on contributions to disease processes. Within the past decade, an upsurge of exciting findings have implicated subtoxic levels of oxidative stress in processes such as maintenance of mucosal homeostasis, the control of protective inflammation and even regulation of tissue wound healing. Resident gut microbial communities have been shown to trigger redox signalling within the mucosa, which expresses similar but distinct enzymes to phagocytes. At the fulcrum of this delicate balance is the colonic mucosal epithelium, and emerging evidence suggests that precise control of redox signalling by these barrier-forming cells may dictate the outcome of an inflammatory event. This Review will address both the spectrum and intensity of redox activity pertaining to host-immune and host-microbiota crosstalk during homeostasis and disease processes in the gastrointestinal tract.
Collapse
|
83
|
Couturier-Maillard A, Froux N, Piotet-Morin J, Michaudel C, Brault L, Le Bérichel J, Sénéchal A, Robinet P, Chenuet P, Jejou S, Dumoutier L, Renauld JC, Iovanna J, Huber S, Chamaillard M, Quesniaux V, Sokol H, Chamaillard M, Ryffel B. Interleukin-22-deficiency and microbiota contribute to the exacerbation of Toxoplasma gondii-induced intestinal inflammation. Mucosal Immunol 2018; 11:1181-1190. [PMID: 29728643 DOI: 10.1038/s41385-018-0005-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 01/04/2018] [Accepted: 01/09/2018] [Indexed: 02/04/2023]
Abstract
Upon oral infection with Toxoplasma gondii cysts (76 K strain) tachyzoites are released into the intestinal lumen and cross the epithelial barrier causing damage and acute intestinal inflammation in C57BL/6 (B6) mice. Here we investigated the role of microbiota and IL-22 in T.gondii-induced small intestinal inflammation. Oral T.gondii infection in B6 mice causes inflammation with IFNγ and IL-22 production. In IL-22-deficient mice, T.gondii infection augments the Th1 driven inflammation. Deficiency in either IL-22bp, the soluble IL-22 receptor or Reg3γ, an IL-22-dependent antimicrobial lectin/peptide, did not reduce inflammation. Under germ-free conditions, T.gondii-induced inflammation was reduced in correlation with parasite load. But intestinal inflammation is still present in germ-free mice, at low level, in the lamina propria, independently of IL-22 expression. Exacerbated intestinal inflammation driven by absence of IL-22 appears to be independent of IL-22 deficiency associated-dysbiosis as similar inflammation was observed after fecal transplantation of IL-22-/- or WT microbiota to germ-free-WT mice. Our results suggest cooperation between parasite and intestinal microbiota in small intestine inflammation development and endogenous IL-22 seems to exert a protective role independently of its effect on the microbiota. In conclusion, IL-22 participates in T.gondii induced acute small intestinal inflammation independently of microbiota and Reg3γ.
Collapse
Affiliation(s)
- A Couturier-Maillard
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - N Froux
- CNRS UPS44 -TAAM, Orléans, France
| | - J Piotet-Morin
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - C Michaudel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - L Brault
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - J Le Bérichel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | | | - P Robinet
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - P Chenuet
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - S Jejou
- Sorbonne Universités, UPMC Univ. Paris 06, École Normale Supérieure, PSL Research University, CNRS, INSERM, APHP, Laboratoire des Biomolécules (LBM), 27 rue de Chaligny, 75005, Paris, France
| | - L Dumoutier
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, Brussels, Belgium
| | - J C Renauld
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, Brussels, Belgium
| | - J Iovanna
- INSERM U1068, Centre de Recherche en Cancérologie de Marseille (CRCM), Aix-Marseille Université and Institut Paoli-Calmette, Parc Scientifique et Technologique de Luminy, CNRS UMR 7258, Marseille, France
| | - S Huber
- Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, 20246, Germany
| | | | - Vfj Quesniaux
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - H Sokol
- Sorbonne Universités, UPMC Univ. Paris 06, École Normale Supérieure, PSL Research University, CNRS, INSERM, APHP, Laboratoire des Biomolécules (LBM), 27 rue de Chaligny, 75005, Paris, France
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, 78352, France
- Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique-Hopitaux de Paris, UPMC, Paris, France
| | - M Chamaillard
- Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique-Hopitaux de Paris, UPMC, Paris, France
| | - B Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France.
