201
|
Sun M, He C, Cong Y, Liu Z. Regulatory immune cells in regulation of intestinal inflammatory response to microbiota. Mucosal Immunol 2015; 8:969-978. [PMID: 26080708 PMCID: PMC4540654 DOI: 10.1038/mi.2015.49] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 05/04/2015] [Indexed: 02/07/2023]
Abstract
The intestinal lumen harbors nearly 100 trillion commensal bacteria that exert crucial function for health. An elaborate balance between immune responses and tolerance to intestinal microbiota is required to maintain intestinal homeostasis. This process depends on diverse regulatory mechanisms, including both innate and adaptive immunity. Dysregulation of the homeostasis between intestinal immune systems and microbiota has been shown to be associated with the development of inflammatory bowel diseases (IBD) in genetically susceptible populations. In this review, we discuss the recent progress reported in studies of distinct types of regulatory immune cells in the gut, including intestinal intraepithelial lymphocytes, Foxp3(+) regulatory T cells, regulatory B cells, alternatively activated macrophages, dendritic cells, and innate lymphoid cells, and how dysfunction of this immune regulatory system contributes to intestinal diseases such as IBD. Moreover, we discuss the manipulation of these regulatory immune cells as a potential therapeutic method for management of intestinal inflammatory disorders.
Collapse
Affiliation(s)
- M Sun
- Department of Gastroenterology, Institute for Intestinal Diseases, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - C He
- Department of Gastroenterology, Institute for Intestinal Diseases, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Y Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Z Liu
- Department of Gastroenterology, Institute for Intestinal Diseases, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
202
|
Pasztoi M, Pezoldt J, Huehn J. Microenvironment Matters: Unique Conditions Within Gut-Draining Lymph Nodes Favor Efficient De Novo Induction of Regulatory T Cells. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 136:35-56. [PMID: 26615091 DOI: 10.1016/bs.pmbts.2015.07.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The gastrointestinal tract constitutes the largest surface of the body and thus has developed multitude mechanisms to either prevent pathogen entry or to efficiently eliminate invading pathogens. At the same time, the gastrointestinal system has to avoid unwanted immune responses against self and harmless nonself antigens, such as nutrients and commensal microbiota. Therefore, it is somewhat not unexpected that the gastrointestinal mucosa serves as the largest repository of immune cells throughout the body, harboring both potent pro- as well as anti-inflammatory properties. One additional key element of this regulatory machinery is created by trillions of symbiotic commensal bacteria in the gut. The microbiota not only simply contribute to the breakdown of nutrients, but are essential in limiting the expansion of pathogens, directing the development of the intestinal immune system, and establishing mucosal tolerance by fostering the induction of regulatory T cells (Tregs). In this review, we will discuss our current understanding about the microenvironmental factors fostering the de novo generation of Tregs within the gastrointestinal immune system, focusing on unique properties of antigen-presenting cells, tolerogenic cytokines, commensal-derived metabolites and the contribution of lymph node stromal cells.
Collapse
Affiliation(s)
- Maria Pasztoi
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Joern Pezoldt
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.
| |
Collapse
|
203
|
Wang S, An W, Yao Y, Chen R, Zheng X, Yang W, Zhao Y, Hu X, Jiang E, Bie Y, Chen Z, Ouyang P, Zhang H, Xiong H. Interleukin 37 Expression Inhibits STAT3 to Suppress the Proliferation and Invasion of Human Cervical Cancer Cells. J Cancer 2015; 6:962-9. [PMID: 26316892 PMCID: PMC4543756 DOI: 10.7150/jca.12266] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 07/20/2015] [Indexed: 01/05/2023] Open
Abstract
Objectives: The most recently discovered cytokine interleukin 37 (IL-37) received growing attention. Its function on tumor is largely unknown. Here, we investigated the biological function of IL-37 on cervical cancer (CC). Materials and methods: HPV+ Hela cells and HPV- C33A cells were used. RT-qPCR was performed to detect the transcription of IL-37, STAT3, TNF-αand IL-1β. Western blotting was used for protein detection. CCK-8 assay and transwell assay were employed for cell proliferation and invasion detection, respectively. Results: Successful gene transfection of IL-37 suppressed the proliferation and invasion of CC. Interestingly, IL-37 showed higher anticancer ability in HPV+ Hela cells than that in HPV- C33A cells. Then, the molecular mechanism of IL-37 anticancer was explored. Firstly, we found that IL-37 inhibited STAT3 expression at both mRNA and protein levels. IL-37 also down regulated the phosphorylation of STAT3. Secondly, blockage of STAT3 using siRNAs reduced significantly the ability of IL-37 to suppress cell proliferation and invasion. Thirdly, STAT3 knockdown reduced markedly the inhibition of IL-37 on the transcription of tumor-derived TNF-α and IL-1β, indicating the contribution of STAT3 for the cancer associated antiinflammation of IL-37. Finally, STAT3 up regulation restored the ability of cell proliferation, cell invasion and the expression of inflammatory cytokines, TNF-α and IL-1β. Conclusions: IL-37 suppressed cell proliferation and invasion of CC and STAT3 is involved in this process. Thus, IL-37 emerges as a new anticancer cytokine for CC. This study demonstrated a new biological function of IL-37 and offered a potential molecule for CC treatment.
Collapse
Affiliation(s)
- Sen Wang
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China
| | - Weifang An
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China
| | - Yunhong Yao
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China
| | - Renhuai Chen
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China
| | - Xiaoxuan Zheng
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China; ; 2. Clinical Laboratory, Guanghua hospital, Dongguan 523808, China
| | - Wanyong Yang
- 2. Clinical Laboratory, Guanghua hospital, Dongguan 523808, China
| | - Yi Zhao
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China
| | - Xinrong Hu
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China
| | - Enping Jiang
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China
| | - Yanhong Bie
- 3. Pathology, People's Hospital of Xiaolan, Zhongshan 528415, China
| | - Zhangquan Chen
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China
| | - Ping Ouyang
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China
| | - He Zhang
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China
| | - Hui Xiong
- 1. Cancer Institute, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
204
|
Wu W, Wang W, Wang Y, Li W, Yu G, Li Z, Fang C, Shen Y, Sun Z, Han L, Yu J, Fang L, Chen S, Dong K, Han Z, Liu H, Luo Y, Feng X. IL-37b suppresses T cell priming by modulating dendritic cell maturation and cytokine production via dampening ERK/NF-κB/S6K signalings. Acta Biochim Biophys Sin (Shanghai) 2015; 47:597-603. [PMID: 26094142 DOI: 10.1093/abbs/gmv058] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/16/2015] [Indexed: 12/20/2022] Open
Abstract
Interleukin 37b (IL-37b) plays a key role in suppressing immune responses, partially by modulating the function of dendritic cells (DCs). However, the precise mechanisms are still largely unknown. Here, we investigated the effects of IL-37b on DC maturation and T cell responses induced by DCs, and explored the involved signaling pathways. It was found that IL-37b down-regulated the expressions of co-stimulatory molecules CD80 and CD86 on DCs in vitro. At the same time, the expressions of pro-inflammatory cytokines, such as TNF-α and IL-6, were suppressed, while the expression of the T cell inhibitory cytokine TGF-β was increased in IL-37b-treated DCs. In addition, the activation effect of DCs on T cells was impaired by IL-37b. We further revealed that extracellular single-regulated kinase (ERK), nuclear factor-κB (NF-κB), and mTOR-S6K signaling pathways were involved in the inhibition of DCs induced by IL-37b. This was confirmed by the similarly suppressive effect of chemical inhibitors against NF-κB, ERK, and S6K on the expressions of IL-6 and TNF-α in DCs. In conclusion, these results demonstrated that IL-37b suppressed DC maturation and immunostimulatory capacity in T cell priming by involving in ERK, NF-κB, and S6K-based inhibitory signaling pathways.
