1
|
Mandal A, Banerjee S, Ghosh S, Biswas S, Bagchi A, Sil PC. α-ketoglutarate ameliorates colitis through modulation of inflammation, ER stress, and apoptosis. Toxicol Rep 2025; 14:101897. [PMID: 39886045 PMCID: PMC11780160 DOI: 10.1016/j.toxrep.2025.101897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 02/01/2025] Open
Abstract
Colitis is an inflammatory disorder of the gastrointestinal tract. A widely consumed dietary nutrient, α-ketoglutarate (α-KG) is known to play a crucial role in cellular metabolism and provide protection to intestinal epithelium under various pathophysiological conditions. In this study, 2,4,6-trinitrobenzenesulfonic acid (TNBS) was used to induce colitis in Wistar rats. After 36 hours of TNBS administration, the rats were orally treated with a solution of α-KG at 1 g/kg body weight for 5 days. Development of colitis was confirmed by observable physical symptoms of repeated loose blood-mixed stool, apathy for food and weight loss. Macroscopic inspection revealed an inflamed colonic surface with ulcerations. Histopathological observations included alterations in crypts-structure and disruption in both epithelial and mucosal layers of colon in colitis induced rats. Colitis resulted in elevated levels of pro-inflammatory cytokines, ER stress-mediated cell death and intrinsic apoptosis pathway. The ameliorative effects of α-KG against TNBS-mediated toxicity were confirmed through molecular technics and docking analysis. Additionally, there were no instances of toxicity of α-KG. Therefore, α-KG can be considered as a valuable therapeutic agent for further comprehensive research.
Collapse
Affiliation(s)
- Ankita Mandal
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| | - Sharmistha Banerjee
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| | - Sumit Ghosh
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| | - Sima Biswas
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, Kalyani, West Bengal 741235, India
| | - Angshuman Bagchi
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, Kalyani, West Bengal 741235, India
| | - Parames C. Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| |
Collapse
|
2
|
Li W, Liu T, Chen Y, Sun Y, Li C, Dong Y. Regulation and therapeutic potential of NLRP3 inflammasome in intestinal diseases. J Leukoc Biol 2025; 117:qiaf014. [PMID: 40276926 DOI: 10.1093/jleuko/qiaf014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Indexed: 04/26/2025] Open
Abstract
The NOD-like receptor family, particularly the protein 3 that contains the pyrin domain (NLRP3), is an intracellular sensing protein complex responsible for detecting patterns associated with pathogens and injuries. NLRP3 plays a crucial role in the innate immune response. Currently, a wide range of research has indicated the crucial importance of NLRP3 in various inflammatory conditions. Similarly, the NLRP3 inflammasome plays a significant role in preserving intestinal balance and impacting the advancement of diseases. In addition, several randomized trials have demonstrated the safety and efficacy of targeting NLRP3 in the treatment of colitis, colorectal cancer, and related diseases. This review explores the mechanisms of NLRP3 assembly and activation in the gut. We describe its pathological significance in intestinal diseases. Finally, we summarize current and future therapeutic approaches targeting NLRP3 for intestinal diseases.
Collapse
Affiliation(s)
- Wenxue Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Tianya Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Yaoxing Chen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| | - Yan Sun
- Department of Horticulture and Landscape Architecture, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China
| | - Chengzhong Li
- Department of Horticulture and Landscape Architecture, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China
| | - Yulan Dong
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, Beijing 100193, China
| |
Collapse
|
3
|
Prasad S, Singh S, Menge S, Mohapatra I, Kim S, Helland L, Singh G, Singh A. Gut redox and microbiome: charting the roadmap to T-cell regulation. Front Immunol 2024; 15:1387903. [PMID: 39234241 PMCID: PMC11371728 DOI: 10.3389/fimmu.2024.1387903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
The gastrointestinal (GI) tract redox environment, influenced by commensal microbiota and bacterial-derived metabolites, is crucial in shaping T-cell responses. Specifically, metabolites from gut microbiota (GM) exhibit robust anti-inflammatory effects, fostering the differentiation and regulation of CD8+ tissue-resident memory (TRM) cells, mucosal-associated invariant T (MAIT) cells, and stabilizing gut-resident Treg cells. Nitric oxide (NO), a pivotal redox mediator, emerges as a central regulator of T-cell functions and gut inflammation. NO impacts the composition of the gut microbiome, driving the differentiation of pro-inflammatory Th17 cells and exacerbating intestinal inflammation, and supports Treg expansion, showcasing its dual role in immune homeostasis. This review delves into the complex interplay between GI redox balance and GM metabolites, elucidating their profound impact on T-cell regulation. Additionally, it comprehensively emphasizes the critical role of GI redox, particularly reactive oxygen species (ROS) and NO, in shaping T-cell phenotype and functions. These insights offer valuable perspectives on disease mechanisms and potential therapeutic strategies for conditions associated with oxidative stress. Understanding the complex cross-talk between GI redox, GM metabolites, and T-cell responses provides valuable insights into potential therapeutic avenues for immune-mediated diseases, underscoring the significance of maintaining GI redox balance for optimal immune health.
Collapse
Affiliation(s)
- Sujata Prasad
- Translational Division, MLM Labs, LLC, Oakdale, MN, United States
| | - Shilpi Singh
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Samuel Menge
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
| | - Iteeshree Mohapatra
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, United States
| | - Stefan Kim
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Logan Helland
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Gatikrushna Singh
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Amar Singh
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
4
|
Yamada S, Honzawa Y, Yamamoto S, Matsuura M, Kitamoto H, Okabe M, Kakiuchi N, Toyonaga T, Kobayashi T, Hibi T, Seno H, Nakase H. Single Nucleotide Polymorphisms of the MEFV Gene E148Q Are Highly Associated With Disease Phenotype in Crohn's Disease. Inflamm Bowel Dis 2024; 30:970-980. [PMID: 37951297 DOI: 10.1093/ibd/izad259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Indexed: 11/13/2023]
Abstract
BACKGROUND Single nucleotide polymorphisms (SNPs) of the MEFV gene may modify inflammatory bowel disease (IBD) activity. The prevalence of MEFV gene SNPs in IBD patients and their involvement in IBD pathophysiology remains unclear. METHODS We analyzed 12 MEFV gene SNPs in peripheral leukocytes of Japanese IBD patients (Crohn's disease [CD]: 69 patients, ulcerative colitis: 32 patients) by polymerase chain reaction using next-generation DNA sequencing and evaluated their prevalence and association with the disease characteristics. Inflammasome activity and mature interleukin (IL)-1β and IL-18 production were evaluated in peripheral blood mononuclear cells obtained from CD patients stimulated with lipopolysaccharides and adenosine triphosphate, and compared between those with and without the E148Q SNP. COL1A1 and HSP47 gene expression was analyzed in CCD-18Co cells costimulated with IL-1β and other inflammatory cytokines. RESULTS The prevalence of MEFV gene SNPs in IBD patients was similar to that in the human gene database. E148Q was the most common SNP. Compared with CD patients without E148Q, those with E148Q had a significantly greater frequency of the stricture phenotype, and their peripheral blood mononuclear cells exhibited significantly higher IL-1β and IL-18 levels and higher caspase-1 activity. IL-1β and IL-17A synergistically increased COL1A1 and HSP47 gene expression. CONCLUSIONS MEFV gene SNPs, including E148Q, modify the behavior of CD. IL-1β and IL-18 are produced through enhanced caspase-1 activity in monocytes of CD patients with E148Q. IL-1β promotes gene expression of fibrosis-related genes by cooperating with IL-17A in myofibroblasts. Therefore, E148Q might be a disease-modifying gene associated with the fibrostenosis phenotype in CD patients.
Collapse
Affiliation(s)
- Satoshi Yamada
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yusuke Honzawa
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Shuji Yamamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Minoru Matsuura
- Third Department of Internal Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Hiroki Kitamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makoto Okabe
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nobuyuki Kakiuchi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiko Toyonaga
- Center for Advanced IBD Research and Treatment, Kitasato University Hospital, Kitasato University, Tokyo, Japan
| | - Taku Kobayashi
- Center for Advanced IBD Research and Treatment, Kitasato University Hospital, Kitasato University, Tokyo, Japan
| | - Toshifumi Hibi
- Center for Advanced IBD Research and Treatment, Kitasato University Hospital, Kitasato University, Tokyo, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
5
|
Arrè V, Scialpi R, Centonze M, Giannelli G, Scavo MP, Negro R. The 'speck'-tacular oversight of the NLRP3-pyroptosis pathway on gastrointestinal inflammatory diseases and tumorigenesis. J Biomed Sci 2023; 30:90. [PMID: 37891577 PMCID: PMC10612184 DOI: 10.1186/s12929-023-00983-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023] Open
Abstract
The NLRP3 inflammasome is an intracellular sensor and an essential component of the innate immune system involved in danger recognition. An important hallmark of inflammasome activation is the formation of a single supramolecular punctum, known as a speck, per cell, which is the site where the pro-inflammatory cytokines IL-1β and IL-18 are converted into their bioactive form. Speck also provides the platform for gasdermin D protein activation, whose N-terminus domain perforates the plasma membrane, allowing the release of mature cytokines alongside with a highly inflammatory form of cell death, namely pyroptosis. Although controlled NLRP3 inflammasome-pyroptosis pathway activation preserves mucosal immunity homeostasis and contributes to host defense, a prolonged trigger is deleterious and could lead, in genetically predisposed subjects, to the onset of inflammatory bowel disease, including Crohn's disease and ulcerative colitis, as well as to gastrointestinal cancer. Experimental evidence shows that the NLRP3 inflammasome has both protective and pathogenic abilities. In this review we highlight the impact of the NLRP3-pyroptosis axis on the pathophysiology of the gastrointestinal tract at molecular level, focusing on newly discovered features bearing pro- and anti-inflammatory and neoplastic activity, and on targeted therapies tested in preclinical and clinical trials.
Collapse
Affiliation(s)
- Valentina Arrè
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Rosanna Scialpi
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Matteo Centonze
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Maria Principia Scavo
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Roberto Negro
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy.
| |
Collapse
|
6
|
Wang J, Hua S, Bao H, Yuan J, Zhao Y, Chen S. Pyroptosis and inflammasomes in cancer and inflammation. MedComm (Beijing) 2023; 4:e374. [PMID: 37752941 PMCID: PMC10518439 DOI: 10.1002/mco2.374] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Nonprogrammed cell death (NPCD) and programmed cell death (PCD) are two types of cell death. Cell death is significantly linked to tumor development, medication resistance, cancer recurrence, and metastatic dissemination. Therefore, a comprehensive understanding of cell death is essential for the treatment of cancer. Pyroptosis is a kind of PCD distinct from autophagy and apoptosis in terms of the structure and function of cells. The defining features of pyroptosis include the release of an inflammatory cascade reaction and the expulsion of lysosomes, inflammatory mediators, and other cellular substances from within the cell. Additionally, it displays variations in osmotic pressure both within and outside the cell. Pyroptosis, as evidenced by a growing body of research, is critical for controlling the development of inflammatory diseases and cancer. In this paper, we reviewed the current level of knowledge on the mechanism of pyroptosis and inflammasomes and their connection to cancer and inflammatory diseases. This article presents a theoretical framework for investigating the potential of therapeutic targets in cancer and inflammatory diseases, overcoming medication resistance, establishing nanomedicines associated with pyroptosis, and developing risk prediction models in refractory cancer. Given the link between pyroptosis and the emergence of cancer and inflammatory diseases, pyroptosis-targeted treatments may be a cutting-edge treatment strategy.