| |
Collapse
|
84
|
Diminished circulating plasma and elevated lymph node culture supernatant levels of IL-10 family cytokines in tuberculous lymphadenitis. Cytokine 2018; 111:511-517. [PMID: 29871780 DOI: 10.1016/j.cyto.2018.05.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/23/2018] [Accepted: 05/29/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND IL-10 family cytokines are associated with the host immune response to pulmonary tuberculosis (PTB), but their association with host response in tuberculous lymphadenitis (TBL) is not known. METHODS Hence, we examined the circulating levels of the whole panel of IL-10 family cytokines in TBL (n = 44) and compared them to the levels in PTB (n = 44) and healthy control (HC, n = 44) individuals. We also assessed the pre and post-treatment cytokine levels in TBL individuals following the completion of anti-tuberculosis treatment (ATT). Next, we also compared the levels of IL-10 family cytokine in circulation versus lymph node (LN) culture supernatants in a subset of TBL individuals (n = 22). Finally, we also measured the levels of IL-10 family cytokines in tuberculosis antigen (purified protein derivative, PPD) stimulated and unstimulated LN culture supernatants. RESULTS TBL individuals exhibit significantly decreased levels of IL-10, IL-19, IL-20, IL-24, IL-28B and IL-29 in the circulation when compared to PTB (except IL-10) and HC (except IL-20 and IL-28B) and significantly increased levels of IL-22 when compared to PTB individuals. Following ATT, TBL individuals exhibit significantly elevated levels of IL-10, IL-19, IL-20, IL-24, IL-28B and IL-29 and significantly diminished levels of IL-26. Similarly, TBL individuals also exhibited significantly increased levels of IL-10, IL-19, IL-20, IL-24, IL-28A and IL-29 in LN culture supernatants compared to plasma and significantly decreased levels of IL-22. This was associated with enhanced levels of IL-19, IL-20, IL-24, IL-28B and IL-29 upon PPD stimulation of LN cultures. CONCLUSIONS Therefore, we demonstrate that TBL is associated with significantly diminished plasma and elevated LN culture supernatant levels of most of the IL-10 family cytokines. This to our knowledge is the first comprehensive examination of IL-10 family cytokines in TBL.
Collapse
|
85
|
Inflammasome components caspase-1 and adaptor protein apoptosis-associated speck-like proteins are important in resistance to Cryptosporidium parvum. Microbes Infect 2018; 20:369-375. [PMID: 29842985 DOI: 10.1016/j.micinf.2018.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/22/2018] [Accepted: 04/24/2018] [Indexed: 02/07/2023]
Abstract
Cryptosporidium spp. are opportunistic protozoan parasites that infect epithelial cells in the intestinal tract and cause a flu-like diarrheal illness. Innate immunity is key to limiting the expansion of parasitic stages early in infection. One mechanism in which it does this is through the generation of early cytokines, such as IL-18. The processing and secretion of mature IL-18 (and IL-1β) is mediated by caspase-1 which is activated within an inflammasome following the engagement of inflammasome-initiating sensors. We examined how the absence of caspase-1 and caspase-11, the adapter protein Asc, and other inflammasome components affects susceptibility to cryptosporidial infection by these and other key cytokines in the gut. We found that Casp-11-/-Casp-1-/- knockout mice have increased susceptibility to Cryptosporidium parvum infection as demonstrated by the 35-fold higher oocyst production (at peak infection) compared to wild-type mice. Susceptibility correlated with a lack of IL-18 in caspase-1 and caspase1/11 knockout mice, whereas IL-18 is significantly elevated in wildtype mice. IL-1β was not generated in any significant amount following infection nor was any increased susceptibility observed in IL-1β knockout mice. We also show that the adapter protein Asc is important to susceptibility, and that the caspase-1 canonical inflammasome signaling pathway is the dominant pathway in C. parvum resistance.
Collapse
|
86
|
Heimesaat MM, Escher U, Grunau A, Fiebiger U, Bereswill S. Peroral Low-Dose Toxoplasma gondii Infection of Human Microbiota-Associated Mice - A Subacute Ileitis Model to Unravel Pathogen-Host Interactions. Eur J Microbiol Immunol (Bp) 2018; 8:53-61. [PMID: 29997912 PMCID: PMC6038537 DOI: 10.1556/1886.2018.00005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 03/16/2018] [Indexed: 02/06/2023] Open
Abstract
Within 1 week following high-dose Toxoplasma gondii infection, mice develop lethal necrotizing ileitis. However, data from a subacute T. gondii-induced ileitis model are scarce. Therefore, mice harboring a human gut microbiota were perorally infected with one cyst of T. gondii. Within 9 days post-infection, the intestinal microbiota composition shifted towards higher loads of commensal enterobacteria and enterococci. Following T. gondii infection, mice were clinically only mildly affected, whereas ≈60% of mice displayed fecal blood and mild-to-moderate ileal histopathological changes. Intestinal inflammation was further characterized by increased apoptotic intestinal epithelial cells, which were accompanied by elevated proliferating gut epithelial cell numbers. As compared to naive controls, infected mice displayed elevated numbers of intestinal T lymphocytes and regulatory T-cells and increased pro-inflammatory mediator secretion. Remarkably, T. gondii-induced apoptotic and pro-inflammatory immune responses were not restricted to the gut, but could also be observed in extra-intestinal compartments including kidney, liver, and lung. Strikingly, low-dose T. gondii infection resulted in increased serum levels of pro- and anti-inflammatory cytokines. In conclusion, the here presented subacute ileitis model following peroral low-dose T. gondii infection of humanized mice allows for detailed investigations of the molecular mechanism underlying the “ménage à trois” of pathogens, human gut microbiota, and immunity.