Collapse
Affiliation(s)
- Wantong Wu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China Tianjin Dongli Hospital, Tianjin 300300, China
| | - Weiqiang Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Yun Wang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Wenwen Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Gang Yu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Zhonglong Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Chunmin Fang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Yue Shen
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Zhina Sun
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Ling Han
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Juan Yu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Lijun Fang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Song Chen
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Kui Dong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Zhongchao Han
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Hanzhi Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Yuechen Luo
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences, Tianjin 300020, China
| |
Collapse
|
205
|
Masterson JC, McNamee EN, Fillon SA, Hosford L, Harris R, Fernando SD, Jedlicka P, Iwamoto R, Jacobsen E, Protheroe C, Eltzschig HK, Colgan SP, Arita M, Lee JJ, Furuta GT. Eosinophil-mediated signalling attenuates inflammatory responses in experimental colitis. Gut 2015; 64:1236-47. [PMID: 25209655 PMCID: PMC4515997 DOI: 10.1136/gutjnl-2014-306998] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 08/19/2014] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Eosinophils reside in the colonic mucosa and increase significantly during disease. Although a number of studies have suggested that eosinophils contribute to the pathogenesis of GI inflammation, the expanding scope of eosinophil-mediated activities indicate that they also regulate local immune responses and modulate tissue inflammation. We sought to define the impact of eosinophils that respond to acute phases of colitis in mice. DESIGN Acute colitis was induced in mice by administration of dextran sulfate sodium, 2,4,6-trinitrobenzenesulfonic acid or oxazolone to C57BL/6J (control) or eosinophil deficient (PHIL) mice. Eosinophils were also depleted from mice using antibodies against interleukin (IL)-5 or by grafting bone marrow from PHIL mice into control mice. Colon tissues were collected and analysed by immunohistochemistry, flow cytometry and reverse transcription PCR; lipids were analysed by mass spectroscopy. RESULTS Eosinophil-deficient mice developed significantly more severe colitis, and their colon tissues contained a greater number of neutrophils, than controls. This compensatory increase in neutrophils was accompanied by increased levels of the chemokines CXCL1 and CXCL2, which attract neutrophils. Lipidomic analyses of colonic tissue from eosinophil-deficient mice identified a deficiency in the docosahexaenoic acid-derived anti-inflammatory mediator 10, 17- dihydroxydocosahexaenoic acid (diHDoHE), namely protectin D1 (PD1). Administration of an exogenous PD1-isomer (10S, 17S-DiHDoHE) reduced the severity of colitis in eosinophil-deficient mice. The PD1-isomer also attenuated neutrophil infiltration and reduced levels of tumour necrosis factor-α, IL-1β, IL-6 and inducible NO-synthase in colons of mice. Finally, in vitro assays identified a direct inhibitory effect of PD1-isomer on neutrophil transepithelial migration. CONCLUSIONS Eosinophils exert a protective effect in acute mouse colitis, via production of anti-inflammatory lipid mediators.
Collapse
Affiliation(s)
- Joanne C Masterson
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics; Digestive Health Institute, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA,Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado, USA,University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Eóin N McNamee
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado, USA,University of Colorado School of Medicine, Aurora, Colorado, USA,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Sophie A Fillon
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics; Digestive Health Institute, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA,Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado, USA,University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Lindsay Hosford
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics; Digestive Health Institute, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA,Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado, USA,University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Rachel Harris
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics; Digestive Health Institute, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA,Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado, USA,University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Shahan D Fernando
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics; Digestive Health Institute, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA,Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado, USA,University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Paul Jedlicka
- University of Colorado School of Medicine, Aurora, Colorado, USA,Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ryo Iwamoto
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Elizabeth Jacobsen
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan,Department of Biochemistry and Molecular Biology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Cheryl Protheroe
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan,Department of Biochemistry and Molecular Biology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Holger K Eltzschig
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado, USA,University of Colorado School of Medicine, Aurora, Colorado, USA,Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Sean P Colgan
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado, USA,University of Colorado School of Medicine, Aurora, Colorado, USA,Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Makoto Arita
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan,Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - James J Lee
- Department of Biochemistry and Molecular Biology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Glenn T Furuta
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics; Digestive Health Institute, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA,Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado, USA,University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
206
|
Deciphering the crosstalk among IL-1 and IL-10 family cytokines in intestinal immunity. Trends Immunol 2015; 36:471-8. [PMID: 26184648 DOI: 10.1016/j.it.2015.06.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 06/18/2015] [Accepted: 06/19/2015] [Indexed: 12/16/2022]
Abstract
The IL-1 and IL-10 family cytokines are important regulators of intestinal immunity. Whereas these cytokines have protective roles in response to mucosal damage or infection, they also contribute to pathology in certain settings. How these cytokines function to maintain intestinal homoeostasis, and under what circumstances they contribute to disease is poorly understood. Recent studies have revealed a multi-layered regulatory network wherein IL-1 and IL-10 family cytokines impact each other's production. The workings of this network vary in different intestinal regions, reflecting the influence of resident microbiota and the distribution of distinct immune cell populations in different regions of the intestine. We review these findings here, and discuss them in the context of the current understanding of the functions of these cytokine families in health and disease. We further highlight important areas of future investigation.
Collapse
|
207
|
Palomo J, Dietrich D, Martin P, Palmer G, Gabay C. The interleukin (IL)-1 cytokine family--Balance between agonists and antagonists in inflammatory diseases. Cytokine 2015; 76:25-37. [PMID: 26185894 DOI: 10.1016/j.cyto.2015.06.017] [Citation(s) in RCA: 326] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 06/29/2015] [Indexed: 12/14/2022]
Abstract
The interleukin (IL)-1 family of cytokines comprises 11 members, including 7 pro-inflammatory agonists (IL-1α, IL-1β, IL-18, IL-33, IL-36α, IL-36β, IL-36γ) and 4 defined or putative antagonists (IL-1R antagonist (IL-1Ra), IL-36Ra, IL-37, and IL-38) exerting anti-inflammatory activities. Except for IL-1Ra, IL-1 cytokines do not possess a leader sequence and are secreted via an unconventional pathway. In addition, IL-1β and IL-18 are produced as biologically inert pro-peptides that require cleavage by caspase-1 in their N-terminal region to generate active proteins. N-terminal processing is also required for full activity of IL-36 cytokines. The IL-1 receptor (IL-1R) family comprises 10 members and includes cytokine-specific receptors, co-receptors and inhibitory receptors. The signaling IL-1Rs share a common structure with three extracellular immunoglobulin (Ig) domains and an intracellular Toll-like/IL-1R (TIR) domain. IL-1 cytokines bind to their specific receptor, which leads to the recruitment of a co-receptor and intracellular signaling. IL-1 cytokines induce potent inflammatory responses and their activity is tightly controlled at the level of production, protein processing and maturation, receptor binding and post-receptor signaling by naturally occurring inhibitors. Some of these inhibitors are IL-1 family antagonists, while others are IL-1R family members acting as membrane-bound or soluble decoy receptors. An imbalance between agonist and antagonist levels can lead to exaggerated inflammatory responses. Several genetic modifications or mutations associated with dysregulated IL-1 activity and autoinflammatory disorders were identified in mouse models and in patients. These findings paved the road to the successful use of IL-1 inhibitors in diseases that were previously considered as untreatable.
Collapse
Affiliation(s)
- Jennifer Palomo
- Division of Rheumatology, Departments of Internal Medicine Specialties and of Pathology-Immunology, University of Geneva School of Medicine, Switzerland
| | - Damien Dietrich
- Division of Rheumatology, Departments of Internal Medicine Specialties and of Pathology-Immunology, University of Geneva School of Medicine, Switzerland
| | - Praxedis Martin
- Division of Rheumatology, Departments of Internal Medicine Specialties and of Pathology-Immunology, University of Geneva School of Medicine, Switzerland
| | - Gaby Palmer
- Division of Rheumatology, Departments of Internal Medicine Specialties and of Pathology-Immunology, University of Geneva School of Medicine, Switzerland
| | - Cem Gabay
- Division of Rheumatology, Departments of Internal Medicine Specialties and of Pathology-Immunology, University of Geneva School of Medicine, Switzerland.