Collapse
Affiliation(s)
- Jie‐Lin Wang
- Department of Obstetrics and GynecologyGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Gynecologic Oncology Research OfficeGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Sheng‐Ni Hua
- Department of Radiation OncologyZhuhai Peoples HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiChina
| | - Hai‐Juan Bao
- Department of Obstetrics and GynecologyGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Gynecologic Oncology Research OfficeGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Jing Yuan
- Department of Obstetrics and GynecologyGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Gynecologic Oncology Research OfficeGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Yang Zhao
- Department of Obstetrics and GynecologyGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Gynecologic Oncology Research OfficeGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Shuo Chen
- Department of Obstetrics and GynecologyGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Department of Gynecologic Oncology Research OfficeGuangzhou Key Laboratory of Targeted Therapy for Gynecologic OncologyGuangdong Provincial Key Laboratory of Major Obstetric DiseasesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
7
|
Godala M, Gaszyńska E, Walczak K, Małecka-Wojciesko E. Role of Serum Interleukin-6, Interleukin-1β and Interleukin-10 in Assessment of Disease Activity and Nutritional Status in Patients with Inflammatory Bowel Disease. J Clin Med 2023; 12:5956. [PMID: 37762896 PMCID: PMC10532332 DOI: 10.3390/jcm12185956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are characterised by multifactorial and chronic inflammation. Much attention has been paid to immune dysfunction in inflammatory bowel diseases. The aim of this study was to assess the usefulness of serum IL-6, IL-1β and IL-10 in determining the activity and nutritional status in IBD patients. The case-control study was carried out on 82 patients with IBD; the control group consisted of 25 clinically healthy subjects. The serum concentrations of IL-6, IL-1 β and IL-10 were determined by the quantitative sandwich enzyme-linked immunosorbent assay. There were no significant differences in IL-6 and IL-1β levels in UC and CD patients according to disease activity as assessed by the Montreal classification, Partial Mayo Score and CDAI. Significantly higher IL-6 levels were found in patients with low body fat in comparison to patients with normal body fat. Furthermore, significantly higher mean IL-6 levels were observed in patients with excess body fat in comparison to patients with normal body fat, and also in comparison to patients with deficient body fat. IL-6 and IL-1β may provide extra information regarding the nutritional status of IBD patients. IL-10 can be considered a non-invasive biomarker of IBD activity.
Collapse
Affiliation(s)
- Małgorzata Godala
- Department of Nutrition and Epidemiology, Medical University of Lodz, 90-752 Lodz, Poland;
| | - Ewelina Gaszyńska
- Department of Nutrition and Epidemiology, Medical University of Lodz, 90-752 Lodz, Poland;
| | - Konrad Walczak
- Department of Internal Medicine and Nephrodiabetology, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Ewa Małecka-Wojciesko
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-419 Lodz, Poland;
| |
Collapse
|
8
|
Inflammatory cytokines directly disrupt the bovine intestinal epithelial barrier. Sci Rep 2022; 12:14578. [PMID: 36028741 PMCID: PMC9418144 DOI: 10.1038/s41598-022-18771-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/18/2022] [Indexed: 11/08/2022] Open
Abstract
The small intestinal mucosa constitutes a physical barrier separating the gut lumen from sterile internal tissues. Junctional complexes between cells regulate transport across the barrier, preventing water loss and the entry of noxious molecules or pathogens. Inflammatory diseases in cattle disrupt this barrier; nonetheless, mechanisms of barrier disruption in cattle are poorly understood. We investigated the direct effects of three inflammatory cytokines, TNFα, IFNγ, and IL-18, on the bovine intestinal barrier utilizing intestinal organoids. Flux of fluorescein isothiocyanate (FITC)-labeled dextran was used to investigate barrier permeability. Immunocytochemistry and transmission electron microscopy were used to investigate junctional morphology, specifically tortuosity and length/width, respectively. Immunocytochemistry and flow cytometry was used to investigate cellular turnover via proliferation and apoptosis. Our study shows that 24-h cytokine treatment with TNFα or IFNγ significantly increased dextran permeability and tight junctional tortuosity, and reduced cellular proliferation. TNFα reduced the percentage of G2/M phase cells, and IFNγ treatment increased cell apoptotic rate. IL-18 did not directly induce significant changes to barrier permeability or cellular turnover. Our study concludes that the inflammatory cytokines, TNFα and IFNγ, directly induce intestinal epithelial barrier dysfunction and alter the tight junctional morphology and rate of cellular turnover in bovine intestinal epithelial cells.
Collapse
|
9
|
Ihim SA, Abubakar SD, Zian Z, Sasaki T, Saffarioun M, Maleknia S, Azizi G. Interleukin-18 cytokine in immunity, inflammation, and autoimmunity: Biological role in induction, regulation, and treatment. Front Immunol 2022; 13:919973. [PMID: 36032110 PMCID: PMC9410767 DOI: 10.3389/fimmu.2022.919973] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/25/2022] [Indexed: 12/13/2022] Open
Abstract
Interleukin-18 (IL-18) is a potent pro-inflammatory cytokine involved in host defense against infections and regulates the innate and acquired immune response. IL-18 is produced by both hematopoietic and non-hematopoietic cells, including monocytes, macrophages, keratinocytes and mesenchymal cell. IL-18 could potentially induce inflammatory and cytotoxic immune cell activities leading to autoimmunity. Its elevated levels have been reported in the blood of patients with some immune-related diseases, including rheumatoid arthritis, systemic lupus erythematosus, type I diabetes mellitus, atopic dermatitis, psoriasis, and inflammatory bowel disease. In the present review, we aimed to summarize the biological properties of IL-18 and its pathological role in different autoimmune diseases. We also reported some monoclonal antibodies and drugs targeting IL-18. Most of these monoclonal antibodies and drugs have only produced partial effectiveness or complete ineffectiveness in vitro, in vivo and human studies. The ineffectiveness of these drugs targeting IL-18 may be largely due to the loophole caused by the involvement of other cytokines and proteins in the signaling pathway of many inflammatory diseases besides the involvement of IL-18. Combination drug therapies, that focus on IL-18 inhibition, in addition to other cytokines, are highly recommended to be considered as an important area of research that needs to be explored.
Collapse
Affiliation(s)
- Stella Amarachi Ihim
- Department of Molecular and Cellular Pharmacology, University of Shizuoka, Shizuoka, Japan
- Department of Pharmacology and Toxicology, University of Nigeria, Nsukka, Nigeria
- Department of Science Laboratory Technology, University of Nigeria, Nsukka, Nigeria
| | - Sharafudeen Dahiru Abubakar
- Division of Molecular Pathology, Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan
- Department of Medical Laboratory Science, College of Medical Science, Ahmadu Bello University, Zaria, Nigeria
| | - Zeineb Zian
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Takanori Sasaki
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Mohammad Saffarioun
- Biopharmaceutical Research Center, AryoGen Pharmed Inc., Alborz University of Medical Sciences, Karaj, Iran
| | - Shayan Maleknia
- Biopharmaceutical Research Center, AryoGen Pharmed Inc., Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- *Correspondence: Gholamreza Azizi,
| |
Collapse
|
10
|
Li Y, Lin X, Wang W, Wang W, Cheng S, Huang Y, Zou Y, Ke J, Zhu L. The Proinflammatory Role of Guanylate-Binding Protein 5 in Inflammatory Bowel Diseases. Front Microbiol 2022; 13:926915. [PMID: 35722277 PMCID: PMC9201962 DOI: 10.3389/fmicb.2022.926915] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/09/2022] [Indexed: 01/31/2023] Open
Abstract
NLRP3 inflammasome is implicated in the pathogenesis of inflammatory bowel diseases (IBD). Since guanylate-binding protein 5 (GBP5) induces the NLRP3 inflammasome activity, we aim to investigate the potential role of GBP5 in IBD pathogenesis. The expression of GBP5, NLRP3 inflammasome, and related cytokines and chemokines was examined in two cohorts of IBD patients and healthy controls, by microarray transcriptome analysis and quantitative real-time PCR. Cellular localization of GBP5 in colonic biopsies was examined by immunohistochemistry and immunofluorescence with confocal microscopy. For functional studies, GBP5 was induced by interferon γ or silenced by siRNA or CRISPR/CAS9 technique, and inflammatory activities were evaluated at mRNA and protein levels. We found that the expression of GBP5 was elevated in colonic mucosa in two geographically and culturally distinct IBD cohorts. In colonic tissues of IBD patients, GBP5 expression was mainly confined to immune cells and the levels of GBP5 expression were correlated with those of the inflammatory cytokines and chemokines. In cultured T and macrophage cells, the expression of proinflammatory cytokines and chemokines was increased when GBP5 was induced, while GBP5 deficiency leads to decreased expression of proinflammatory mediators including gasdermin D, caspase 1, cytokines, and chemokines. We conclude that GBP5 is required in the expression of many proinflammatory cytokines and chemokines in intestinal immune cells. In addition, GBP5 may upregulate inflammatory reactions through an inflammasome-mediated mechanism. Since GBP5 plays a proinflammatory role at the early steps of the inflammatory cascades of IBD pathogenesis, and is implicated in IBD patients of distinct genetic and environmental backgrounds, targeting GBP5 could be an effective strategy for the management of IBD.