Collapse
Affiliation(s)
- Markus M Heimesaat
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Microbiology and Infection Immunology, Berlin, Germany
| | - Ulrike Escher
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Microbiology and Infection Immunology, Berlin, Germany
| | - Anne Grunau
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Microbiology and Infection Immunology, Berlin, Germany
| | - Ulrike Fiebiger
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Microbiology and Infection Immunology, Berlin, Germany
| | - Stefan Bereswill
- Charité -Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Microbiology and Infection Immunology, Berlin, Germany
| |
Collapse
|
87
|
Sharma D, Malik A, Guy CS, Karki R, Vogel P, Kanneganti TD. Pyrin Inflammasome Regulates Tight Junction Integrity to Restrict Colitis and Tumorigenesis. Gastroenterology 2018; 154:948-964.e8. [PMID: 29203393 PMCID: PMC5847456 DOI: 10.1053/j.gastro.2017.11.276] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 11/22/2017] [Accepted: 11/28/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Inflammatory bowel diseases (IBD) increase risk for colorectal cancer. Mutations in the Mediterranean fever gene (MEFV or pyrin) are associated with hereditary autoinflammatory disease and severe IBD. Expression of MEFV, a sensor protein that the initiates assembly of the inflammasome complex, is increased in colon biopsies from patients with IBD. We investigated the role of pyrin in intestinal homeostasis in mice. METHODS Mefv-/- mice and C57/BL6 mice (controls) were given azoxymethane followed by multiple rounds of dextran sodium sulfate (DSS) to induce colitis and tumorigenesis. In some experiments, Mefv-/- mice were given injections of recombinant interleukin 18 (rIL18) or saline (control) during DSS administration. Colon tissues were collected at different time points during colitis development and analyzed by histology, immunohistochemistry, immunoblots, or ELISAs (to measure cytokines). Spleen and mesenteric lymph node were collected, processed, and analyzed by flow cytometry. Colon epithelial permeability was measured in mice with colitis by gavage of fluorescent dextran and quantification of serum levels. RESULTS MEFV was expressed in colons of control mice and expression increased during chronic and acute inflammation; high levels were detected in colon tumor and adjacent non-tumor tissues. Mefv-/- mice developed more severe colitis than control mice, with a greater extent of epithelial hyperplasia and a larger tumor burden. Levels of inflammatory cytokines (IL6) and chemokines were significantly higher in colons of Mefv-/- mice than control mice following colitis induction, whereas the level IL18, which depends on the inflammasome for maturation and release, was significantly lower in colons of Mefv-/- mice. Mefv-/- mice had increased epithelial permeability following administration of DSS than control mice, and loss of the tight junction proteins occludin and claudin-2 from intercellular junctions. STAT3 was activated (phosphorylated) in inflamed colon tissues from Mefv-/-, which also had increased expression of stem cell markers (OLFM4, BMI1, and MSI1) compared with colons from control mice. Administration of rIL18 to Mefv-/- mice reduced epithelial permeability, intestinal inflammation, the severity of colitis, and colon tumorigenesis. CONCLUSIONS In studies with DSS-induced colitis, we found that pyrin (MEFV) is required for inflammasome activation and IL18 maturation, which promote intestinal barrier integrity and prevent colon inflammation and tumorigenesis. Strategies to increase activity of MEFV or IL18 might be developed for the treatment of IBD and prevention of colitis-associated tumorigenesis.
Collapse
Affiliation(s)
- Deepika Sharma
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ankit Malik
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Clifford S Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Peter Vogel
- Animal Resources Center and the Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | |
Collapse
|
88
|
Rathinam VAK, Chan FKM. Inflammasome, Inflammation, and Tissue Homeostasis. Trends Mol Med 2018; 24:304-318. [PMID: 29433944 DOI: 10.1016/j.molmed.2018.01.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/06/2018] [Accepted: 01/14/2018] [Indexed: 02/07/2023]
Abstract
Organismal fitness demands proper response to neutralize the threat from infection or injury. At the mammalian intestinal epithelium barrier, the inflammasome coordinates an elaborate tissue repair response marked by the induction of antimicrobial peptides, wound-healing cytokines, and reparative proliferation of epithelial stem cells. The inflammasome in myeloid and intestinal epithelial compartments exerts these effects in part through maintenance of a healthy microbiota. Disease-associated mutations and elevated expression of certain inflammasome sensors have been identified. In many cases, inhibition of inflammasome activity has dramatic effects on disease outcome in mouse models of experimental colitis. Here, we discuss recent studies on the role of distinct inflammasome sensors in intestinal homeostasis and how this knowledge may be translated into a therapeutic setting.