| |
Collapse
|
208
|
Chai M, Ji Q, Zhang H, Zhou Y, Yang Q, Zhou Y, Guo G, Liu W, Han W, Yang L, Zhang L, Liang J, Liu Y, Shi D, Zhao Y. The Protective Effect of Interleukin-37 on Vascular Calcification and Atherosclerosis in Apolipoprotein E-Deficient Mice with Diabetes. J Interferon Cytokine Res 2015; 35:530-9. [PMID: 25866993 DOI: 10.1089/jir.2014.0212] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Meng Chai
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| | - Qingwei Ji
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| | - Haitao Zhang
- Department of Cardiology, General Hospital of the Air Force, Beijing, China
| | - Yujie Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| | - Qing Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| | - Yangwei Zhou
- School of General Medicine and Continuing Education, Capital Medical University, Beijing, China
| | - Guangjin Guo
- Department of Cardiology, General Hospital of the Air Force, Beijing, China
| | - Wei Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| | - Wei Han
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| | - Lixia Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| | - Linlin Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| | - Jing Liang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| | - Yuyang Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| | - Dongmei Shi
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| | - Yingxin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Institute of Heart, Lung and Blood Vessel Disease, Ministry of Education, Beijing, China
| |
Collapse
|
209
|
Fonseca-Camarillo G, Furuzawa-Carballeda J, Yamamoto-Furusho JK. Interleukin 35 (IL-35) and IL-37: Intestinal and peripheral expression by T and B regulatory cells in patients with Inflammatory Bowel Disease. Cytokine 2015; 75:389-402. [PMID: 26141420 DOI: 10.1016/j.cyto.2015.04.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/24/2015] [Accepted: 04/16/2015] [Indexed: 12/11/2022]
Abstract
The aim of the study was to characterize and to quantify peripheral and tissue. IL-35- and IL-37-producing cells in Inflammatory Bowel Disease (IBD) patients. We studied a total of 38 active UC, 31 inactive UC, 17 active CD, and 13 inactive CD and 50 non-inflamed tissues as control group. Gene expression was measured by real time polymerase chain reaction (RT-PCR) and protein expression was evaluated in tissue by immunohistochemistry and in peripheral blood mononuclear cells by flow cytometry. Higher levels of IL-35 was produced by intestinal regulatory B cells and circulating regulatory CD4(+) and CD8(+) T cells in active vs. inactive disease or healthy donors (P<0.05). The IL-37 was conspicuously synthesized by circulating B cells, active natural killer cells and monocytes. These results suggest that down-regulation of inflammation in active IBD patients might be based on the increased expression of IL-35 and IL-37.
Collapse
Affiliation(s)
- Gabriela Fonseca-Camarillo
- Inflammatory Bowel Disease Clinic, Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, Vasco de Quiroga No. 15, Col. Sección XVI, CP 14000 Mexico City, Mexico
| | - Janette Furuzawa-Carballeda
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, Vasco de Quiroga No. 15, Col. Sección XVI, CP 14000 Mexico City, Mexico
| | - Jesús K Yamamoto-Furusho
- Inflammatory Bowel Disease Clinic, Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, Vasco de Quiroga No. 15, Col. Sección XVI, CP 14000 Mexico City, Mexico.
| |
Collapse
|
210
|
Afonina I, Müller C, Martin S, Beyaert R. Proteolytic Processing of Interleukin-1 Family Cytokines: Variations on a Common Theme. Immunity 2015; 42:991-1004. [DOI: 10.1016/j.immuni.2015.06.003] [Citation(s) in RCA: 369] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Indexed: 12/22/2022]
|
211
|
Højen JF, Rasmussen TA, Andersen KLD, Winckelmann AA, Laursen RR, Gunst JD, Møller HJ, Fujita M, Østergaard L, Søgaard OS, Dinarello CA, Tolstrup M. Interleukin-37 Expression Is Increased in Chronic HIV-1-Infected Individuals and Is Associated with Inflammation and the Size of the Total Viral Reservoir. Mol Med 2015; 21:337-45. [PMID: 25879630 DOI: 10.2119/molmed.2015.00031] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/14/2015] [Indexed: 12/11/2022] Open
Abstract
Interleukin-37 (IL-37) is a recently identified cytokine with potent antiinflammatory and immunosuppressive functions. The objective of this study was to compare levels of IL-37 mRNA in immunological subgroups of chronic human immunodeficiency virus-1 (HIV-1)-infected individuals and noninfected controls, to determine IL-37's association with biomarkers of inflammation and reservoir size. This was a cross-sectional study. The HIV-1-infected patients were categorized in three subgroups depending on their combination antiretroviral therapy (cART) treatment status and CD4(+) T-cell count. Quantitative RT-PCR was used for the detection of IL-37 mRNA and HIV-1 DNA in peripheral blood mononuclear cells (PBMCs). Biomarkers in plasma were quantified by enzyme-linked immunosorbent assay (ELISA), whereas T-cell activation was determined by flow cytometry. Lastly, lipopolysaccharide (LPS) stimulations of patients PBMCs were carried out to determine differences in IL-37 mRNA response between the subgroups. Sixty HIV-1-infected patients and 20 noninfected controls were included in the study. Steady-state IL-37 mRNA levels in PBMCs were significantly higher in HIV-1-infected individuals compared with noninfected controls: 2.4-fold (p ≤ 0.01) cART-naïve subjects; 3.9-fold (p ≤ 0.0001) inadequate immunological responders; and 4.0-fold (p ≤ 0.0001) in immunological responders compared with non-infected controls. Additionally, levels of the monocyte inflammatory marker sCD14 correlated with IL-37 mRNA (p = 0.03), whereas there was no association with T-cell activation. Finally, we observed a significant correlation between total viral HIV-1 DNA and IL-37 mRNA in PBMCs (p < 0.0001). Collectively, our data shows that the level of IL-37 mRNA is affected by chronic HIV-1-infection. A relationship with the activation of the monocyte compartment is suggested by the correlation with sCD14 and, interestingly, IL-37 could be related to the size of the total viral HIV-1 reservoir.
Collapse
Affiliation(s)
- Jesper F Højen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas A Rasmussen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | | | - Anni A Winckelmann
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Rune R Laursen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Jesper D Gunst
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Holger J Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Mayumi Fujita
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Ole S Søgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Charles A Dinarello
- Division of Infectious Diseases, University of Colorado Denver, Aurora, Colorado, United States of America.,Department of Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
212
|
Ye Z, Wang C, Tang J, Zhou Y, Bai L, Liu Y, Kijlstra A, Yang P. Decreased Interleukin-37 Expression in Vogt-Koyanagi-Harada Disease and Upregulation Following Immunosuppressive Treatment. J Interferon Cytokine Res 2015; 35:265-72. [PMID: 25343528 DOI: 10.1089/jir.2014.0042] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Zi Ye
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Chaokui Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Jihong Tang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Yan Zhou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Lin Bai
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Yunjia Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, People's Republic of China
| | - Aize Kijlstra
- University Eye Clinic Maastricht, Maastricht, The Netherlands
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, People's Republic of China
| |
Collapse
|
213
|
Lunding L, Webering S, Vock C, Schröder A, Raedler D, Schaub B, Fehrenbach H, Wegmann M. IL-37 requires IL-18Rα and SIGIRR/IL-1R8 to diminish allergic airway inflammation in mice. Allergy 2015; 70:366-73. [PMID: 25557042 DOI: 10.1111/all.12566] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND Interleukin (IL) 37 has been described as a negative regulator of innate immunity, as it reduces the activation and cytokine production of different innate immune cells. Recently, results from the CLARA childhood asthma cohort suggested an implication of IL-37 for human asthma pathogenesis. This study aimed to investigate the effects of IL-37 on allergic airway inflammation in a mouse model of experimental asthma. METHODS Peripheral blood mononuclear cells (PBMCs) of children were cultured for 48 h (anti-CD3/anti-CD28 stimulation or unstimulated), and IL-37 concentrations in supernatants were determined. Wild-type, IL-18Rα-deficient ((-/-) ), and SIGIRR(-/-) C57BL/6 mice were sensitized to ovalbumin (OVA) and challenged with OVA aerosol to induce acute experimental asthma, and IL-37 was applied intranasally prior to each OVA challenge. Airway hyper-responsiveness (AHR), airway inflammation, cytokine levels in broncho-alveolar lavage fluid, and mucus production were determined. RESULTS IL-37 production of human PBMCs was significantly lower in allergic asthmatics vs healthy children. In wild-type mice, intranasal administration of IL-37 ablated allergic airway inflammation as well as cytokine production and subsequently diminished the hallmarks of experimental asthma including mucus hyperproduction and AHR. In contrast, local application of IL-37 produced none of these effects in mice lacking either IL18Rα or SIGIRR/IL-1R8. CONCLUSIONS This study demonstrates that IL-37 is able to ablate a TH2 cell-directed allergic inflammatory response and the hallmarks of experimental asthma in mice, suggesting that IL-37 may be critical for asthma pathogenesis. Furthermore, these data suggest a mode of action of IL-37 that involves IL18Rα as well as the orphan receptor SIGIRR/IL-1R8.