Collapse
Affiliation(s)
- Yichen Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, The Sixth Affiliated Hospital, Guangdong Institute of Gastroenterology, Sun Yat-sen University, Guangzhou, China
| | - Xutao Lin
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Guangdong Institute of Gastroenterology, Sun Yat-sen University, Guangzhou, China
| | - Wenxia Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, The Sixth Affiliated Hospital, Guangdong Institute of Gastroenterology, Sun Yat-sen University, Guangzhou, China
| | - Wenyu Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, The Sixth Affiliated Hospital, Guangdong Institute of Gastroenterology, Sun Yat-sen University, Guangzhou, China
| | - Sijing Cheng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, The Sixth Affiliated Hospital, Guangdong Institute of Gastroenterology, Sun Yat-sen University, Guangzhou, China.,School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Yibo Huang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, The Sixth Affiliated Hospital, Guangdong Institute of Gastroenterology, Sun Yat-sen University, Guangzhou, China
| | - Yifeng Zou
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, The Sixth Affiliated Hospital, Guangdong Institute of Gastroenterology, Sun Yat-sen University, Guangzhou, China
| | - Jia Ke
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, The Sixth Affiliated Hospital, Guangdong Institute of Gastroenterology, Sun Yat-sen University, Guangzhou, China
| | - Lixin Zhu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, The Sixth Affiliated Hospital, Guangdong Institute of Gastroenterology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
11
|
Abraham C, Abreu MT, Turner JR. Pattern Recognition Receptor Signaling and Cytokine Networks in Microbial Defenses and Regulation of Intestinal Barriers: Implications for Inflammatory Bowel Disease. Gastroenterology 2022; 162:1602-1616.e6. [PMID: 35149024 PMCID: PMC9112237 DOI: 10.1053/j.gastro.2021.12.288] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/30/2021] [Accepted: 12/10/2021] [Indexed: 12/23/2022]
Abstract
Inflammatory bowel disease is characterized by defects in epithelial function and dysregulated inflammatory signaling by lamina propria mononuclear cells including macrophages and dendritic cells in response to microbiota. In this review, we focus on the role of pattern recognition receptors in the inflammatory response as well as epithelial barrier regulation. We explore cytokine networks that increase inflammation, regulate paracellular permeability, cause epithelial damage, up-regulate epithelial proliferation, and trigger restitutive processes. We focus on studies using patient samples as well as speculate on pathways that can be targeted to more holistically treat patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Clara Abraham
- Department of Internal Medicine, Yale University, New Haven, Connecticut.
| | - Maria T. Abreu
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Miami Leonard Miller School of Medicine, Miami, FL
| | - Jerrold R. Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
12
|
Ambrose PA, Goodman WA. Impact of COVID-19 on Patients with Inflammatory Bowel Disease. JOURNAL OF EXPLORATORY RESEARCH IN PHARMACOLOGY 2022; 7:37-44. [PMID: 35966234 PMCID: PMC9373928 DOI: 10.14218/jerp.2021.00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first identified in Wuhan, China, in late 2019. Responsible for the ongoing coronavirus disease 2019 (COVID-19) pandemic, SARS-CoV-2 is one of three structurally similar beta-coronaviruses that can cause a strong upregulation of cytokines referred to as cytokine release syndrome (CRS). Unresolved CRS leads to respiratory symptoms, including pneumonia, and in more severe cases, acute respiratory distress syndrome (ARDS). Although COVID-19 is widely known for these hallmark respiratory symptoms, it also impacts the gut, causing gastrointestinal (GI) tract inflammation and diarrhea. COVID-19's GI symptoms may be due to the high intestinal expression of angiotensin converting enzyme-2 receptors, which are for the binding of SARS-CoV-2 viral particles. Reports have shown that SARS-CoV-2 can be passed through fecal matter, with one study finding that 48.1% of COVID-19 patients expressed viral SARS-CoV-2 mRNA in their stool. Given that the GI tract is a target tissue affected by COVID-19, this causes concern for those with underlying GI pathologies, such as inflammatory bowel disease (IBD). Regrettably, there have been only limited studies on the impact of COVID-19 on gut health, and the impact of COVID-19 on intestinal inflammation among IBD patients remains unclear. In particular, questions regarding susceptibility to SARS-CoV-2 infection, clinical impact of COVID-19 on IBD, and the potential influence of age, sex, and immunosuppressant medications are still poorly understood. An improved understanding of these issues is needed to address the unique risks of COVID-19 among IBD patients, as well as the potential impact of SARS-CoV-2 on the host intestinal microbiota.
Collapse
Affiliation(s)
- Paula A. Ambrose
- Department of Pathology, Case Western Reserve University School of Medicine, OH, USA
| | - Wendy A. Goodman
- Department of Pathology, Case Western Reserve University School of Medicine, OH, USA
| |
Collapse
|
13
|
Sarmento A, Simões CD. Gut Microbiota Dysbiosis and Chronic Intestinal Inflammation. COMPREHENSIVE GUT MICROBIOTA 2022:423-441. [DOI: 10.1016/b978-0-12-819265-8.00057-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
SEĞMEN B, YURTTUTAN S, SERİNGEÇ AKKEÇECİ BN, İNANÇ TOLUN F, BOZKAYA A. Neonatal Sepsis Şüphesi Olan Yenidoğanlarda Akut Faz Reaktanı Olarak CRP, Prokalsitonin, İnterlökin-18 Düzeylerinin Değerlendirilmesi. KAHRAMANMARAŞ SÜTÇÜ İMAM ÜNIVERSITESI TIP FAKÜLTESI DERGISI 2021. [DOI: 10.17517/ksutfd.879147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
15
|
Jarret A, Jackson R, Duizer C, Healy ME, Zhao J, Rone JM, Bielecki P, Sefik E, Roulis M, Rice T, Sivanathan KN, Zhou T, Solis AG, Honcharova-Biletska H, Vélez K, Hartner S, Low JS, Qu R, de Zoete MR, Palm NW, Ring AM, Weber A, Moor AE, Kluger Y, Nowarski R, Flavell RA. Enteric Nervous System-Derived IL-18 Orchestrates Mucosal Barrier Immunity. Cell 2020; 180:50-63.e12. [PMID: 31923399 PMCID: PMC7339937 DOI: 10.1016/j.cell.2019.12.016] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 11/01/2019] [Accepted: 12/12/2019] [Indexed: 12/18/2022]
Abstract
Mucosal barrier immunity is essential for the maintenance of the commensal microflora and combating invasive bacterial infection. Although immune and epithelial cells are thought to be the canonical orchestrators of this complex equilibrium, here, we show that the enteric nervous system (ENS) plays an essential and non-redundant role in governing the antimicrobial protein (AMP) response. Using confocal microscopy and single-molecule fluorescence in situ mRNA hybridization (smFISH) studies, we observed that intestinal neurons produce the pleiotropic cytokine IL-18. Strikingly, deletion of IL-18 from the enteric neurons alone, but not immune or epithelial cells, rendered mice susceptible to invasive Salmonella typhimurium (S.t.) infection. Mechanistically, unbiased RNA sequencing and single-cell sequencing revealed that enteric neuronal IL-18 is specifically required for homeostatic goblet cell AMP production. Together, we show that neuron-derived IL-18 signaling controls tissue-wide intestinal immunity and has profound consequences on the mucosal barrier and invasive bacterial killing.
Collapse
Affiliation(s)
- Abigail Jarret
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ruaidhrí Jackson
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Coco Duizer
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Marc E Healy
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich 8091, Switzerland; Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Jun Zhao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Joseph M Rone
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Piotr Bielecki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Esen Sefik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Manolis Roulis
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tyler Rice
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kisha N Sivanathan
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ting Zhou
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Angel G Solis
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Hanna Honcharova-Biletska
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich 8091, Switzerland
| | - Karelia Vélez
- Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Saskia Hartner
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; University of Vienna, Universitätsring 1, Wien 1010, Austria
| | - Jun Siong Low
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rihao Qu
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Marcel R de Zoete
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Aaron M Ring
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich 8091, Switzerland; Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Andreas E Moor
- Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Yuval Kluger
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA; Applied Mathematics Program, Yale University, New Haven, CT 06511, USA
| | - Roni Nowarski
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
16
|
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: 0.8] [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
|
17
|
An innate interaction between IL-18 and the propeptide that inactivates its precursor form. Sci Rep 2019; 9:6160. [PMID: 30992532 PMCID: PMC6467916 DOI: 10.1038/s41598-019-42661-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/01/2019] [Indexed: 01/03/2023] Open
Abstract
Uncontrolled secretion of mature interleukin (IL)-1β and IL-18 is responsible for severe autoinflammatory or autoimmune disorders and various allergic diseases. Here we report an intramolecular interaction between IL-18 and its propeptide, which is proteolytically removed from its precursor proIL-18 during maturation. The intramolecular interaction was recapitulated intermolecularly using recombinant propeptide. These results suggest the possibility of developing a novel class of peptide-based IL-18 inhibitors that could serve as therapeutic agents for IL-18-related inflammatory diseases.
Collapse
|
18
|
Abstract
NLRP3 inflammasome can be widely found in epithelial cells and immune cells. The NOD-like receptors (NLRs) family member NLRP3 contains a central nucleotide-binding and oligomerization (NACHT) domain which facilitates self-oligomerization and has ATPase activity. The C-terminal conserves a leucine-rich repeats (LRRs) domain which can modulate NLRP3 activity and sense endogenous alarmins and microbial ligands. In contrast, the N-terminal pyrin domain (PYD) can account for homotypic interactions with the adaptor protein-ASC of NLRP3 inflammasome. These characters enable it function in innate immunity. Its downstream effector proteins include caspase-1 and IL-1β etc. which exhibit protective or detrimental roles in mucosal immunity in different studies. Here, we comprehensively review the current literature regarding the physiology of NLRP3 inflammasome and its potential roles in the pathogenesis of IBD. We also discuss about the complex interactions among the NLRP3 inflammasome, mucosal immune response, and gut homeostasis as found in experimental models and IBD patients.
Collapse
Affiliation(s)
- Yu Zhen
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- The Centre of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Hu Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- The Centre of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Wosen JE, Mukhopadhyay D, Macaubas C, Mellins ED. Epithelial MHC Class II Expression and Its Role in Antigen Presentation in the Gastrointestinal and Respiratory Tracts. Front Immunol 2018; 9:2144. [PMID: 30319613 PMCID: PMC6167424 DOI: 10.3389/fimmu.2018.02144] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/30/2018] [Indexed: 12/13/2022] Open
Abstract
As the primary barrier between an organism and its environment, epithelial cells are well-positioned to regulate tolerance while preserving immunity against pathogens. Class II major histocompatibility complex molecules (MHC class II) are highly expressed on the surface of epithelial cells (ECs) in both the lung and intestine, although the functional consequences of this expression are not fully understood. Here, we summarize current information regarding the interactions that regulate the expression of EC MHC class II in health and disease. We then evaluate the potential role of EC as non-professional antigen presenting cells. Finally, we explore future areas of study and the potential contribution of epithelial surfaces to gut-lung crosstalk.
Collapse
Affiliation(s)
- Jonathan E Wosen
- Program in Immunology, Department of Pediatrics, Stanford University, Stanford, CA, United States
| | - Dhriti Mukhopadhyay
- Program in Immunology, Department of Pediatrics, Stanford University, Stanford, CA, United States
| | - Claudia Macaubas
- Program in Immunology, Department of Pediatrics, Stanford University, Stanford, CA, United States
| | - Elizabeth D Mellins
- Program in Immunology, Department of Pediatrics, Stanford University, Stanford, CA, United States
| |
Collapse
|
20
|
NLRP1 restricts butyrate producing commensals to exacerbate inflammatory bowel disease. Nat Commun 2018; 9:3728. [PMID: 30214011 PMCID: PMC6137172 DOI: 10.1038/s41467-018-06125-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 08/21/2018] [Indexed: 12/26/2022] Open
Abstract
Anti-microbial signaling pathways are normally triggered by innate immune receptors when detecting pathogenic microbes to provide protective immunity. Here we show that the inflammasome sensor Nlrp1 aggravates DSS-induced experimental mouse colitis by limiting beneficial, butyrate-producing Clostridiales in the gut. The colitis-protective effects of Nlrp1 deficiency are thus reversed by vancomycin treatment, but recapitulated with butyrate supplementation in wild-type mice. Moreover, an activating mutation in Nlrp1a increases IL-18 and IFNγ production, and decreases colonic butyrate to exacerbate colitis. We also show that, in patients with ulcerative colitis, increased NLRP1 in inflamed regions of the colon is associated with increased IFN-γ. In this context, NLRP1, IL-18 or IFN-γ expression negatively correlates with the abundance of Clostridiales in human rectal mucosal biopsies. Our data identify the NLRP1 inflammasome to be a key negative regulator of protective, butyrate-producing commensals, which therefore promotes inflammatory bowel disease.