Collapse
Affiliation(s)
- Vijay A K Rathinam
- Department of Immunology, UConn Health School of Medicine, Farmington, CT 06030, USA.
| | - Francis Ka-Ming Chan
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
89
|
Dunay IR, Diefenbach A. Group 1 innate lymphoid cells in Toxoplasma gondii infection. Parasite Immunol 2018; 40. [PMID: 29315653 DOI: 10.1111/pim.12516] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/04/2018] [Indexed: 12/31/2022]
Abstract
Innate lymphoid cells (ILCs) are a group of lymphocytes that carry out important functions in immunity to infections and in organ homeostasis at epithelial barrier surfaces. ILCs are innate immune cells that provide an early source of cytokines to initiate immune responses against pathogens. Cytotoxic ILCs (i.e. conventional (c)NK cells) and several subsets of helper-like ILCs are the major branches of the ILC family. Conventional NK cells and group 1 ILCs share several characteristics such as surface receptors and the ability to produce IFN-γ upon activation, but they differ in their developmental paths and in their dependence on specific transcription factors. Infection of mice with the intracellular parasite Toxoplasma gondii is followed by a strong Th1-mediated immune response. Previous studies indicate that NK1.1+ cells contribute to the production of IFN-γ and TNF and cytotoxicity during acute T. gondii infection. Upon oral infection, the parasite infects intestinal enterocytes, and within the lamina propria, innate immune responses lead to initial parasite control although the infection disseminates widely and persists long-term in immune privileged sites despite adaptive immunity. Upon parasite entry into the small intestine, during the acute stage, ILC1 produce high levels of IFN-γ and TNF protecting barrier surfaces, thus essentially contributing to early parasite control. We will discuss here the role of innate lymphocytes during T. gondii infection in the context of the only recently appreciated diversity of ILC subsets.
Collapse
Affiliation(s)
- I R Dunay
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - A Diefenbach
- Department of Microbiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
90
|
Liu X, Lu J, Liu Z, Zhao J, Sun H, Wu N, Liu H, Liu W, Hu Z, Meng G, Shen L, Miller AW, Su B, Li X, Kang Z. Intestinal Epithelial Cell-Derived LKB1 Suppresses Colitogenic Microbiota. THE JOURNAL OF IMMUNOLOGY 2018; 200:1889-1900. [PMID: 29352002 DOI: 10.4049/jimmunol.1700547] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 12/20/2017] [Indexed: 12/14/2022]
Abstract
Dysregulation of the immune barrier function of the intestinal epithelium can often result in dysbiosis. In this study we report a novel role of intestinal epithelial cell (IEC)-derived liver kinase B1 (LKB1) in suppressing colitogenic microbiota. IEC-specific deletion of LKB1 (LKB1ΔIEC) resulted in an increased susceptibility to dextran sodium sulfate (DSS)-induced colitis and a definitive shift in the composition of the microbial population in the mouse intestine. Importantly, transfer of the microbiota from LKB1ΔIEC mice was sufficient to confer increased susceptibility to DSS-induced colitis in wild-type recipient mice. Collectively, the data indicate that LKB1 deficiency in intestinal epithelial cells nurtures the outgrowth of colitogenic bacteria in the commensal community. In addition, LKB1 deficiency in the intestinal epithelium reduced the production of IL-18 and antimicrobial peptides in the colon. Administration of exogenous IL-18 restored the expression of antimicrobial peptides, corrected the outgrowth of several bacterial genera, and rescued the LKB1ΔIEC mice from increased sensitivity to DSS challenge. Taken together, our study reveals an important function of LKB1 in IECs for suppressing colitogenic microbiota by IL-18 expression.