Collapse
Affiliation(s)
- L. Lunding
- Division of Asthma Mouse Models; Priority Area Asthma & Allergy, Research Center Borstel; Airway Research Center North; Member of the German Center for Lung Research; Borstel Germany
| | - S. Webering
- Division of Experimental Pneumology; Priority Area Asthma & Allergy, Research Center Borstel; Airway Research Center North; Member of the German Center for Lung Research; Borstel Germany
| | - C. Vock
- Division of Experimental Pneumology; Priority Area Asthma & Allergy, Research Center Borstel; Airway Research Center North; Member of the German Center for Lung Research; Borstel Germany
| | - A. Schröder
- Division of Asthma Mouse Models; Priority Area Asthma & Allergy, Research Center Borstel; Airway Research Center North; Member of the German Center for Lung Research; Borstel Germany
| | - D. Raedler
- Department of Pulmonary & Allergy; LMU Munich, Comprehensive Pneumology Center-Munich; Member of the German Center for Lung Research; University Children's Hospital Munich; Munich Germany
| | - B. Schaub
- Department of Pulmonary & Allergy; LMU Munich, Comprehensive Pneumology Center-Munich; Member of the German Center for Lung Research; University Children's Hospital Munich; Munich Germany
| | - H. Fehrenbach
- Division of Experimental Pneumology; Priority Area Asthma & Allergy, Research Center Borstel; Airway Research Center North; Member of the German Center for Lung Research; Borstel Germany
| | - M. Wegmann
- Division of Asthma Mouse Models; Priority Area Asthma & Allergy, Research Center Borstel; Airway Research Center North; Member of the German Center for Lung Research; Borstel Germany
| |
Collapse
|
214
|
Transfer of the IL-37b gene elicits anti-tumor responses in mice bearing 4T1 breast cancer. Acta Pharmacol Sin 2015; 36:528-34. [PMID: 25832432 DOI: 10.1038/aps.2015.3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/15/2014] [Indexed: 12/29/2022]
Abstract
AIM IL-37b has shown anti-cancer activities in addition to its anti-inflammatory properties. In this study, we investigated the effects of IL-37b on breast carcinoma growth in mice and to determine the involvement of T cell activation in the effects. METHODS IL-37b gene was transferred into mouse breast carcinoma cell line 4T1 (4T1-IL37b cells), the expression of secretory IL-37b by the cells was detected, and the effects of IL-37b expression on the cell proliferation in vitro was evaluated. After injection of 4T1 cells or 4T1-IL37b cells into immunocompetent BALB/c mice, immunodeficient BALB/c nude mice and NOD-SCID mice, the tumor growth and survival rate were measured. The proliferation of T cells in vitro was also detected. RESULTS IL-37b was detected in the supernatants of 4T1-IL37b cells with a concentration of 12.02 ± 0.875 ng/mL. IL-37b expression did not affect 4T1 cell proliferation in vitro. BALB/c mice inoculated with 4T1-IL37b cells showed significant retardation of tumor growth. BALB/c mice inoculated with both 4T1 cells and mitomycin C-treated 4T1-IL37b cells also showed significant retardation of tumor growth. But the anti-cancer activity of IL-37b was abrogated in BALB/c nude mice and NOD-SCID mice inoculated with 4T1-IL37b cells. Recombinant IL-37b slightly promoted CD4(+) T cell proliferation without affecting CD8(+) T cell proliferation. CONCLUSION IL-37b exerts anti-4T1 breast carcinoma effects in vivo by modulating the tumor microenvironment and influencing T cell activation.
Collapse
|
215
|
Nold-Petry CA, Lo CY, Rudloff I, Elgass KD, Li S, Gantier MP, Lotz-Havla AS, Gersting SW, Cho SX, Lao JC, Ellisdon AM, Rotter B, Azam T, Mangan NE, Rossello FJ, Whisstock JC, Bufler P, Garlanda C, Mantovani A, Dinarello CA, Nold MF. IL-37 requires the receptors IL-18Rα and IL-1R8 (SIGIRR) to carry out its multifaceted anti-inflammatory program upon innate signal transduction. Nat Immunol 2015; 16:354-65. [PMID: 25729923 DOI: 10.1038/ni.3103] [Citation(s) in RCA: 333] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 01/12/2015] [Indexed: 12/12/2022]
Abstract
Interleukin 37 (IL-37) and IL-1R8 (SIGIRR or TIR8) are anti-inflammatory orphan members of the IL-1 ligand family and IL-1 receptor family, respectively. Here we demonstrate formation and function of the endogenous ligand-receptor complex IL-37-IL-1R8-IL-18Rα. The tripartite complex assembled rapidly on the surface of peripheral blood mononuclear cells upon stimulation with lipopolysaccharide. Silencing of IL-1R8 or IL-18Rα impaired the anti-inflammatory activity of IL-37. Whereas mice with transgenic expression of IL-37 (IL-37tg mice) with intact IL-1R8 were protected from endotoxemia, IL-1R8-deficient IL-37tg mice were not. Proteomic and transcriptomic investigations revealed that IL-37 used IL-1R8 to harness the anti-inflammatory properties of the signaling molecules Mer, PTEN, STAT3 and p62(dok) and to inhibit the kinases Fyn and TAK1 and the transcription factor NF-κB, as well as mitogen-activated protein kinases. Furthermore, IL-37-IL-1R8 exerted a pseudo-starvational effect on the metabolic checkpoint kinase mTOR. IL-37 thus bound to IL-18Rα and exploited IL-1R8 to activate a multifaceted intracellular anti-inflammatory program.
Collapse
Affiliation(s)
- Claudia A Nold-Petry
- 1] Ritchie Centre, MIMR-PHI Institute of Medical Research, Melbourne, Victoria, Australia. [2] Department of Paediatrics, Monash University, Melbourne, Australia
| | - Camden Y Lo
- 1] Ritchie Centre, MIMR-PHI Institute of Medical Research, Melbourne, Victoria, Australia. [2] Monash Micro Imaging, Monash University, Melbourne, Australia
| | - Ina Rudloff
- 1] Ritchie Centre, MIMR-PHI Institute of Medical Research, Melbourne, Victoria, Australia. [2] Department of Paediatrics, Monash University, Melbourne, Australia
| | | | - Suzhao Li
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Michael P Gantier
- 1] Centre for Cancer Research, MIMR-PHI Institute of Medical Research, Melbourne, Victoria, Australia. [2] Department of Molecular and Translational Science, Monash University, Melbourne, Australia
| | | | - Søren W Gersting
- Molecular Pediatrics, Ludwig-Maximilians University, Munich, Germany
| | - Steven X Cho
- 1] Ritchie Centre, MIMR-PHI Institute of Medical Research, Melbourne, Victoria, Australia. [2] Department of Paediatrics, Monash University, Melbourne, Australia
| | - Jason C Lao
- 1] Ritchie Centre, MIMR-PHI Institute of Medical Research, Melbourne, Victoria, Australia. [2] Department of Paediatrics, Monash University, Melbourne, Australia
| | - Andrew M Ellisdon
- 1] Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia. [2] Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, Australia
| | | | - Tania Azam
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Niamh E Mangan
- 1] Department of Molecular and Translational Science, Monash University, Melbourne, Australia. [2] Centre for Innate Immunity and Infectious Diseases, MIMR-PHI Institute of Medical Research, Melbourne, Australia
| | - Fernando J Rossello
- Victorian Bioinformatics Consortium, Monash University, Melbourne, Australia
| | - James C Whisstock
- 1] Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia. [2] Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, Australia
| | - Philip Bufler
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | | | - Alberto Mantovani
- 1] Humanitas Clinical and Research Center, Rozzano, Italy. [2] Humanitas University, Rozzano, Italy
| | - Charles A Dinarello
- 1] Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA. [2] Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Marcel F Nold
- 1] Ritchie Centre, MIMR-PHI Institute of Medical Research, Melbourne, Victoria, Australia. [2] Department of Paediatrics, Monash University, Melbourne, Australia. [3] Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
216
|
Chen HM, Fujita M. IL-37: A new player in immune tolerance. Cytokine 2015; 72:113-4. [DOI: 10.1016/j.cyto.2014.11.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 11/24/2014] [Indexed: 01/03/2023]
|
217
|
Extracellular forms of IL-37 inhibit innate inflammation in vitro and in vivo but require the IL-1 family decoy receptor IL-1R8. Proc Natl Acad Sci U S A 2015; 112:2497-502. [PMID: 25654981 DOI: 10.1073/pnas.1424626112] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Similar to IL-1α and IL-33, IL-1 family member IL-37b translocates to the nucleus and is associated with suppression of innate and adaptive immunity. Here we demonstrate an extracellular function of the IL-37 precursor and a processed form. Recombinant IL-37 precursor reduced LPS-induced IL-6 by 50% (P < 0.001) in highly inflammatory human blood-derived M1 differentiated macrophages derived from selective subjects but not M2 macrophages. In contrast, a neutralizing monoclonal anti-IL-37 increased LPS-induced IL-6, TNFα and IL-1β (P < 0.01). The suppression by IL-37 was consistently observed at low picomolar but not nanomolar concentrations. Whereas LPS induced a 12-fold increase in TNFα mRNA, IL-37 pretreatment decreased the expression to only 3-fold over background (P < 0.01). Mechanistically, LPS-induced p38 and pERK were reduced by IL-37. Recombinant IL-37 bound to the immobilized ligand binding α-chain of the IL-18 receptor as well as to the decoy receptor IL-1R8. In M1 macrophages, LPS increased the surface expression of IL-1R8. Compared with human blood monocytes, resting M1 cells express more surface IL-1R8 as well as total IL-1R8; there was a 16-fold increase in IL-1R8 mRNA levels when pretreated with IL-37. IL-37 reduced LPS-induced TNFα and IL-6 by 50-55% in mouse bone marrow-derived dendritic cells, but not in dendritic cells derived from IL-1R8-deficient mice. In mice subjected to systemic LPS-induced inflammation, pretreatment with IL-37 reduced circulating and organ cytokine levels. Thus, in addition to a nuclear function, IL-37 acts as an extracellular cytokine by binding to the IL-18 receptor but using the IL-1R8 for its anti-inflammatory properties.