Collapse
|
21
|
Abstract
Danger signals are a hallmark of many common inflammatory diseases, and these stimuli can function to activate the cytosolic innate immune signalling receptor NLRP3 (NOD-, LRR- and pyrin domain-containing 3). Once activated, NLRP3 nucleates the assembly of an inflammasome, leading to caspase 1-mediated proteolytic activation of the interleukin-1β (IL-1β) family of cytokines, and induces an inflammatory, pyroptotic cell death. Pharmacological inhibition of NLRP3 activation results in potent therapeutic effects in a wide variety of rodent models of inflammatory diseases, effects that are mirrored by genetic ablation of NLRP3. Although these findings highlight the potential of NLRP3 as a drug target, an understanding of NLRP3 structure and activation mechanisms is incomplete, which has hampered the discovery and development of novel therapeutics against this target. Here, we review recent advances in our understanding of NLRP3 activation and regulation, highlight the evolving landscape of NLRP3 modulators and discuss opportunities for pharmacologically targeting NLRP3 with novel small molecules.
Collapse
|
22
|
Kurioka A, Cosgrove C, Simoni Y, van Wilgenburg B, Geremia A, Björkander S, Sverremark-Ekström E, Thurnheer C, Günthard HF, Khanna N, Walker LJ, Arancibia-Cárcamo CV, Newell EW, Willberg CB, Klenerman P. CD161 Defines a Functionally Distinct Subset of Pro-Inflammatory Natural Killer Cells. Front Immunol 2018; 9:486. [PMID: 29686665 PMCID: PMC5900032 DOI: 10.3389/fimmu.2018.00486] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 02/23/2018] [Indexed: 01/22/2023] Open
Abstract
CD161 is a C-type lectin-like receptor expressed on the majority of natural killer (NK) cells; however, the significance of CD161 expression on NK cells has not been comprehensively investigated. Recently, we found that CD161 expression identifies a transcriptional and innate functional phenotype that is shared across various T cell populations. Using mass cytometry and microarray experiments, we demonstrate that this functional phenotype extends to NK cells. CD161 marks NK cells that have retained the ability to respond to innate cytokines during their differentiation, and is lost upon cytomegalovirus-induced maturation in both healthy and human immunodeficiency virus (HIV)-infected patients. These pro-inflammatory NK cells are present in the inflamed lamina propria where they are enriched for integrin CD103 expression. Thus, CD161 expression identifies NK cells that may contribute to inflammatory disease pathogenesis and correlates with an innate responsiveness to cytokines in both T and NK cells.
Collapse
Affiliation(s)
- Ayako Kurioka
- The Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Cormac Cosgrove
- Ragon Institute of Massachusetts General Hospital, Harvard University, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Yannick Simoni
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore, Singapore
| | - Bonnie van Wilgenburg
- The Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Alessandra Geremia
- Translational Gastroenterology Unit, Experimental Medicine Division, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Sophia Björkander
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Christine Thurnheer
- Division of Infectious Diseases, University Hospital Berne, University of Berne, Berne, Switzerland
| | - Huldrych F. Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Nina Khanna
- Division of Infectious Diseases, University Hospital Basel, Basel, Switzerland
| | - Lucy Jane Walker
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Carolina V. Arancibia-Cárcamo
- Translational Gastroenterology Unit, Experimental Medicine Division, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Evan W. Newell
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore, Singapore
| | - Christian B. Willberg
- The Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Paul Klenerman
- The Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
23
|
Nold-Petry CA, Nold MF, Levy O, Kliger Y, Oren A, Borukhov I, Becker C, Wirtz S, Sandhu MK, Neurath M, Dinarello CA. Gp96 Peptide Antagonist gp96-II Confers Therapeutic Effects in Murine Intestinal Inflammation. Front Immunol 2017; 8:1531. [PMID: 29312281 PMCID: PMC5732239 DOI: 10.3389/fimmu.2017.01531] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/27/2017] [Indexed: 01/31/2023] Open
Abstract
Background The expression of heat shock protein gp96 is strongly correlated with the degree of tissue inflammation in ulcerative colitis and Crohn’s disease, thereby leading us to the hypothesis that inhibition of expression via gp96-II peptide prevents intestinal inflammation. Methods We employed daily injections of gp96-II peptide in two murine models of intestinal inflammation, the first resulting from five daily injections of IL-12/IL-18, the second via a single intrarectal application of TNBS (2,4,6-trinitrobenzenesulfonic acid). We also assessed the effectiveness of gp96-II peptide in murine and human primary cell culture. Results In the IL-12/IL-18 model, all gp96-II peptide-treated animals survived until day 5, whereas 80% of placebo-injected animals died. gp96-II peptide reduced IL-12/IL-18-induced plasma IFNγ by 89%, IL-1β by 63%, IL-6 by 43% and tumor necrosis factor (TNF) by 70% compared to controls. The clinical assessment Disease Activity Index of intestinal inflammation severity was found to be significantly lower in the gp96-II-treated animals when compared to vehicle-injected mice. gp96-II peptide treatment in the TNBS model limited weight loss to 5% on day 7 compared with prednisolone treatment, whereas placebo-treated animals suffered a 20% weight loss. Histological disease severity was reduced equally by prednisolone (by 40%) and gp96-II peptide (35%). Mice treated with either gp96-II peptide or prednisolone exhibited improved endoscopic scores compared with vehicle-treated control mice: vascularity, fibrin, granularity, and translucency scores were reduced by up to 49% by prednisolone and by up to 30% by gp96-II peptide. In vitro, gp96-II peptide reduced TLR2-, TLR4- and IL-12/IL-18-induced cytokine expression in murine splenocytes, with declines in constitutive IL-6 (54%), lipopolysaccharide-induced TNF (48%), IL-6 (81%) and in Staphylococcus epidermidis-induced TNF (67%) and IL-6 (81%), as well as IL-12/IL-18-induced IFNγ (75%). gp96-II peptide reduced IL–1β, IL-6, TNF and GM-CSF in human peripheral blood mononuclear cells to a similar degree without affecting cell viability, whereas RANTES, IL-25 and MIF were twofold to threefold increased. Conclusion gp96-II peptide protects against murine intestinal inflammation by regulating inflammation in vivo and in vitro, pointing to its promise as a novel treatment for inflammatory bowel disease.
Collapse
Affiliation(s)
- Claudia A Nold-Petry
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Marcel F Nold
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | | | | | | | | | - Christoph Becker
- Medical Clinic 1, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Stefan Wirtz
- Medical Clinic 1, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Manjeet K Sandhu
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Gastroenterology, Monash Health, Clayton, VIC, Australia
| | - Markus Neurath
- Medical Clinic 1, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| |
Collapse
|
24
|
Inflammasomes and intestinal inflammation. Mucosal Immunol 2017; 10:865-883. [PMID: 28401932 DOI: 10.1038/mi.2017.19] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 02/19/2017] [Indexed: 02/04/2023]
Abstract
The inflammasome is a cytosolic multi-protein innate immune rheostat, sensing a variety of endogenous and environmental stimuli, and regulating homeostasis or damage control. In the gastrointestinal tract, inflammasomes orchestrate immune tolerance to microbial and potentially food-related signals or drive the initiation of inflammatory responses to invading pathogens. When inadequately regulated, intestinal inflammasome activation leads to a perpetuated inflammatory response leading to immune pathology and tissue damage. In this review, we present the main features of the predominant types of inflammasomes participating in intestinal homeostasis and inflammation. We then discuss current controversies and open questions related to their functions and implications in disease, highlighting how pathological inflammasome over-activation or impaired function impact gut homeostasis, the microbiome ecosystem, and the propensity to develop gut-associated diseases. Collectively, understanding of the molecular basis of intestinal inflammasome signaling may be translated into clinical manipulation of this fundamental pathway as a potential immune modulatory therapeutic intervention.
Collapse
|
25
|
Ono Y, Kanmura S, Morinaga Y, Oda K, Kawabata K, Arima S, Sasaki F, Nasu Y, Tanoue S, Hashimoto S, Taguchi H, Uto H, Tsubouchi H, Ido A. The utility of apoptosis inhibitor of macrophages as a possible diagnostic marker in patients with Crohn's disease. BMC Gastroenterol 2017; 17:40. [PMID: 28284201 PMCID: PMC5346245 DOI: 10.1186/s12876-017-0591-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 02/25/2017] [Indexed: 01/14/2023] Open
Abstract
Background Apoptosis inhibitor of macrophages (AIM) was initially identified as an apoptosis inhibitor that supports the survival of macrophages against various apoptosis-inducing stimuli, and AIM produced by macrophages may contribute to the pathogenesis of inflammatory bowel diseases (IBDs). However, there have been no reports on the kinetics of AIM in IBD and the impact of AIM on the pathogenesis of IBD. In this study, we aimed to investigate the diagnostic utility of levels of AIM and their correlation with the activity of Crohn’s disease (CD) and IBD. Methods We used an enzyme-linked immunosorbent assay (ELISA) to examine AIM serum levels in 16 healthy subjects and 90 patients with inflammatory bowel diseases, namely 39 with CD and 51 with ulcerative colitis (UC), as well as 17 patients with Behcet’s disease (BD) as intestinal disease controls. We compared serum AIM levels among groups and examined whether there were correlations between serum AIM levels and disease activity and type. We also performed immunohistochemical staining of AIM in intestinal tissues of patients with CD. Results Serum AIM levels were significantly higher in patients with CD than in patients with UC, BD, and controls (3.27 ± 2.14, 1.88 ± 1.43, 2.34 ± 1.37, and 2.13 ± 0.64 μg/ml, respectively; P < 0.01). There was no difference in serum AIM levels before and after treatment in patients with CD. However, in these patients the diagnostic rate using AIM was better than that based on anti-Saccharomyces cerevisiae antibodies. AIM was expressed in macrophages that were positive for CD14, CD16, or both in the intestinal tissues of patients with CD. Conclusions AIM is a novel biomarker of CD that can distinguish CD from UC or BD. It is suggested that AIM may contribute to intestinal inflammation by inhibiting the apoptosis of macrophages.