Collapse
Affiliation(s)
- Xia'nan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jinfeng Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhenshan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junjie Zhao
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195
| | - Hongxiang Sun
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ningbo Wu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hongzhi Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weiwei Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhuqin Hu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guangxun Meng
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lei Shen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Aaron W Miller
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195.,Department of Urology, Cleveland Clinic, Cleveland, OH 44195; and
| | - Bing Su
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoxia Li
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| | - Zizhen Kang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; .,Department of Immunology, Cleveland Clinic, Cleveland, OH 44195.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| |
Collapse
|
91
|
Consequences of Epithelial Inflammasome Activation by Bacterial Pathogens. J Mol Biol 2018; 430:193-206. [DOI: 10.1016/j.jmb.2017.03.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/17/2017] [Accepted: 03/19/2017] [Indexed: 01/02/2023]
|
92
|
Geremia A, Arancibia-Cárcamo CV. Innate Lymphoid Cells in Intestinal Inflammation. Front Immunol 2017; 8:1296. [PMID: 29081776 PMCID: PMC5645495 DOI: 10.3389/fimmu.2017.01296] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/27/2017] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the intestine that encompasses Crohn's disease (CD) and ulcerative colitis. The cause of IBD is unknown, but the evidence suggests that an aberrant immune response toward the commensal bacterial flora is responsible for disease in genetically susceptible individuals. Results from animal models of colitis and human studies indicate a role for innate lymphoid cells (ILC) in the pathogenesis of chronic intestinal inflammation in IBD. ILC are a population of lymphocytes that are enriched at mucosal sites, where they play a protective role against pathogens including extracellular bacteria, helminthes, and viruses. ILC lack an antigen-specific receptor, but can respond to environmental stress signals contributing to the rapid orchestration of an early immune response. Several subsets of ILC reflecting functional characteristics of T helper subsets have been described. ILC1 express the transcription factor T-bet and are characterized by secretion of IFNγ, ILC2 are GATA3+ and secrete IL5 and IL13 and ILC3 depend on expression of RORγt and secrete IL17 and IL22. However, ILC retain a degree of plasticity depending on exposure to cytokines and environmental factors. IL23 responsive ILC have been implicated in the pathogenesis of colitis in several innate murine models through the production of IL17, IFNγ, and GM-CSF. We have previously identified IL23 responsive ILC in the human intestine and found that they accumulate in the inflamed colon and small bowel of patients with CD. Other studies have confirmed accumulation of ILC in CD with increased frequencies of IFNγ-secreting ILC1 in both the intestinal lamina propria and the epithelium. Moreover, IL23 driven IL22 producing ILC have been shown to drive bacteria-induced colitis-associated cancer in mice. Interestingly, our data show increased ILC accumulation in patients with IBD and primary sclerosing cholangitis, who carry an increased risk of developing colorectal cancer. ILC may play an important amplifying role in IBD and IBD-associated cancer, through secretion of inflammatory cytokines and interaction with other immune and non-immune cells. Here, we will review the evidence indicating a role for ILC in the pathogenesis of chronic intestinal inflammation.
Collapse
Affiliation(s)
- Alessandra Geremia
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom
| | - Carolina V Arancibia-Cárcamo
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
93
|
Victor AR, Nalin AP, Dong W, McClory S, Wei M, Mao C, Kladney RD, Youssef Y, Chan WK, Briercheck EL, Hughes T, Scoville SD, Pitarresi JR, Chen C, Manz S, Wu LC, Zhang J, Ostrowski MC, Freud AG, Leone GW, Caligiuri MA, Yu J. IL-18 Drives ILC3 Proliferation and Promotes IL-22 Production via NF-κB. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:2333-2342. [PMID: 28842466 PMCID: PMC5624342 DOI: 10.4049/jimmunol.1601554] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 07/27/2017] [Indexed: 12/13/2022]
Abstract
Group 3 innate lymphoid cells (ILC3s) are important regulators of the immune system, maintaining homeostasis in the presence of commensal bacteria, but activating immune defenses in response to microbial pathogens. ILC3s are a robust source of IL-22, a cytokine critical for stimulating the antimicrobial response. We sought to identify cytokines that can promote proliferation and induce or maintain IL-22 production by ILC3s and determine a molecular mechanism for this process. We identified IL-18 as a cytokine that cooperates with an ILC3 survival factor, IL-15, to induce proliferation of human ILC3s, as well as induce and maintain IL-22 production. To determine a mechanism of action, we examined the NF-κB pathway, which is activated by IL-18 signaling. We found that the NF-κB complex signaling component, p65, binds to the proximal region of the IL22 promoter and promotes transcriptional activity. Finally, we observed that CD11c+ dendritic cells expressing IL-18 are found in close proximity to ILC3s in human tonsils in situ. Therefore, we identify a new mechanism by which human ILC3s proliferate and produce IL-22, and identify NF-κB as a potential therapeutic target to be considered in pathologic states characterized by overproduction of IL-18 and/or IL-22.