Collapse
|
218
|
Yang Y, Zhang ZX, Lian D, Haig A, Bhattacharjee RN, Jevnikar AM. IL-37 inhibits IL-18-induced tubular epithelial cell expression of pro-inflammatory cytokines and renal ischemia-reperfusion injury. Kidney Int 2015; 87:396-408. [PMID: 25207880 DOI: 10.1038/ki.2014.295] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 06/24/2014] [Accepted: 06/26/2014] [Indexed: 12/21/2022]
Abstract
Cytokines and chemokines produced by tubular epithelial and infiltrating cells are critical to inflammation in renal ischemia-reperfusion injury. IL-37, a newly described IL-1 family member, inhibits IL-18-dependent pro-inflammatory cytokine production by its binding to IL-18 receptors and IL-18 binding protein. The potential role of IL-37 in renal ischemia-reperfusion injury is unknown. Here we found that exposure of tubular epithelial cells to exogenous IL-37 downregulated hypoxia and the IL-18-induced expression of TNFα, IL-6, and IL-1β. Importantly, human PT-2 tubular epithelial cells have inducible expression of IL-37. Moreover, pro-inflammatory cytokine expression was augmented in IL-37 mRNA-silenced tubular epithelial cells and inhibited by transfection with pCMV6-XL5-IL-37. In a mouse ischemic injury model, transgenic expression of human IL-37 inhibited kidney expression of TNFα, IL-6, and IL-1β and improved mononuclear cell infiltration, kidney injury, and function. Thus, human tubular epithelial cells express the IL-18 contra-regulatory protein IL-37 as an endogenous control mechanism to reduce inflammation. Augmenting kidney IL-37 may represent a novel strategy to suppress renal injury responses and promote kidney function after renal ischemic injury and transplantation.
Collapse
Affiliation(s)
- Yunbo Yang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Zhu-Xu Zhang
- 1] Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada [2] Department of Pathology, London Health Sciences, Western University, London, Ontario, Canada [3] Department of Medicine, London Health Sciences, Western University, London, Ontario, Canada
| | - Dameng Lian
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Aaron Haig
- Department of Pathology, London Health Sciences, Western University, London, Ontario, Canada
| | - Rabindra N Bhattacharjee
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Anthony M Jevnikar
- 1] Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada [2] Department of Medicine, London Health Sciences, Western University, London, Ontario, Canada
| |
Collapse
|
219
|
Kang B, Cheng S, Peng J, Yan J, Zhang S. Interleukin-37 gene variants segregated anciently coexist during hominid evolution. Eur J Hum Genet 2015; 23:1392-8. [PMID: 25626704 DOI: 10.1038/ejhg.2014.302] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 12/11/2014] [Accepted: 12/19/2014] [Indexed: 11/09/2022] Open
Abstract
IL37 is a member of IL-1 cytokine family but conveys anti-inflammatory functions. The biological characteristic and genetic heterogeneity of IL37 are not fully understood yet. Here using the whole-genome sequencing data from 1000 Genomes Project, we performed population and evolutionary genetic analysis of human IL37 gene. First, 2184 IL37 gene sequences from different human populations were retrieved. The IL37 protein sequences were inferred from the coding DNA sequences and multiple species alignment was made. Then, the phylogenetic tree of IL37 was built and dN/dS ratios were calculated for each evolutionary branch, the classic McDonald and Kreitman test was also performed. Next, we conducted intraspecific evolutionary genetic analysis and built the genealogy network of 116 unique IL37 haplotypes through median-joining network analysis. Finally, we compared IL37 sequences between the modern and archaic humans. Our results for the first time provide solid evidence that common IL37 variants other than NCBI reference sequence are present worldwide. Our data also supports that IL37 variants are shaped and maintained by selection instead of neutral evolution. We further identified that human IL37 variants consist of two major haplogroups and their presence in archaic humans corroborates its ancient origin in hominid evolution. In conclusion, these data indicate that common IL37 variants are maintained among human populations by selective force, suggesting their potential involvements in immune regulation and human diseases. In addition, the ancient history of IL37 variants reveals interesting insight into the complicated human evolutionary history.
Collapse
Affiliation(s)
- Bin Kang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shimeng Cheng
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jinbiao Peng
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jingjing Yan
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shuye Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology, Institute of Medical Microbiology, Shanghai Medical College of Fudan University, Shanghai, China
| |
Collapse
|
220
|
Chen B, Huang K, Ye L, Li Y, Zhang J, Zhang J, Fan X, Liu X, Li L, Sun J, Du J, Huang Z. Interleukin-37 is increased in ankylosing spondylitis patients and associated with disease activity. J Transl Med 2015; 13:36. [PMID: 25627863 PMCID: PMC4323018 DOI: 10.1186/s12967-015-0394-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 01/13/2015] [Indexed: 12/17/2022] Open
Abstract
Background Interleukin-37 (IL-37) has been known to play an immunosuppressive role in various inflammatory disorders, but whether it participates in the regulation of pathogenesis of ankylosing spondylitis (AS) has not been investigated. Here, we examined the serum levels of IL-37 and its clinical association in AS, and explored the anti-inflammatory effects of IL-37 on peripheral blood mononuclear cells (PBMCs) from AS patients. Methods The mRNA levels of IL-37, TNF-α, IL-6, IL-17, and IL-23 in PBMCs and their serum concentrations from 46 AS patients were examined by real-time polymerase chain reaction (RT-PCR) and enzyme-linked immunoassay (ELISA), respectively. The correlations between serum IL-37 levels with disease activity, laboratory values and pro-inflammatory cytokines in AS were analyzed by Spearman correlation test. PBMCs from 46 AS patients were stimulated with recombinant IL-37 protein, expressions of TNF-α, IL-6, IL-17 and IL-23 were determined by RT-PCR and ELISA. Results Compared to healthy controls (HC), AS patients and active AS patients showed higher levels of IL-37 in PBMCs and serum respectively. Strikingly, serum IL-37 levels were higher in AS patients with osteoporosis than those without. Serum levels of IL-37 were correlated with laboratory values as well as TNF-α, IL-6 and IL-17, but not IL-23 in patients with AS. The productions of pro-inflammatory cytokines such as TNF-α, IL-6, IL-17, IL-23 in PBMCs from AS patients were obviously attenuated after recombinant IL-37 stimulation, but not in the HC. Conclusion The higher levels of IL-37 were found in AS patients, which were correlated with disease activity and AS related pro-inflammatory cytokines. More importantly, IL-37 inhibits the expressions of the pro-inflammatory cytokines from PBMCs in AS patients, indicating the potential anti-inflammatory role of IL-37 in AS. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0394-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bingni Chen
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, 518060, Guangdong, China. .,Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China. .,Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, 518060, China.