Collapse
Affiliation(s)
- Yohei Ono
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Shuji Kanmura
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan.
| | - Yuko Morinaga
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Kohei Oda
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Katsuto Kawabata
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Shiho Arima
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Fumisato Sasaki
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Yuichirou Nasu
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Shiroh Tanoue
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Shinichi Hashimoto
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Hiroki Taguchi
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Hirofumi Uto
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan.,Center for Digestive and Liver Diseases, Miyazaki Medical Center Hospital, Miyazaki, Japan
| | | | - Akio Ido
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| |
Collapse
|
26
|
Zhou Z, Ding M, Huang L, Gilkeson G, Lang R, Jiang W. Toll-like receptor-mediated immune responses in intestinal macrophages; implications for mucosal immunity and autoimmune diseases. Clin Immunol 2016; 173:81-86. [PMID: 27620642 PMCID: PMC5148676 DOI: 10.1016/j.clim.2016.09.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/04/2016] [Accepted: 09/07/2016] [Indexed: 12/21/2022]
Abstract
Monocytes are precursors of macrophages and key players during inflammation and pathogen challenge in the periphery, whereas intestinal resident macrophages act as innate effector cells to engulf and clear bacteria, secrete cytokines, and maintain intestinal immunity and homeostasis. However, perturbation of toll-like receptor signaling pathway in intestinal macrophages has been associated with tolerance breakdown in autoimmune diseases. In the present review, we have summarized and discussed the role of toll-like receptor signals in human intestinal macrophages, and the role of human intestinal macrophages in keeping human intestinal immunity, homeostasis, and autoimmune diseases.
Collapse
Affiliation(s)
- Zejun Zhou
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston 29425, USA
| | - Miao Ding
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston 29425, USA
| | - Lei Huang
- The 302 Hospital of PLA, Treatment and Research Center for Infectious Diseases, Beijing 100039, China
| | - Gary Gilkeson
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston 29425, SC, USA
| | - Ren Lang
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 10020, China.
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston 29425, USA; Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston 29425, USA.
| |
Collapse
|
27
|
Timperi E, Pacella I, Schinzari V, Focaccetti C, Sacco L, Farelli F, Caronna R, Del Bene G, Longo F, Ciardi A, Morelli S, Vestri AR, Chirletti P, Barnaba V, Piconese S. Regulatory T cells with multiple suppressive and potentially pro-tumor activities accumulate in human colorectal cancer. Oncoimmunology 2016; 5:e1175800. [PMID: 27622025 PMCID: PMC5006916 DOI: 10.1080/2162402x.2016.1175800] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/01/2016] [Accepted: 04/01/2016] [Indexed: 01/05/2023] Open
Abstract
Tregs can contribute to tumor progression by suppressing antitumor immunity. Exceptionally, in human colorectal cancer (CRC), Tregs are thought to exert beneficial roles in controlling pro-tumor chronic inflammation. The goal of our study was to characterize CRC-infiltrating Tregs at multiple levels, by phenotypical, molecular and functional evaluation of Tregs from the tumor site, compared to non-tumoral mucosa and peripheral blood of CRC patients. The frequency of Tregs was higher in mucosa than in blood, and further significantly increased in tumor. Ex vivo, those Tregs suppressed the proliferation of tumor-infiltrating CD8(+) and CD4(+) T cells. A differential compartmentalization was detected between Helios(high) and Helios(low) Treg subsets (thymus-derived versus peripherally induced): while Helios(low) Tregs were enriched in both sites, only Helios(high) Tregs accumulated significantly and specifically in tumors, displayed a highly demethylated TSDR region and contained high proportions of cells expressing CD39 and OX40, markers of activation and suppression. Besides the suppression of T cells, Tregs may contribute to CRC progression also through releasing IL-17, or differentiating into Tfr cells that potentially antagonize a protective Tfh response, events that were both detected in tumor-associated Tregs. Overall, our data indicate that Treg accumulation may contribute through multiple mechanisms to CRC establishment and progression.
Collapse
Affiliation(s)
- Eleonora Timperi
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Ilenia Pacella
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Valeria Schinzari
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Chiara Focaccetti
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Luca Sacco
- Sezione di Chirurgia Interdisciplinare “F. Durante”, Sapienza Università di Roma, Rome, Italy
| | - Francesco Farelli
- Sezione di Chirurgia Interdisciplinare “F. Durante”, Sapienza Università di Roma, Rome, Italy
| | - Roberto Caronna
- Sezione di Chirurgia Interdisciplinare “F. Durante”, Sapienza Università di Roma, Rome, Italy
| | - Gabriella Del Bene
- Dipartimento di Medicina Molecolare, Sapienza Università di Roma, Rome, Italy
| | - Flavia Longo
- Dipartimento di Medicina Molecolare, Sapienza Università di Roma, Rome, Italy
| | - Antonio Ciardi
- Dipartimento di Scienze Radiologiche, Oncologiche e Anatomo-Patologiche, Sapienza Università di Roma, Rome, Italy
| | - Sergio Morelli
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Anna Rita Vestri
- Dipartimento di Sanità Pubblica e Malattie Infettive, Sapienza Università di Roma, Rome, Italy
| | - Piero Chirletti
- Sezione di Chirurgia Interdisciplinare “F. Durante”, Sapienza Università di Roma, Rome, Italy
| | - Vincenzo Barnaba
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Silvia Piconese
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
28
|
Cassol E, Rossouw T, Malfeld S, Mahasha P, Slavik T, Seebregts C, Bond R, du Plessis J, Janssen C, Roskams T, Nevens F, Alfano M, Poli G, van der Merwe SW. CD14(+) macrophages that accumulate in the colon of African AIDS patients express pro-inflammatory cytokines and are responsive to lipopolysaccharide. BMC Infect Dis 2015; 15:430. [PMID: 26475133 PMCID: PMC4609115 DOI: 10.1186/s12879-015-1176-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 10/05/2015] [Indexed: 01/05/2023] Open
Abstract
Background Intestinal macrophages are key regulators of inflammatory responses to the gut microbiome and play a central role in maintaining tissue homeostasis and epithelial integrity. However, little is known about the role of these cells in HIV infection, a disease fuelled by intestinal inflammation, a loss of epithelial barrier function and increased microbial translocation (MT). Methods Phenotypic and functional characterization of intestinal macrophages was performed for 23 African AIDS patients with chronic diarrhea and/or weight loss and 11 HIV-negative Africans with and without inflammatory bowel disease (IBD). AIDS patients were treated with cotrimoxazole for the prevention of opportunistic infections (OIs). Macrophage phenotype was assessed by flow cytometry and immuno-histochemistry (IHC); production of proinflammatory mediators by IHC and Qiagen PCR Arrays; in vitro secretion of cytokines by the Bio-Plex Suspension Array System. Statistical analyses were performed using Spearman’s correlation and Wilcoxon matched-pair tests. Results between groups were analyzed using the Kruskal-Wallis with Dunn’s post-test and the Mann–Whitney U tests. Results None of the study participants had evidence of enteric co-infections as assessed by stool analysis and histology. Compared to healthy HIV-negative controls, the colon of AIDS patients was highly inflamed with increased infiltration of inflammatory cells and increased mRNA expression of proinflammatory cytokine (tumour necrosis factor (TNF)-α, interleukin (IL)-1β, IFN-γ, and IL-18), chemokines (chemokine (C-C motif) ligand (CCL)2 and chemokine (C-X-C) motif ligand (CXCL)10) and transcription factors (TNF receptor-associated factor (TRAF)6 and T-box (TXB)21). IHC revealed significant co-localization of TNF-α and IL-1β with CD68+ cells. As in IBD, HIV was associated with a marked increase in macrophages expressing innate response receptors including CD14, the co-receptor for lipopolysaccharide (LPS). The frequency of CD14+ macrophages correlated positively with plasma LPS, a marker of MT. Total unfractionated mucosal mononuclear cells (MMC) isolated from the colon of AIDS patients, but not MMC depleted of CD14+ cells, secreted increased levels of proinflammatory cytokines ex vivo in response to LPS. Conclusions Intestinal macrophages, in the absence of overt OIs, play an important role in driving persistent inflammation in HIV patients with late-stage disease and diarrhea. These results suggest intensified treatment strategies that target inflammatory processes in intestinal macrophages may be highly beneficial in restoring the epithelial barrier and limiting MT in HIV-infected patients.
Collapse
Affiliation(s)
- Edana Cassol
- MRC Unit for Inflammation and Immunity, Department of Immunology and the Tshwane Academic Division of the National Health Laboratory Service, University of Pretoria, Pretoria, South Africa. .,Department of Health Sciences, Carleton University, 5433 Herzberg Laboratories, 1125 Colonel By Drive, Ottawa, Ontario, K1S 5B6, Canada.
| | - Theresa Rossouw
- MRC Unit for Inflammation and Immunity, Department of Immunology and the Tshwane Academic Division of the National Health Laboratory Service, University of Pretoria, Pretoria, South Africa. .,Department of Family Medicine, University of Pretoria, Pretoria, South Africa.
| | - Susan Malfeld
- MRC Unit for Inflammation and Immunity, Department of Immunology and the Tshwane Academic Division of the National Health Laboratory Service, University of Pretoria, Pretoria, South Africa.
| | - Phetole Mahasha
- MRC Unit for Inflammation and Immunity, Department of Immunology and the Tshwane Academic Division of the National Health Laboratory Service, University of Pretoria, Pretoria, South Africa.
| | - Tomas Slavik
- Department of Anatomical Pathology, University of Pretoria and Ampath Pathology Laboratories, Pretoria, South Africa.
| | - Chris Seebregts
- Jembi Health Systems NPC, Durban, South Africa. .,School of Mathematics, Statistics and Computer Science, University of KwaZulu-Natal, Durban, South Africa.
| | - Robert Bond
- Hepatology and GI-Research Laboratory, University of Pretoria, Pretoria, South Africa.
| | - Johannie du Plessis
- Hepatology and GI-Research Laboratory, University of Pretoria, Pretoria, South Africa.
| | - Carl Janssen
- Hepatology and GI-Research Laboratory, University of Pretoria, Pretoria, South Africa.
| | - Tania Roskams
- Translational Cell and Tissue Research, Department of Imaging and Pathology, University of Leuven, Leuven, Belgium.
| | - Frederik Nevens
- Department of Hepatology, University of Leuven, Leuven, Belgium.
| | - Massimo Alfano
- San Raffaele Scientific Institute, School of Medicine, Milan, Italy. .,Present Address: Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Guido Poli
- San Raffaele Scientific Institute, School of Medicine, Milan, Italy. .,Vita-Salute San Raffaele University, School of Medicine, Milan, Italy. .,Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Schalk W van der Merwe
- MRC Unit for Inflammation and Immunity, Department of Immunology and the Tshwane Academic Division of the National Health Laboratory Service, University of Pretoria, Pretoria, South Africa. .,Department of Internal Medicine, Division of Liver and Biliopancreatic Disorders, University of Leuven, Leuven, Belgium.