Collapse
Affiliation(s)
- Aaron R Victor
- Medical Scientist Training Program, Ohio State University, Columbus, OH 43210
| | - Ansel P Nalin
- Medical Scientist Training Program, Ohio State University, Columbus, OH 43210
| | - Wenjuan Dong
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Susan McClory
- Medical Scientist Training Program, Ohio State University, Columbus, OH 43210
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Min Wei
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Charlene Mao
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Raleigh D Kladney
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, OH 43210
| | - Youssef Youssef
- Department of Pathology, The Ohio State University, Columbus, OH 43210
| | - Wing Keung Chan
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Edward L Briercheck
- Medical Scientist Training Program, Ohio State University, Columbus, OH 43210
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Tiffany Hughes
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Steven D Scoville
- Medical Scientist Training Program, Ohio State University, Columbus, OH 43210
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Jason R Pitarresi
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Charlie Chen
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Sarah Manz
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Lai-Chu Wu
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Jianying Zhang
- Center for Biostatistics, Department of Bioinformatics, The Ohio State University, Columbus, OH 43210; and
| | - Michael C Ostrowski
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Aharon G Freud
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
- Department of Pathology, The Ohio State University, Columbus, OH 43210
| | - Gustavo W Leone
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, OH 43210
| | - Michael A Caligiuri
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210;
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Jianhua Yu
- The James Cancer Hospital and Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210;
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
94
|
Group III phospholipase A 2 promotes colitis and colorectal cancer. Sci Rep 2017; 7:12261. [PMID: 28947740 PMCID: PMC5612992 DOI: 10.1038/s41598-017-12434-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/11/2017] [Indexed: 12/21/2022] Open
Abstract
Lipid mediators play pivotal roles in colorectal cancer and colitis, but only a limited member of the phospholipase A2 (PLA2) subtypes, which lie upstream of various lipid mediators, have been implicated in the positive or negative regulation of these diseases. Clinical and biochemical evidence suggests that secreted PLA2 group III (sPLA2-III) is associated with colorectal cancer, although its precise role remains obscure. Here we have found that sPLA2-III-null (Pla2g3 -/-) mice are highly resistant to colon carcinogenesis. Furthermore, Pla2g3 -/- mice are less susceptible to dextran sulfate-induced colitis, implying that the amelioration of colonic inflammation by sPLA2-III ablation may underlie the protective effect against colon cancer. Lipidomics analysis of the colon revealed significant reduction of pro-inflammatory/pro-tumorigenic lysophosholipids as well as unusual steady-state elevation of colon-protective fatty acids and their oxygenated metabolites in Pla2g3 -/- mice. Overall, our results establish a role of sPLA2-III in the promotion of colorectal inflammation and cancer, expand our understanding of the divergent roles of multiple PLA2 enzymes in the gastrointestinal tract, and point to sPLA2-III as a novel druggable target for colorectal diseases.
Collapse
|
95
|
Lei-Leston AC, Murphy AG, Maloy KJ. Epithelial Cell Inflammasomes in Intestinal Immunity and Inflammation. Front Immunol 2017; 8:1168. [PMID: 28979266 PMCID: PMC5611393 DOI: 10.3389/fimmu.2017.01168] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/04/2017] [Indexed: 12/12/2022] Open
Abstract
Pattern recognition receptors (PRR), such as NOD-like receptors (NLRs), sense conserved microbial signatures, and host danger signals leading to the coordination of appropriate immune responses. Upon activation, a subset of NLR initiate the assembly of a multimeric protein complex known as the inflammasome, which processes pro-inflammatory cytokines and mediates a specialized form of cell death known as pyroptosis. The identification of inflammasome-associated genes as inflammatory bowel disease susceptibility genes implicates a role for the inflammasome in intestinal inflammation. Despite the fact that the functional importance of inflammasomes within immune cells has been well established, the contribution of inflammasome expression in non-hematopoietic cells remains comparatively understudied. Given that intestinal epithelial cells (IEC) act as a barrier between the host and the intestinal microbiota, inflammasome expression by these cells is likely important for intestinal immune homeostasis. Accumulating evidence suggests that the inflammasome plays a key role in shaping epithelial responses at the host-lumen interface with many inflammasome components highly expressed by IEC. Recent studies have exposed functional roles of IEC inflammasomes in mucosal immune defense, inflammation, and tumorigenesis. In this review, we present the main features of the predominant inflammasomes and their effector mechanisms contributing to intestinal homeostasis and inflammation. We also discuss existing controversies in the field and open questions related to their implications in disease. A comprehensive understanding of the molecular basis of intestinal inflammasome signaling could hold therapeutic potential for clinical translation.
Collapse
Affiliation(s)
- Andrea C Lei-Leston
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Alison G Murphy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Kevin J Maloy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
96
|
Grunau A, Escher U, Bereswill S, Heimesaat MM. Toll-Like Receptor-4 Dependent Inflammatory Responses Following Intestinal Colonization of Secondary Abiotic IL10-Deficient Mice with Multidrug-Resistant Pseudomonas Aeruginosa. Eur J Microbiol Immunol (Bp) 2017; 7:210-219. [PMID: 29034110 PMCID: PMC5632748 DOI: 10.1556/1886.2017.00023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 08/23/2017] [Indexed: 01/05/2023] Open
Abstract
The rising incidences of infections with multidrug-resistant (MDR) Gram-negative bacteria including Pseudomonas aeruginosa (PA) have gained increasing attention in medicine, but also in the general public and global health politics. The mechanisms underlying opportunistic pathogen–host interactions are unclear, however. To address this, we challenged secondary abiotic IL10–/– mice deficient for Toll-like receptor-4 (TLR4–/– × IL10–/–), the main receptor of the Gram-negative cell wall constituent lipopolysaccharide, with a clinical MDR PA isolate. Despite higher intestinal colonization densities, apoptotic colonic epithelial cell numbers were lower in TLR4–/– × IL10–/– mice as compared to IL10–/– controls at day 14 postinfection (p.i.), whereas proliferating/regenerating cells had increased in the latter only. Furthermore, PA-colonized TLR4–/– × IL10–/– mice displayed less distinct innate and adaptive immune cell responses in the colon as compared to IL10–/– counterparts that were accompanied by lower nitric oxide concentrations in mesenteric lymph nodes in the former at day 14 p.i. Conversely, splenic NO levels were higher in both naive and PA-colonized TLR4-deficient IL10–/– mice versus IL10–/– controls. Remarkably, intestinal MDR PA was able to translocate to extra-intestinal including systemic compartments of TLR4–/– × IL10–/– mice only. Hence, MDR PA-induced intestinal and systemic immune responses observed in secondary abiotic IL10–/– mice are TLR4-dependent.