| | - Kunzhao Huang
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, 518060, Guangdong, China. .,Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China. .,Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, 518060, China.
| | - Liang Ye
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, 518060, Guangdong, China. .,Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China. .,Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, 518060, China.
| | - Yanqun Li
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, 518060, Guangdong, China. .,Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China. .,Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, 518060, China.
| | - Jiawei Zhang
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, 518060, Guangdong, China. .,Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China. .,Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, 518060, China.
| | - Jinshun Zhang
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, 518060, Guangdong, China. .,Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China. .,Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, 518060, China.
| | - Xinmin Fan
- Department of pathology, Shenzhen University School of Medicine, Shenzhen, 518060, China.
| | - Xiaokai Liu
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, 518060, Guangdong, China. .,Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China. .,Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, 518060, China.
| | - Li Li
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, 518060, Guangdong, China. .,Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China. .,Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, 518060, China.
| | - Jinxia Sun
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, 518060, Guangdong, China. .,Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China. .,Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, 518060, China.
| | - Jing Du
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China.
| | - Zhong Huang
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, 518060, Guangdong, China. .,Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China. .,Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, 518060, China.
| |
Collapse
|
221
|
van de Veerdonk FL, Gresnigt MS, Oosting M, van der Meer JWM, Joosten LAB, Netea MG, Dinarello CA. Protective host defense against disseminated candidiasis is impaired in mice expressing human interleukin-37. Front Microbiol 2015; 5:762. [PMID: 25620965 PMCID: PMC4285810 DOI: 10.3389/fmicb.2014.00762] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/15/2014] [Indexed: 11/13/2022] Open
Abstract
The effect of the anti-inflammatory cytokine interleukin-37 (IL-37) on host defense against Candida infections remains unknown. We assessed the role of IL-37 in a murine model of disseminated candidiasis using mice transgenic for human IL-37 (hIL-37Tg). Upon exposure to Candida albicans pseudohyphae, macrophages from hIL-37Tg mice release 39% less TNFα compared to cells from wild-type (WT) mice (p = 0.01). In vivo, hIL-37Tg mice displayed a decreased capacity to recruit neutrophils to the site of infection. These defects were associated with increased mortality and organ fungal growth in hIL-37Tg compared to WT mice. We conclude that IL-37 interferes with the innate protective anti-Candida host response by reducing the production of proinflammatory cytokines and suppressing neutrophil recruitment in response to Candida, resulting in an increased susceptibility to disseminated candidiasis.
Collapse
Affiliation(s)
- Frank L van de Veerdonk
- Department of Medicine, University of Colorado Denver Denver, CO, USA ; Department of Medicine, Radboud University Nijmegen Medical Center Nijmegen, Netherlands ; Radboud Center for Infection Nijmegen, Netherlands
| | - Mark S Gresnigt
- Department of Medicine, Radboud University Nijmegen Medical Center Nijmegen, Netherlands
| | - Marije Oosting
- Department of Medicine, Radboud University Nijmegen Medical Center Nijmegen, Netherlands
| | - Jos W M van der Meer
- Department of Medicine, Radboud University Nijmegen Medical Center Nijmegen, Netherlands
| | - Leo A B Joosten
- Department of Medicine, Radboud University Nijmegen Medical Center Nijmegen, Netherlands ; Radboud Center for Infection Nijmegen, Netherlands
| | - Mihai G Netea
- Department of Medicine, Radboud University Nijmegen Medical Center Nijmegen, Netherlands ; Radboud Center for Infection Nijmegen, Netherlands
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver Denver, CO, USA ; Department of Medicine, Radboud University Nijmegen Medical Center Nijmegen, Netherlands
| |
Collapse
|
222
|
Gu J, Gao X, Pan X, Peng X, Li Y, Li M. High-level expression and one-step purification of a soluble recombinant human interleukin-37b in Escherichia coli. Protein Expr Purif 2015; 108:18-22. [PMID: 25559248 DOI: 10.1016/j.pep.2014.12.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 12/21/2014] [Accepted: 12/23/2014] [Indexed: 01/05/2023]
Abstract
Interleukin (IL)-37 is a novel member of the IL-1 cytokine family. However, as a result of lacking efficient method to generate relatively large quantity of IL-37, little is known of its functions in man. In the present study, the recombinant human IL-37b containing a C-hexahistidine tag was expressed in Escherichia coli (E. coli). The expression level of IL-37b in E. coli was very high after induction with IPTG. Furthermore, IL-37b protein was largely found in the soluble fraction. The expressed protein was readily purified by one-step immobilized metal-ion affinity chromatography using Ni(2+)-nitrilotriacetic acid agarose. The purified IL-37b appeared as a single band on SDS-PAGE and the purity was more than 97%. The yield was 90mg IL-37b from 1l of bacterial culture. Western blotting and N-terminal sequencing confirmed the identity of the purified protein. The purified IL-37b inhibited significantly the release of tumor necrosis factor-α and IL-1β in lipopolysaccharide-activated THP-1 cells. Thus, this method provides an efficient way to obtain an active IL-37 with high yield and high purity.
Collapse
Affiliation(s)
- Jiajie Gu
- Department of Immunology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo 315211, China
| | - Xueming Gao
- Department of Immunology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo 315211, China
| | - Xiuhe Pan
- Department of Immunology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo 315211, China
| | - Xiao Peng
- Department of Immunology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo 315211, China
| | - Yan Li
- Department of Histology and Embryology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo 315211, China.
| | - Mingcai Li
- Department of Immunology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo 315211, China.
| |
Collapse
|
223
|
Netea MG, van de Veerdonk FL, van der Meer JWM, Dinarello CA, Joosten LAB. Inflammasome-independent regulation of IL-1-family cytokines. Annu Rev Immunol 2014; 33:49-77. [PMID: 25493334 DOI: 10.1146/annurev-immunol-032414-112306] [Citation(s) in RCA: 260] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Induction, production, and release of proinflammatory cytokines are essential steps to establish an effective host defense. Cytokines of the interleukin-1 (IL-1) family induce inflammation and regulate T lymphocyte responses while also displaying homeostatic and metabolic activities. With the exception of the IL-1 receptor antagonist, all IL-1 family cytokines lack a signal peptide and require proteolytic processing into an active molecule. One such unique protease is caspase-1, which is activated by protein platforms called the inflammasomes. However, increasing evidence suggests that inflammasomes and caspase-1 are not the only mechanism for processing IL-1 cytokines. IL-1 cytokines are often released as precursors and require extracellular processing for activity. Here we review the inflammasome-independent enzymatic processes that are able to activate IL-1 cytokines, paying special attention to neutrophil-derived serine proteases, which subsequently induce inflammation and modulate host defense. The inflammasome-independent processing of IL-1 cytokines has important consequences for understanding inflammatory diseases, and it impacts the design of IL-1-based modulatory therapies.
Collapse
|
224
|
Wei F, Zhang Y, Jian J, Mundra JJ, Tian Q, Lin J, Lafaille JJ, Tang W, Zhao W, Yu X, Liu CJ. PGRN protects against colitis progression in mice in an IL-10 and TNFR2 dependent manner. Sci Rep 2014; 4:7023. [PMID: 25387791 PMCID: PMC4228332 DOI: 10.1038/srep07023] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/24/2014] [Indexed: 12/16/2022] Open
Abstract
This study was aimed to determine the role and regulation of progranulin (PGRN) in the pathogenesis of inflammatory bowel diseases (IBD). Dextran sulfate sodium (DSS)−, picrylsulfonic acid (TNBS)-induced, bone marrow chimera and CD4+CD45Rbhi T cell transfer colitis model were established and analyzed in wild-type and several genetically-modified mice, including PGRN, IL-10 and TNFR2 deficient mice. Elevated levels of PGRN were found in colitis samples from human IBD patients and mouse colitis models in comparison to the corresponding controls. PGRN-deficient mice became highly susceptible to DSS- and TNBS-induced colitis, whereas recombinant PGRN ameliorated the pathology and reduced the histological score in both DSS and TNBS colitis models. In addition, hematopoietic-derived PGRN was critical for protection against DSS-induced colitis, and lack of PGRN signaling in CD4+ T cells also exacerbated experimental colitis. PGRN-mediated protective effect in colitis was compromised in the absence of IL-10 signaling. In addition, PGRN's effect was also largely lost in the TNFR2-deficient colitis model. Collectively, these findings not only provide the new insight into PGRN's anti-inflammatory action in vivo, but may also present PGRN and its derivatives as novel biological agent for treating IBD.