| |
Collapse
|
29
|
Sattler A, Dang-Heine C, Reinke P, Babel N. IL-15 dependent induction of IL-18 secretion as a feedback mechanism controlling human MAIT-cell effector functions. Eur J Immunol 2015; 45:2286-98. [DOI: 10.1002/eji.201445313] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 04/09/2015] [Accepted: 06/03/2015] [Indexed: 02/04/2023]
Affiliation(s)
- Arne Sattler
- Department of Immunology; Berlin-Brandenburg-Center for Regenerative Therapies (BCRT); Berlin Germany
| | - Chantip Dang-Heine
- Department of Immunology; Berlin-Brandenburg-Center for Regenerative Therapies (BCRT); Berlin Germany
| | - Petra Reinke
- Department of Immunology; Berlin-Brandenburg-Center for Regenerative Therapies (BCRT); Berlin Germany
- Department of Nephrology and Internal Intensive Care; Charite University Hospital; Berlin Germany
| | - Nina Babel
- Department of Immunology; Berlin-Brandenburg-Center for Regenerative Therapies (BCRT); Berlin Germany
- Medical Clinic I, Marien Hospital Herne; Ruhr University Bochum; Germany
| |
Collapse
|
30
|
Abstract
: Inflammasomes are multiprotein complexes that process procytokines into mature forms of interleukin 1β and interleukin 18 and induce pyroptotic cell death. Evidence linking NLRP3, NLRC4, and NLRP6 inflammasomes to intestinal inflammation is reviewed to provide a basis to understand how the innate immune system discriminates pathogenic bacteria from commensal bacteria and shapes microbial ecology. Inflammasomes have a direct and important role limiting colitis by directing effective immune responses against pathogenic bacterial infections in the intestine. Chronic granulomatous disease is presented to reveal a contrasting proinflammatory effect of inflammasomes. This pathogenic effect is unmasked in a state of immunodeficiency where bacterial growth is poorly controlled increasing inflammasome activity. The role of inflammasomes in inflammation associated with Crohn's disease and ulcerative colitis is discussed. Finally, mechanistic studies linking genetic polymorphisms in ATG16L and NOD2 to inflammasome activation provide a basis for new hypotheses to explain how genetic polymorphism associated with Crohn's disease modulate intestinal inflammation. A deeper understanding of the role of inflammasomes in intestinal inflammation is expected to identify new ways of treating inflammatory bowel disease.
Collapse
|
31
|
Guo W, Ye P, Yu H, Liu Z, Yang P, Hunter N. CD24 activates the NLRP3 inflammasome through c-Src kinase activity in a model of the lining epithelium of inflamed periodontal tissues. IMMUNITY INFLAMMATION AND DISEASE 2014; 2:239-53. [PMID: 25866631 PMCID: PMC4386918 DOI: 10.1002/iid3.40] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/10/2014] [Accepted: 10/10/2014] [Indexed: 01/17/2023]
Abstract
Chronic periodontitis is characterized by perturbation of the epithelial attachment to the tooth with subsequent migration of the lining epithelium and formation of a cleft or pocket. This non-keratinized lining epithelium provides initial responses to bacterial products by signalling through receptors of innate immunity to activate inflammasome pathways. These comprise an intracellular network of regulatory and effector molecules leading to synthesis and activation of pro-inflammatory cytokines. Conversely, CD24 is characteristically strongly expressed by the pocket epithelium and is reported to function as an important negative regulator for danger signals, protecting tissues from excessive leukocyte activity. The objective of the study was to determine the impact of ligation of CD24 on expression of inflammasome components. An epithelial mimic of pocket epithelium was used to evaluate activation of the NLRP3 inflammasome pathway. Surprisingly, antibody ligation of CD24 enhanced expression of NLRP3 together with co-activators ASC and caspase-1 resulting in burst release of activated interleukin (IL)-18. Potent product inhibition was detected with IL-18 suppressing expression of NLRP3, ASC, and caspase-1. Scant distribution of these products within pocket epithelium compared with healthy gingival attachment provided indication of potential cycling of NLRP3 inflammasome expression. As subjects with mild chronic periodontitis have increased titres of serum antibodies auto-reactive with CD24 compared with those of subjects with severe periodontitis, a molecular mechanism for regulated expression of the NLRP3 inflammasome mediated by c-Src kinase activity, is proposed. This pathway could be regionally disrupted by products of pathogenic bacteria with profound downregulation in the dysbiosis associated with severe disease.
Collapse
Affiliation(s)
- Wei Guo
- Department of Periodontology, School of Dentistry, Key Laboratory of Oral Biomedicine, Shandong University Shandong Province, China ; Yantai Stomatological Hospital Shandong Province, China
| | - Ping Ye
- Institute of Dental Research, Westmead Millennium Institute and Westmead Centre for Oral Health Westmead Hospital, Australia
| | - Hong Yu
- Microscopy Laboratory, Westmead Millennium Institute Westmead Hospital, Australia
| | - Zhonghao Liu
- Yantai Stomatological Hospital Shandong Province, China
| | - Pishan Yang
- Department of Periodontology, School of Dentistry, Key Laboratory of Oral Biomedicine, Shandong University Shandong Province, China
| | - Neil Hunter
- Institute of Dental Research, Westmead Millennium Institute and Westmead Centre for Oral Health Westmead Hospital, Australia ; Faculty of Dentistry, the University of Sydney Sydney, Australia
| |
Collapse
|
32
|
The relationship between interleukin-18 polymorphisms and allergic disease: a meta-analysis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:290687. [PMID: 24995282 PMCID: PMC4066680 DOI: 10.1155/2014/290687] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 05/05/2014] [Accepted: 05/05/2014] [Indexed: 12/21/2022]
Abstract
Recent studies have suggested that IL-18 −607C/A and −137G/C polymorphisms may be associated with the risk of allergic disease; however, individually published results are inconclusive. Therefore, we performed a meta-analysis to clarify whether IL-18 −607C/A and −137G/C polymorphisms were associated with the risk of allergic disease. A total of 21 studies including 5,331 cases and 9,658 controls were involved in this meta-analysis. In the overall analysis and the subgroup analysis according to ethnicity, we did not find significant association between IL-18 −607C/A or −137G/C polymorphism and the risk of allergic disease (all P > 0.05). However, in a stratified analysis by type of allergic disease, our results indicated that IL-18 −607C/A polymorphism was associated with a significantly decreased risk of allergic asthma in heterozygous comparison and IL-18 −137G/C was associated with a significantly decreased risk of allergic dermatitis in recessive model and homozygous comparison. In the stratified analysis by source of control, IL-18−607C/A showed significantly reduced risk in population-based subgroup, and for IL-18 −137G/C only significantly decreased risk was found in the hospital-based subgroup. Our meta-analysis suggests that IL-18 −607C/A and −137G/C polymorphisms may be protective factors for the risk of allergic asthma and allergic dermatitis, respectively.
Collapse
|
33
|
Abstract
: Inflammatory bowel disease accounts for significant patient morbidity in the Western world. Several immunosuppressive therapies are available but are associated with potential significant adverse effects. In addition, there remains a cohort of patients with refractory or relapsing disease. Therefore, the search for novel therapeutic agents continues. In this review, we evaluate the role of a number of designated cytokines that are candidates in the pathogenesis of inflammatory bowel disease and discuss how their manipulation has been explored as a therapeutic strategy for this disease. The interleukins (ILs) chosen for discussion reflect those that currently show most promise as future therapeutic targets, as well as discussing the role of some of the most recently identified ILs, such as IL-27, IL-33, IL-35, and IL-22, in this context.
Collapse
|
34
|
Lopetuso LR, Chowdhry S, Pizarro TT. Opposing Functions of Classic and Novel IL-1 Family Members in Gut Health and Disease. Front Immunol 2013; 4:181. [PMID: 23847622 PMCID: PMC3705591 DOI: 10.3389/fimmu.2013.00181] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 06/24/2013] [Indexed: 12/17/2022] Open
Abstract
In addition to their well-established role(s) in the pathogenesis of gastrointestinal (GI)-related inflammatory disorders, including inflammatory bowel disease (IBD) and inflammation-associated colorectal cancer (CRC), emerging evidence confirms the critical involvement of the interleukin-1 (IL-1) cytokine family and their ligands in the maintenance of normal gut homeostasis. In fact, the paradigm that IBD occurs in two distinct phases is substantiated by the observation that classic IL-1 family members, such as IL-1, the IL-1 receptor antagonist (IL-1Ra), and IL-18, possess dichotomous functions depending on the phase of disease, as well as on their role in initiating vs. sustaining chronic gut inflammation. Another recently characterized IL-1 family member, IL-33, also possesses dual functions in the gut. IL-33 is upregulated in IBD and potently induces Th2 immune responses, while also amplifying Th1-mediated inflammation. Neutralization studies in acute colitis models, however, have yielded controversial results and recent reports suggest a protective role of IL-33 in epithelial regeneration and mucosal wound healing. Finally, although little is currently known regarding the potential contribution of IL-36 family members in GI inflammation/homeostasis, another IL-1 family member, IL-37, is emerging as a potent anti-inflammatory cytokine with the ability to down-regulate colitis. This new body of information has important translational implications for both the prevention and treatment of patients suffering from IBD and inflammation-associated CRC.
Collapse
Affiliation(s)
- Loris R Lopetuso
- Department of Pathology, Case Western Reserve University School of Medicine , Cleveland, OH , USA ; Internal Medicine, Gastroenterology Division, Catholic University of Rome , Rome , Italy
| | | | | |
Collapse
|
35
|
Yoneno K, Hisamatsu T, Shimamura K, Kamada N, Ichikawa R, Kitazume MT, Mori M, Uo M, Namikawa Y, Matsuoka K, Sato T, Koganei K, Sugita A, Kanai T, Hibi T. TGR5 signalling inhibits the production of pro-inflammatory cytokines by in vitro differentiated inflammatory and intestinal macrophages in Crohn's disease. Immunology 2013; 139:19-29. [PMID: 23566200 PMCID: PMC3634536 DOI: 10.1111/imm.12045] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Revised: 10/30/2012] [Accepted: 11/05/2012] [Indexed: 12/13/2022] Open
Abstract
Bile acids (BAs) play important roles not only in lipid metabolism, but also in signal transduction. TGR5, a transmembrane receptor of BAs, is an immunomodulative factor, but its detailed mechanism remains unclear. Here, we aimed to delineate how BAs operate in immunological responses via the TGR5 pathway in human mononuclear cell lineages. We examined TGR5 expression in human peripheral blood monocytes, several types of in vitro differentiated macrophages (Mϕs) and dendritic cells. Mϕs differentiated with macrophage colony-stimulating factor and interferon-γ (Mγ-Mϕs), which are similar to the human intestinal lamina propria CD14+ Mϕs that contribute to Crohn's disease (CD) pathogenesis by production of pro-inflammatory cytokines, highly expressed TGR5 compared with any other type of differentiated Mϕ and dendritic cells. We also showed that a TGR5 agonist and two types of BAs, deoxycholic acid and lithocholic acid, could inhibit tumour necrosis factor-α production in Mγ-Mϕs stimulated by commensal bacterial antigen or lipopolysaccharide. This inhibitory effect was mediated by the TGR5–cAMP pathway to induce phosphorylation of c-Fos that regulated nuclear factor-κB p65 activation. Next, we analysed TGR5 levels in lamina propria mononuclear cells (LPMCs) obtained from the intestinal mucosa of patients with CD. Compared with non-inflammatory bowel disease, inflamed CD LPMCs contained more TGR5 transcripts. Among LPMCs, isolated CD14+ intestinal Mϕs from patients with CD expressed TGR5. In isolated intestinal CD14+ Mϕs, a TGR5 agonist could inhibit tumour necrosis factor-α production. These results indicate that TGR5 signalling may have the potential to modulate immune responses in inflammatory bowel disease.