Collapse
Affiliation(s)
- Anne Grunau
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Microbiology and Hygiene, Berlin, Germany
| | - Ulrike Escher
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Microbiology and Hygiene, Berlin, Germany
| | - Stefan Bereswill
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Microbiology and Hygiene, Berlin, Germany
| | - Markus M Heimesaat
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Microbiology and Hygiene, Berlin, Germany
| |
Collapse
|
97
|
Kern M, Aschenbach JR, Tedin K, Pieper R, Loss H, Lodemann U. Characterization of Inflammasome Components in Pig Intestine and Analysis of the Influence of Probiotic Enterococcus Faecium during an Escherichia Coli Challenge. Immunol Invest 2017; 46:742-757. [DOI: 10.1080/08820139.2017.1360341] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Martina Kern
- Department of Veterinary Medicine, Institute of Veterinary Physiology, Freie Universitaet Berlin, Berlin, Germany
| | - Jörg R Aschenbach
- Department of Veterinary Medicine, Institute of Veterinary Physiology, Freie Universitaet Berlin, Berlin, Germany
| | - Karsten Tedin
- Department of Veterinary Medicine, Institute of Microbiology and Epizootics, Freie Universitaet Berlin, Berlin, Germany
| | - Robert Pieper
- Department of Veterinary Medicine, Institute of Animal Nutrition, Freie Universitaet Berlin, Berlin, Germany
| | - Henriette Loss
- Department of Veterinary Medicine, Institute of Veterinary Physiology, Freie Universitaet Berlin, Berlin, Germany
| | - Ulrike Lodemann
- Department of Veterinary Medicine, Institute of Veterinary Physiology, Freie Universitaet Berlin, Berlin, Germany
| |
Collapse
|
98
|
Savage AK, Liang HE, Locksley RM. The Development of Steady-State Activation Hubs between Adult LTi ILC3s and Primed Macrophages in Small Intestine. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:1912-1922. [PMID: 28747343 PMCID: PMC5568484 DOI: 10.4049/jimmunol.1700155] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/03/2017] [Indexed: 12/21/2022]
Abstract
Group 3 innate lymphoid cells (ILC3s) are important for intestinal health, particularly in controlling inflammation in response to epithelial dysregulation, but their role during homeostasis remains less well understood. We generated IL-22 reporter mice to assess production of this key cytokine by ILC3s in the small intestine during development and under basal conditions. Although IL-22 is produced by a variety of lymphocyte populations, constitutively high IL-22 expression was limited to lymphoid-tissue inducer (LTi) cells residing in lymph node-like structures in the gut called solitary intestinal lymphoid tissues (SILT). Constitutive IL-22 expression was dependent on the microbiota and MyD88 signaling, appeared upon weaning, and was present across the spectrum of SILT, including in cryptopatches. Activated SILT LTi cells colocalized with a rare subpopulation of activated macrophages constitutively positive for IL-12/23 p40 and capable of activating neonatal LTi cells in response to TLR stimulus. Thus, weaning leads to the organization of innate immune activation hubs at SILT that mature and are continuously sustained by signals from the microbiota. This functional and anatomic organization constitutes a significant portion of the steady-state IL-23/IL-22 axis.