Collapse
Affiliation(s)
- Fanhua Wei
- 1] Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003 [2] Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, 250012
| | - Yuying Zhang
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Jinlong Jian
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Jyoti Joshi Mundra
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Qingyun Tian
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Jiqiang Lin
- Department of Pathology, New York University Medical Center, New York, NY, 10016
| | - Juan Jose Lafaille
- Department of Pathology, New York University Medical Center, New York, NY, 10016
| | - Wei Tang
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, 250012
| | - Weiming Zhao
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, 250012
| | - Xiuping Yu
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, 250012
| | - Chuan-Ju Liu
- 1] Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003 [2] Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
225
|
IL-37 inhibits inflammasome activation and disease severity in murine aspergillosis. PLoS Pathog 2014; 10:e1004462. [PMID: 25375146 PMCID: PMC4223056 DOI: 10.1371/journal.ppat.1004462] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 09/10/2014] [Indexed: 12/19/2022] Open
Abstract
Since IL-37 transgenic mice possesses broad anti-inflammatory properties, we assessed whether recombinant IL-37 affects inflammation in a murine model of invasive pulmonary aspergillosis. Recombinant human IL-37 was injected intraperitoneally into mice prior to infection and the effects on lung inflammation and inflammasome activation were evaluated. IL-37 markedly reduced NLRP3-dependent neutrophil recruitment and steady state mRNA levels of IL-1β production and mitigated lung inflammation and damage in a relevant clinical model, namely aspergillosis in mice with cystic fibrosis. The anti-inflammatory activity of IL-37 requires the IL-1 family decoy receptor TIR-8/SIGIRR. Thus, by preventing activation of the NLRP3 inflammasome and reducing IL-1β secretion, IL-37 functions as a broad spectrum inhibitor of the innate response to infection-mediated inflammation, and could be considered to be therapeutic in reducing the pulmonary damage due to non-resolving Aspergillus infection and disease. IL-37, firstly identified by in silico research in the year 2000, is a member of the IL-1 family. The biological properties of IL-37 are mainly those of down-regulating inflammation in models of septic shock, chemical colitis, cardiac ischemia and contact dermatitis. Whether and how IL-37 down-regulates the inflammation of infection, and its consequences, is not known. We observed that IL-37 limits inflammation and disease severity in murine invasive aspergillosis, an infection model in which cytokines of the IL-1 family have important roles. However, given that IL-1R1-deficient or caspase 1-deficient mice are resistant to lung inflammation during infection and that IL-1 signaling could drive the differentiation of antifungal inflammatory Th17 cells, the pro-inflammatory properties of IL 1-induced inflammation in aspergillosis is potentially dangerous for the host. IL-37 markedly reduced NLRP3-dependent neutrophil recruitment and steady state mRNA levels of IL-1β production and mitigated lung inflammation and damage in a relevant clinical model, namely aspergillosis in mice with cystic fibrosis. The anti-inflammatory activity of IL-37 requires the IL-1 receptor family decoy TIR-8/SIGIRR. Thus, IL-37 functions as a broad spectrum inhibitor of infection-mediated inflammation, and could be considered to be therapeutic in reducing the pulmonary damage due to non-resolving Aspergillus infection and disease.
Collapse
|
226
|
Luo Y, Cai X, Liu S, Wang S, Nold-Petry CA, Nold MF, Bufler P, Norris D, Dinarello CA, Fujita M. Suppression of antigen-specific adaptive immunity by IL-37 via induction of tolerogenic dendritic cells. Proc Natl Acad Sci U S A 2014; 111:15178-83. [PMID: 25294929 PMCID: PMC4210310 DOI: 10.1073/pnas.1416714111] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
IL-1 family member IL-37 limits innate inflammation in models of colitis and LPS-induced shock, but a role in adaptive immunity remains unknown. Here, we studied mice expressing human IL-37b isoform (IL-37tg) subjected to skin contact hypersensitivity (CHS) to dinitrofluorobenzene. CHS challenge to the hapten was significantly decreased in IL-37tg mice compared with wild-type (WT) mice (-61%; P < 0.001 at 48 h). Skin dendritic cells (DCs) were present and migrated to lymph nodes after antigen uptake in IL-37tg mice. When hapten-sensitized DCs were adoptively transferred to WT mice, antigen challenge was greatly impaired in mice receiving DCs from IL-37tg mice compared with those receiving DCs from WT mice (-60%; P < 0.01 at 48 h). In DCs isolated from IL-37tg mice, LPS-induced increase of MHC II and costimulatory molecule CD40 was reduced by 51 and 31%, respectively. In these DCs, release of IL-1β, IL-6, and IL-12 was reduced whereas IL-10 secretion increased (37%). Consistent with these findings, DCs from IL-37tg mice exhibited a lower ability to stimulate syngeneic and allogeneic naive T cells as well as antigen-specific T cells and displayed enhanced induction of T regulatory (Treg) cells (86%; P < 0.001) in vitro. Histological analysis of CHS skin in mice receiving hapten-sensitized DCs from IL-37tg mice revealed a marked reduction in CD8(+) T cells (-74%) but an increase in Treg cells (2.6-fold). Together, these findings reveal that DCs expressing IL-37 are tolerogenic, thereby impairing activation of effector T-cell responses and inducing Treg cells. IL-37 thus emerges as an inhibitor of adaptive immunity.
Collapse
Affiliation(s)
| | - Xiangna Cai
- Departments of Dermatology and Department of Plastic and Reconstruct Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou City 515041, People's Republic of China
| | | | - Sen Wang
- Departments of Dermatology and Department of Plastic and Reconstruct Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou City 515041, People's Republic of China
| | - Claudia A Nold-Petry
- The Ritchie Centre, Monash Institute of Medical Research, Monash University, Melbourne, VIC 3800, Australia
| | - Marcel F Nold
- The Ritchie Centre, Monash Institute of Medical Research, Monash University, Melbourne, VIC 3800, Australia
| | - Philip Bufler
- Children's Hospital, Ludwig-Maximilians University, 80539 Munich, Germany; and
| | - David Norris
- Departments of Dermatology and Denver Veterans Affairs Medical Center, Denver, CO 80220
| | | | - Mayumi Fujita
- Departments of Dermatology and Denver Veterans Affairs Medical Center, Denver, CO 80220
| |
Collapse
|
227
|
Quirk S, Agrawal DK. Immunobiology of IL-37: mechanism of action and clinical perspectives. Expert Rev Clin Immunol 2014; 10:1703-9. [PMID: 25327443 DOI: 10.1586/1744666x.2014.971014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This article provides an overview of the biological function of a recently discovered cytokine, IL-37, formerly referred to as IL-1F7, and its role in chronic inflammation and autoimmune disease. Much has been discovered about IL-37 in the past decade, including its ability to down-regulate systemic and local inflammation by lowering levels of pro-inflammatory molecules. Here, we critically review the published reports. Future research is necessary to understand the receptor-dependent effects of IL-37, its intracellular and extracellular functions in both normal and diseased states and its potential role as a biomarker and pharmacological target in human disease.
Collapse
Affiliation(s)
- Shannon Quirk
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| | | |
Collapse
|
228
|
Role of cytokines and Toll-like receptors in the immunopathogenesis of Guillain-Barré syndrome. Mediators Inflamm 2014; 2014:758639. [PMID: 25614713 PMCID: PMC4189947 DOI: 10.1155/2014/758639] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 08/13/2014] [Indexed: 11/17/2022] Open
Abstract
Guillain-Barré syndrome (GBS) is an autoimmune disease of the peripheral nervous system, mostly triggered by an aberrant immune response to an infectious pathogen. Although several infections have been implicated in the pathogenesis of GBS, not all such infected individuals develop this disease. Moreover, infection with a single agent might also lead to different subtypes of GBS emphasizing the role of host factors in the development of GBS. The host factors regulate a broad range of inflammatory processes that are involved in the pathogenesis of autoimmune diseases including GBS. Evidences suggest that systemically and locally released cytokines and their involvement in immune-mediated demyelination and axonal damage of peripheral nerves are important in the pathogenesis of GBS. Toll-like receptors (TLRs) link innate and adaptive immunity through transcription of several proinflammatory cytokines. TLR genes may increase susceptibility to microbial infections; an attenuated immune response towards antigen and downregulation of cytokines occurs due to mutation in the gene. Herein, we discuss the crucial role of host factors such as cytokines and TLRs that activate the immune response and are involved in the pathogenesis of the disease.