Collapse
Affiliation(s)
- Kazuaki Yoneno
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bilsborough J, Viney JL. From model to mechanism: lessons of mice and men in the discovery of protein biologicals for the treatment of inflammatory bowel disease. Expert Opin Drug Discov 2013; 1:69-83. [PMID: 23506033 DOI: 10.1517/17460441.1.1.69] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Successful therapeutics for inflammatory bowel disease (IBD) must be able to reverse effectively the complex processes involved in the manifestation of inflammatory pathology in intact tissues. Although studies of human tissue samples are important to confirm the biological rationale for developing a particular therapeutic, in vivo rodent models of IBD provide a biological 'flask' in which therapeutics can be tested in a more representative setting. Moreover, gene targeting and transgenic technologies in rodents have exponentially increased the repertoire of available IBD models and provided insight into possible contributions that certain genes may have in the pathogenesis of disease. These models have been key in generating the current arsenal of biological therapeutics that are available, or are presently under investigation, for the treatment of IBD patients.
Collapse
Affiliation(s)
- Janine Bilsborough
- ZymoGenetics, Inc., Department of Autoimmunity and Inflammation, 1201 Eastlake Avenue East, Seattle, WA 98102, USA
| | | |
Collapse
|
37
|
Elinav E, Henao-Mejia J, Flavell RA. Integrative inflammasome activity in the regulation of intestinal mucosal immune responses. Mucosal Immunol 2013; 6:4-13. [PMID: 23212196 DOI: 10.1038/mi.2012.115] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The mammalian intestinal tract harbors a vast and diverse ecosystem of microbes that are separated from the sterile host milieu by a single layer of epithelial cells. While this bio-geographical configuration is critical for host biological processes, it imposes a risk for microbial penetration and life-threatening systemic invasion. Inflammasomes are cytosolic multi-protein platforms that sense both microbial and damage-associated molecular patterns and initiate a potent innate immune anti-microbial response. In this review, we will highlight the role of inflammasomes in the orchestration and regulation of the intestinal immune response, focusing on the roles of inflammasomes in maintenance of intestinal homeostasis, enteric infection, auto-inflammation, and tumorigenesis. We highlight the centrality of inflammasome signaling in the complex cross-talk between host mucosal immune arms and the environment, in particular the microflora, with emphasis on the spatial and temporal integration of inflammasome activation with signals from other innate signaling platforms.
Collapse
Affiliation(s)
- E Elinav
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | |
Collapse
|
38
|
Struck D, Frank I, Enders S, Steinhoff U, Schmidt C, Stallmach A, Liesenfeld O, Heimesaat MM. Treatment with interleukin-18 binding protein ameliorates Toxoplasma gondii-induced small intestinal pathology that is induced by bone marrow cell-derived interleukin-18. Eur J Microbiol Immunol (Bp) 2012; 2:249-57. [PMID: 24688772 DOI: 10.1556/eujmi.2.2012.3.11] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 06/29/2012] [Indexed: 01/08/2023] Open
Abstract
Peroral infection with Toxoplasma gondii results in a Th1-type immunopathology characterized by small intestinal necrosis and is dependent on IL-18. In the present study, we investigated whether treatment with IL-18 binding protein (IL-18bp) prevents ileal pathology. We observed increased expression of IL-18bp in intestinal biopsies of mice following infection. Whereas small intestines of control mice showed severe necrosis with complete destruction of the small intestinal architecture, mice treated with IL-18bp daily displayed only mild inflammatory changes including flattening of villi and edema in the space between the epithelium and lamina propria. Small intestinal parasite loads and concentrations of pro-inflammatory cytokines did not differ in control and IL-18bp-treated mice. Binding of IL-18 to immobilized IL-18bp revealed a remarkably slow dissociation rate, indicating high affinity. Using chimeric mice we observed that bone marrow-derived rather than stromal cells were the primary source of IL-18 that resulted in small intestinal pathology following peroral infection with T. gondii. In conclusion, the results presented here suggest that IL-18bp may be an effective and safe treatment for small intestinal inflammation. Antigen-presenting rather than epithelial cells appear to be the main source of IL-18 in T. gondii-induced small intestinal inflammation.
Collapse
|
39
|
Peters CD, Jespersen B, Nørregaard R. AlphaLISA versus ELISA-based detection of interleukin 18 in healthy subjects and patients with end-stage renal disease. Scandinavian Journal of Clinical and Laboratory Investigation 2012; 72:583-92. [DOI: 10.3109/00365513.2012.713175] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
40
|
Lapointe TK, Buret AG. Interleukin-18 facilitates neutrophil transmigration via myosin light chain kinase-dependent disruption of occludin, without altering epithelial permeability. Am J Physiol Gastrointest Liver Physiol 2012; 302:G343-51. [PMID: 22135309 DOI: 10.1152/ajpgi.00202.2011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Compromised epithelial barrier function and tight junction alterations are hallmarks of a number of gastrointestinal disorders, including inflammatory bowel disease (IBD). Increased levels of IL-18 have been observed in mucosal samples from Crohn's disease and ulcerative colitis patients. Remarkably, several reports have demonstrated that immunological or genetic blockage of IL-18 ameliorates the severity of colitis in multiple in vivo models of IBD. Nevertheless, the effects of IL-18 on intestinal epithelial barrier function remain unclear. We hypothesized that IL-18 could disrupt intestinal epithelial barrier structure and function, thus contributing to tissue damage in the context of IBD. The aims of the present study were to determine the effects of IL-18 on epithelial barrier structure and function and to characterize the mechanisms involved in these modulatory properties. Human colonic epithelial Caco-2 monolayers were coincubated with IL-18 for 24 h and processed for immunocytochemistry, immunoblotting, quantitative PCR, and permeability measurements (transepithelial resistance, FITC-dextran fluxes, and bacterial translocation). Our findings indicate that IL-18 selectively disrupts tight junctional occludin, without affecting the distribution pattern of claudin-4, claudin-5, zonula occludens-1, or E-cadherin. This effect coincided with a significant increase in myosin light chain kinase (MLCK) protein levels and activity. Pharmacological inhibition of MLCK and NF-κB prevented IL-18-induced loss of occludin. Although too subtle to alter paracellular permeability, these fine changes correlated with an MLCK-dependent increase in neutrophil transepithelial migration. In conclusion, our data suggest that IL-18 may potentiate inflammation in the context of IBD by facilitating neutrophil transepithelial migration via MLCK-dependent disruption of tight junctional occludin.
Collapse
Affiliation(s)
- Tamia K Lapointe
- Dept. of Biological Sciences, Univ. of Calgary, Calgary, AB, Canada
| | | |
Collapse
|
41
|
Figueredo CM, Brito F, Barros FC, Menegat JSB, Pedreira RR, Fischer RG, Gustafsson A. Expression of cytokines in the gingival crevicular fluid and serum from patients with inflammatory bowel disease and untreated chronic periodontitis. J Periodontal Res 2011; 46:141-6. [DOI: 10.1111/j.1600-0765.2010.01303.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
42
|
Abstract
Over recent years it has become increasingly clear that innate immune responses can shape the adaptive immune response. Among the most potent molecules of the innate immune system are the interleukin-1 (IL-1) family members. These evolutionarily ancient cytokines are made by and act on innate immune cells to influence their survival and function. In addition, they act directly on lymphocytes to reinforce certain adaptive immune responses. This Review provides an overview of both the long-established and more recently characterized members of the IL-1 family. In addition to their effects on immune cells, their involvement in human disease and disease models is discussed.
Collapse
Affiliation(s)
- John E Sims
- Amgen, 1201 Amgen Court West, Seattle, Washington 98119, USA.
| | | |
Collapse
|
43
|
Sakuraba A, Sato T, Kamada N, Kitazume M, Sugita A, Hibi T. Th1/Th17 immune response is induced by mesenteric lymph node dendritic cells in Crohn's disease. Gastroenterology 2009; 137:1736-45. [PMID: 19632232 DOI: 10.1053/j.gastro.2009.07.049] [Citation(s) in RCA: 181] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Revised: 06/24/2009] [Accepted: 07/16/2009] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Dendritic cells (DCs) possess the most potent ability to induce acquired immunity. However, their involvement in the pathogenesis of Crohn's disease (CD) has not yet been determined. We aimed to establish the immune status of mesenteric lymph nodes, the major gut-associated lymphoid tissue, and isolated DCs and determine their involvement in the pathogenesis of CD. METHODS CD4(+) T cells and DCs were isolated from mesenteric lymph nodes of CD, ulcerative colitis, and normal control. The immune status of CD4(+) T cells was analyzed by cytokine production and transcriptional profile. Surface phenotype of DCs was analyzed by flow cytometry. Cytokine production by myeloid DCs was analyzed by real-time polymerase chain reaction and exogenous bacterial stimulation. Immune stimulating activity of DCs was determined by mixed lymphocyte reaction. RESULTS In CD, mesenteric lymph node CD4(+) T cells produced higher amounts of interferon-gamma and interleukin (IL)-17 compared with ulcerative colitis and normal control, and this was dictated by increased T-bet and retinoic acid-related orphan receptor-gamma expression. Three subtypes of DCs, myeloid DC, plasmacytoid DC, and mature DC, were identified in all groups. When stimulated with exogenous bacterial derivative, myeloid DCs from CD produced a higher amount of IL-23 and a lower amount of IL-10. Myeloid DCs from CD induced stronger T helper cell (Th)1 immune response in mixed lymphocyte reaction compared with those from ulcerative colitis and normal control. CONCLUSIONS Our findings revealed that mesenteric lymph node is the key pathogenic location of CD elicited by the unique cytokine milieu produced by DCs leading to a dysregulated Th1/Th17 immune response.