Collapse
Affiliation(s)
- Adam K Savage
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158
| | - Hong-Erh Liang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143; and
| | - Richard M Locksley
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158;
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143; and
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94115
| |
Collapse
|
99
|
Dolinay T, Himes BE, Shumyatcher M, Lawrence GG, Margulies SS. Integrated Stress Response Mediates Epithelial Injury in Mechanical Ventilation. Am J Respir Cell Mol Biol 2017; 57:193-203. [PMID: 28363030 DOI: 10.1165/rcmb.2016-0404oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Ventilator-induced lung injury (VILI) is a severe complication of mechanical ventilation that can lead to acute respiratory distress syndrome. VILI is characterized by damage to the epithelial barrier with subsequent pulmonary edema and profound hypoxia. Available lung-protective ventilator strategies offer only a modest benefit in preventing VILI because they cannot impede alveolar overdistension and concomitant epithelial barrier dysfunction in the inflamed lung regions. There are currently no effective biochemical therapies to mitigate injury to the alveolar epithelium. We hypothesize that alveolar stretch activates the integrated stress response (ISR) pathway and that the chemical inhibition of this pathway mitigates alveolar barrier disruption during stretch and mechanical ventilation. Using our established rat primary type I-like alveolar epithelial cell monolayer stretch model and in vivo rat mechanical ventilation that mimics the alveolar overdistension seen in acute respiratory distress syndrome, we studied epithelial responses to mechanical stress. Our studies revealed that the ISR signaling pathway is a key modulator of epithelial permeability. We show that prolonged epithelial stretch and injurious mechanical ventilation activate the ISR, leading to increased alveolar permeability, cell death, and proinflammatory signaling. Chemical inhibition of protein kinase RNA-like endoplasmic reticulum kinase, an upstream regulator of the pathway, resulted in decreased injury signaling and improved barrier function after prolonged cyclic stretch and injurious mechanical ventilation. Our results provide new evidence that therapeutic targeting of the ISR can mitigate VILI.
Collapse
Affiliation(s)
- Tamas Dolinay
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Blanca E Himes
- 2 Department of Biostatistics, Epidemiology and Informatics, and
| | - Maya Shumyatcher
- 2 Department of Biostatistics, Epidemiology and Informatics, and
| | - Gladys Gray Lawrence
- 3 Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan S Margulies
- 3 Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
100
|
Heimesaat MM, Grundmann U, Alutis ME, Fischer A, Bereswill S. Absence of Nucleotide-Oligomerization-Domain-2 Is Associated with Less Distinct Disease in Campylobacter jejuni Infected Secondary Abiotic IL-10 Deficient Mice. Front Cell Infect Microbiol 2017; 7:322. [PMID: 28752081 PMCID: PMC5508002 DOI: 10.3389/fcimb.2017.00322] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 06/29/2017] [Indexed: 01/07/2023] Open
Abstract
Human Campylobacter jejuni-infections are progressively increasing worldwide. Despite their high prevalence and socioeconomic impact the underlying mechanisms of pathogen-host-interactions are only incompletely understood. Given that the innate immune receptor nucleotide-oligomerization-domain-2 (Nod2) is involved in clearance of enteropathogens, we here evaluated its role in murine campylobacteriosis. To address this, we applied Nod2-deficient IL-10-/- (Nod2-/- IL-10-/-) mice and IL-10-/- counterparts both with a depleted intestinal microbiota to warrant pathogen-induced enterocolitis. At day 7 following peroral C. jejuni strain 81-176 infection, Nod2 mRNA was down-regulated in the colon of secondary abiotic IL-10-/- and wildtype mice. Nod2-deficiency did neither affect gastrointestinal colonization nor extra-intestinal and systemic translocation properties of C. jejuni. Colonic mucin-2 mRNA was, however, down-regulated upon C. jejuni-infection of both Nod2-/- IL-10-/- and IL-10-/- mice, whereas expression levels were lower in infected, but also naive Nod2-/- IL-10-/- mice as compared to respective IL-10-/- controls. Remarkably, C. jejuni-infected Nod2-/- IL-10-/- mice were less compromised than IL-10-/- counterparts and displayed less distinct apoptotic, but higher regenerative cell responses in colonic epithelia. Conversely, innate as well as adaptive immune cells such as macrophages and monocytes as well as T lymphocytes and regulatory T-cells, respectively, were even more abundant in large intestines of Nod2-/- IL-10-/- as compared to IL-10-/- mice at day 7 post-infection. Furthermore, IFN-γ concentrations were higher in ex vivo biopsies derived from intestinal compartments including colon and mesenteric lymph nodes as well as in systemic tissue sites such as the spleen of C. jejuni infected Nod2-/- IL-10-/- as compared to IL10-/- counterparts. Whereas, at day 7 postinfection anti-inflammatory IL-22 mRNA levels were up-regulated, IL-18 mRNA was down-regulated in large intestines of Nod2-/- IL-10-/- vs. IL-10-/- mice. In summary, C. jejuni-infection induced less clinical signs and apoptosis, but more distinct colonic pro- and (of note) anti-inflammatory immune as well as regenerative cell responses in Nod2 deficient IL-10-/- as compared to IL-10-/- control mice. We conclude that, even though colonic Nod2 mRNA was down-regulated upon pathogenic challenge, Nod2-signaling is essentially involved in the well-balanced innate and adaptive immune responses upon C. jejuni-infection of secondary abiotic IL-10-/- mice, but does neither impact pathogenic colonization nor translocation.
Collapse
Affiliation(s)
- Markus M. Heimesaat
- Department of Microbiology and Hygiene, Charité—University Medicine BerlinBerlin, Germany
| | | | | | | | | |
Collapse
|