Collapse
Affiliation(s)
- Atsushi Sakuraba
- Division of Gastroenterology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Okamoto M, Azuma K, Hoshino T, Imaoka H, Ikeda J, Kinoshita T, Takamori S, Ohshima K, Edakuni N, Kato S, Iwanaga T, Aizawa H. Correlation of decreased survival and IL-18 in bone metastasis. Intern Med 2009; 48:763-73. [PMID: 19443970 DOI: 10.2169/internalmedicine.48.1851] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Previous studies have reported that serum IL-18 levels are increased in some cancers. We investigated whether IL-18 production is increased in sera and cancer cells of patients with non-small cell lung cancer (NSCLC). PATIENTS OR MATERIALS Serum levels of IFN-gamma and IL-18 and thioredoxin 1 (TRX1) were measured in 79 patients (51 males, 28 females, median age 67 years) with advanced NSCLC (57 adenocarcinoma, 22 squamous cell carcinoma; TNM stages IIIA [n=11], IIIB [n=24], and IV [n=44]) and 75 healthy age-matched controls (44 males, 31 females, median age 65 years) by enzyme-linked immunosorbent assay. We examined IL-18 production in the lungs and sites of bone metastasis of adenocarcinoma by immunohistochemistry. RESULTS Serum IL-18, IFN-gamma, and TRX1 levels in NSCLC patients were significantly (p<0.0001, p=0.0031, and p<0.0001, respectively) higher than in control subjects, while serum IFN-gamma levels in NSCLC were slightly increased. Serum IL-18, but not IFN-gamma or TRX1, levels were significantly (p=0.0102) and negatively associated with overall survival in NSCLC. The serum IL-18 level was identified as an independent prognostic factor for overall survival in multivariate survival analysis. Moreover, serum IL-18 levels were significantly (p=0.049) higher in NSCLC with bone metastasis than in NSCLC without bone metastasis. Based on immunohistochemistry, we observed that cancer cells in the lungs and bone metastases markedly produced IL-18. CONCLUSION Our results suggest that elevated serum IL-18 levels may be associated with IL-18 producing cancer cells in advanced NSCLC.
Collapse
Affiliation(s)
- Masaki Okamoto
- Division of Respirology, Neurology and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Inflammatory bowel disease (IBD) is closely associated with the abnormal immune response of intestinal immune system to enterobacteria and dietary antigens. Large amounts of mononuclear lymphocyte infiltrates and high expression levels of proinflammatory cytokines and costimulatory molecules are present in inflamed mucosa. NK cells, involved in innate and acquired immune response, have cytolitic activities through secretion of lytic proteins, and produce proinflammatory mediators. Increased infiltration of NK cells present in inflamed mucosa of IBD expresses high levels of activated molecules and proinflammatory cytokines, and plays a role in intestinal mucosal damage.
Collapse
|
46
|
Kamada N, Hisamatsu T, Okamoto S, Chinen H, Kobayashi T, Sato T, Sakuraba A, Kitazume MT, Sugita A, Koganei K, Akagawa KS, Hibi T. Unique CD14 intestinal macrophages contribute to the pathogenesis of Crohn disease via IL-23/IFN-gamma axis. J Clin Invest 2008; 118:2269-80. [PMID: 18497880 DOI: 10.1172/jci34610] [Citation(s) in RCA: 324] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Accepted: 04/09/2008] [Indexed: 02/06/2023] Open
Abstract
Intestinal macrophages play a central role in regulation of immune responses against commensal bacteria. In general, intestinal macrophages lack the expression of innate-immune receptor CD14 and do not produce proinflammatory cytokines against commensal bacteria. In this study, we identified what we believe to be a unique macrophage subset in human intestine. This subset expressed both macrophage (CD14, CD33, CD68) and DC markers (CD205, CD209) and produced larger amounts of proinflammatory cytokines, such as IL-23, TNF-alpha, and IL-6, than typical intestinal resident macrophages (CD14-CD33+ macrophages). In patients with Crohn disease (CD), the number of these CD14+ macrophages were significantly increased compared with normal control subjects. In addition to increased numbers of cells, these cells also produced larger amounts of IL-23 and TNF-alpha compared with those in normal controls or patients with ulcerative colitis. In addition, the CD14+ macrophages contributed to IFN-gamma production rather than IL-17 production by lamina propria mononuclear cells (LPMCs) dependent on IL-23 and TNF-alpha. Furthermore, the IFN-gamma produced by LPMCs triggered further abnormal macrophage differentiation with an IL-23-hyperproducing phenotype. Collectively, these data suggest that this IL-23/IFN-gamma-positive feedback loop induced by abnormal intestinal macrophages contributes to the pathogenesis of chronic intestinal inflammation in patients with CD.
Collapse
Affiliation(s)
- Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ebert EC, Jabri B. Massive interleukin-12-induced interferon-gamma production by interleukin-15-stimulated lamina propria lymphocytes followed by down-regulation of the interleukin-12 receptor. Immunology 2008; 124:453-60. [PMID: 18540964 DOI: 10.1111/j.1365-2567.2007.02796.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The intestinal mucosal immune response must differentiate between harmless foreign antigens and pathogens, a distinction that may depend upon changes in the cytokine milieu. A key cytokine in the adaptive immune response is interleukin-12 (IL-12), secreted by antigen-presenting cells (APC) immediately after encounter with a pathogen. IL-12 is important in the priming and polarization of naïve T cells. Here, we show that IL-12 and IL-15 direct human intestinal lamina propria lymphocytes (LPL) in the absence of T-cell receptor engagement to secrete extremely high amounts of interferon-gamma (IFN-gamma), greater than with any other stimulus. The functional synergy of IL-12 with IL-15 surprisingly operates independently of signal transducer and activator of transcription 1 (STAT1), STAT3, STAT4, or STAT5 phosphorylation and occurs during transcription. Four-colour immunofluorescence showed that IL-12 receptor beta1 is found on the CD4+ T cells expressing intracytoplasmic IFN-gamma. Importantly, IL-12 receptors beta1 and beta2 are not up-regulated by IL-12, unlike findings using antigen-specific T cells, and are lost over time. This study demonstrates the early and massive IFN-gamma response of LPL to IL-12 and IL-15, providing the tools to deal with a pathogen. The down-regulation of IL-12 receptors may curtail any excess damaging inflammation.
Collapse
Affiliation(s)
- Ellen C Ebert
- UMDNJ-Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA.
| | | |
Collapse
|
48
|
Genetic analysis of innate immunity in Crohn's disease and ulcerative colitis identifies two susceptibility loci harboring CARD9 and IL18RAP. Am J Hum Genet 2008; 82:1202-10. [PMID: 18439550 DOI: 10.1016/j.ajhg.2008.03.016] [Citation(s) in RCA: 199] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 03/26/2008] [Accepted: 03/31/2008] [Indexed: 12/18/2022] Open
Abstract
The two main phenotypes of inflammatory bowel disease (IBD)--Crohn's disease (CD) and ulcerative colitis (UC)--are chronic intestinal inflammatory disorders with a complex genetic background. Using a three-stage design, we performed a functional candidate-gene analysis of innate immune pathway in IBD. In phase I, we typed 354 SNPs from 85 innate immunity genes in 520 Dutch IBD patients (284 CD, 236 UC) and 808 controls. In phase II, ten autosomal SNPs showing association at p < 0.006 in phase I were replicated in a second cohort of 545 IBD patients (326 CD, 219 UC) and 360 controls. In phase III, four SNPs with p < 0.01 in the combined phase I and phase II analysis were genotyped in an additional 786 IBD samples (452 CD, 334 UC) and 768 independent controls. Joint analysis of 1851 IBD patients (1062 CD, 789 UC) and 1936 controls demonstrated strong association to the IL18RAP rs917997 SNP for both CD and UC (p(IBD) 1.9 x 10(-8); OR 1.35). Association in CD is independently supported by the Crohn's disease dataset of the Wellcome Trust Case Control Consortium (imputed SNP rs917997, p = 9.19 x 10(-4)). In addition, an association of the CARD9 rs10870077 SNP to CD and UC was observed (p(IBD) = 3.25 x 10(-5); OR 1.21). Both genes are located in extended haplotype blocks on 2q11-2q12 and 9q34.3, respectively. Our results indicate two IBD loci and further support the importance of the innate immune system in the predisposition to both CD and UC.
Collapse
|
49
|
Lee CC, Lin WY, Wan L, Tsai Y, Tsai CH, Huang CM, Chen CP, Tsai FJ. Association of interleukin-18 gene polymorphism with asthma in Chinese patients. J Clin Lab Anal 2008; 22:39-44. [PMID: 18200581 DOI: 10.1002/jcla.20218] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Like other allergic diseases, asthma results from multiple conditions. Asthmatic beginning and severity are mediated by both environmental and genetic factors. In asthma studies, important work is realization of the genetic background and identification of genetic factors resulting in asthma development and phenomena. Here, we investigated whether interleukin (IL)-18 single nucleotide polymorphisms (SNPs) are involved in Chinese asthma patients. IL-18 (IL-18) SNP was detected by polymerase chain reaction (PCR)-based restriction analysis in 201 patients with asthma and 60 normal controls. Significant differences were found in the genotype distribution of IL-18 SNP between asthma patients and controls (P=0.000003). Allelic frequency of the IL-18 gene distinguished asthma patients from controls (P=0.000066). The results revealed a significant difference between asthma patients and normal controls in IL-18 SNP and a statistical correlation between IL-18 polymorphisms (105A/C) and asthma formation. We concluded that Chinese who carry the C/C homozygote of the IL-18-105A/C gene polymorphism in coding regions may have a higher risk of developing asthma.
Collapse
Affiliation(s)
- Cheng-Chun Lee
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Vigorous immune responses are induced in the immune privileged CNS by injury and disease, but the molecular mechanisms regulating innate immunity in the CNS are poorly defined. The inflammatory response initiated by spinal cord injury (SCI) involves activation of interleukin-1beta (IL-1beta) that contributes to secondary cell death. In the peripheral immune response, the inflammasome activates caspase-1 to process proinflammatory cytokines, but the regulation of trauma-induced inflammation in the CNS is not clearly understood. Here we show that a molecular platform [NALP1 (NAcht leucine-rich-repeat protein 1) inflammasome] consisting of caspase-1, caspase-11, ASC (apoptosis-associated speck-like protein containing a caspase-activating recruitment domain), and NALP1 is expressed in neurons of the normal rat spinal cord and forms a protein assembly with the X-linked inhibitor of apoptosis protein (XIAP). Moderate cervical contusive SCI induced processing of IL-1beta, IL-18, activation of caspase-1, cleavage of XIAP, and promoted assembly of the multiprotein complex. Anti-ASC neutralizing antibodies administered to injured rats entered spinal cord neurons via a mechanism that was sensitive to carbenoxolone. Therapeutic neutralization of ASC reduced caspase-1 activation, XIAP cleavage, and interleukin processing, resulting in significant tissue sparing and functional improvement. Thus, rat spinal cord neurons contain a caspase-1, pro-ILbeta, and pro-IL-18 activating complex different from the human NALP1 inflammasome that constitutes an important arm of the innate CNS inflammatory response after SCI.
Collapse
|