Collapse
|
229
|
Li Y, Wang Z, Yu T, Chen B, Zhang J, Huang K, Huang Z. Increased expression of IL-37 in patients with Graves' disease and its contribution to suppression of proinflammatory cytokines production in peripheral blood mononuclear cells. PLoS One 2014; 9:e107183. [PMID: 25226272 PMCID: PMC4165889 DOI: 10.1371/journal.pone.0107183] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 08/07/2014] [Indexed: 12/22/2022] Open
Abstract
Background Intreleukin-37 (IL-37), a member of IL-1 family, is primarily an anti-inflammatory cytokine, which reduces systemic and local inflammation. However, the expression and role of IL-37 in Graves' disease (GD) remains unknown. This study aims to measure the levels of serum and peripheral blood mononuclear cells (PBMCs) IL-37 in patients with Graves' disease and to examine its association with disease activity. Furthermore, we investigate the effect of IL-37 on proinflammatory cytokines involved in the pathogenesis of GD. Methods The expressions of IL-37, TNF-α, IL-6, and IL-17 mRNA in peripheral blood mononuclear cells (PBMCs) of 40 patients with Graves' disease were determined by real-time reverse transcription-polymerase chain reaction (RT-PCR), and the levels of IL-37, TNF-α, IL-6, and IL-17 in serum were detected by enzyme-linked immunoassay (ELISA). The correlation of serum IL-37 levels with cytokines and disease activity in Graves' disease patients were investigated. The expressions of cytokines TNF-α, IL-6, and IL-17 in PBMCs under recombinant IL-37 stimulation were determined by RT-PCR and ELISA respectively. Results The levels of IL-37, TNF-α, IL-6, and IL-17 in PBMCs and serum were significantly increased in patients with GD compared with healthy controls (HC). Serum IL-37 were closely correlated with TNF-α, IL-6, IL-17, thyrotropin (TSH), free thyroxine (FT4),free triiodothyronine (FT3) and thyrotropin receptor antibody (TRAB). GD patients with active disease showed higher IL-37 mRNA and serum protein levels compared with those with inactive disease as well as HC. Moreover, IL-37 suppressed the production of IL-6, IL-17 and TNF-α in PBMCs of patients with GD. Conclusions Increased level of IL-37 in patients with GD are associated with TNF-α, IL-6, IL-17 and disease activity, and it plays a protective role against inflammatory effect in GD by inhibiting the production of proinflammatory cytokines. Thus, IL-37 may provide a novel research target for the pathogenesis and therapy of GD.
Collapse
Affiliation(s)
- Yanqun Li
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, China
- Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, China
| | - Zi Wang
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, China
- Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, China
| | - Ting Yu
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, China
- Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, China
| | - Bingni Chen
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, China
- Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, China
| | - Jinshun Zhang
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, China
- Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, China
| | - Kunzhao Huang
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, China
- Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, China
| | - Zhong Huang
- Biological therapy institute, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pathogen biology and immunology, Shenzhen University School of Medicine, Shenzhen, China
- Shenzhen City Shenzhen University Immunodiagnostic Technology Platforms, Shenzhen, China
- * E-mail:
| |
Collapse
|
230
|
Günaltay S, Nyhlin N, Kumawat AK, Tysk C, Bohr J, Hultgren O, Hörnquist EH. Differential expression of interleukin-1/Toll-like receptor signaling regulators in microscopic and ulcerative colitis. World J Gastroenterol 2014; 20:12249-12259. [PMID: 25232259 PMCID: PMC4161810 DOI: 10.3748/wjg.v20.i34.12249] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 05/05/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate Toll-like receptor (TLR) signaling regulators in microscopic and ulcerative colitis patients.
METHODS: Total RNA and microRNA were isolated from fresh frozen colonic biopsies of non-inflamed controls and patients with active or in-remission collagenous colitis (CC), lymphocytic colitis (LC), or ulcerative colitis (UC). We compared expressions of interleukin-1 receptor-associated kinase (IRAK)-2, IRAK-M, interleukin (IL)-37, microRNA (miR)-146a, miR-155, and miR-21 using quantitative real time reverse transcription polymerase chain reaction.
RESULTS: IRAK-M expression was increased in LC patients with active disease in histopathological remission (LC-HR; P = 0.02) and UC patients (P = 0.01), but no differences in IRAK-2 expression were detected compared to controls. miR-146a, -155 and -21 expressions were increased in LC-HR (P = 0.04, 0.07, and 0.004) and UC (P = 0.02, 0.04 and 0.03) patients. miR-146a and miR-21 expressions were significantly enhanced in UC patients compared to UC remission (UC-R; P = 0.01 and 0.04). Likewise, active CC patients showed significantly increased expression of miR-155 (P = 0.003) and miR-21 (P = 0.006). IL-37 expression was decreased in both CC (P = 0.03) and LC (P = 0.04) patients with a similar trend in UC patients but not statistically significant, whilst it was increased in UC-R patients compared to controls (P = 0.02) and active UC (P = 0.001).
CONCLUSION: The identification of differentially expressed miRNAs, IL-37, and IRAK-M suggests different pathophysiologic mechanisms in various disease stages in LC, CC, and UC.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biopsy
- Case-Control Studies
- Colitis, Collagenous/diagnosis
- Colitis, Collagenous/genetics
- Colitis, Collagenous/immunology
- Colitis, Lymphocytic/diagnosis
- Colitis, Lymphocytic/genetics
- Colitis, Lymphocytic/immunology
- Colitis, Ulcerative/diagnosis
- Colitis, Ulcerative/genetics
- Colitis, Ulcerative/immunology
- Colon/immunology
- Colon/pathology
- Female
- Humans
- Inflammation Mediators/analysis
- Interleukin-1/analysis
- Interleukin-1/metabolism
- Interleukin-1 Receptor-Associated Kinases/analysis
- Intestinal Mucosa/immunology
- Intestinal Mucosa/pathology
- Male
- MicroRNAs/analysis
- Middle Aged
- Signal Transduction
- Toll-Like Receptors/metabolism
Collapse
|
231
|
Cottle DL, Ursino GMA, Ip SCI, Jones LK, Ditommaso T, Hacking DF, Mangan NE, Mellett NA, Henley KJ, Sviridov D, Nold-Petry CA, Nold MF, Meikle PJ, Kile BT, Smyth IM. Fetal inhibition of inflammation improves disease phenotypes in harlequin ichthyosis. Hum Mol Genet 2014; 24:436-49. [PMID: 25209981 DOI: 10.1093/hmg/ddu459] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Harlequin ichthyosis (HI) is a severe skin disease which leads to neonatal death in ∼50% of cases. It is the result of mutations in ABCA12, a protein that transports lipids required to establish the protective skin barrier needed after birth. To better understand the life-threatening newborn HI phenotype, we analysed the developing epidermis for consequences of lipid dysregulation in mouse models. We observed a pro-inflammatory signature which was characterized by chemokine upregulation in embryonic skin which is distinct from that seen in other types of ichthyosis. Inflammation also persisted in grafted HI skin. To examine the contribution of inflammation to disease development, we overexpressed interleukin-37b to globally suppress fetal inflammation, observing considerable improvements in keratinocyte differentiation. These studies highlight inflammation as an unexpected contributor to HI disease development in utero, and suggest that inhibiting inflammation may reduce disease severity.
Collapse
Affiliation(s)
| | | | | | | | | | - Douglas F Hacking
- Department of Anaesthetics, Saint Vincent's Hospital Melbourne, 41 Victoria Parade, Fitzroy, VIC 3065, Australia Department of Paediatric Intensive Care, The Royal Children's Hospital, Melbourne, VIC, Australia
| | | | - Natalie A Mellett
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Katya J Henley
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC 3052, Australia
| | - Dmitri Sviridov
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Claudia A Nold-Petry
- The Ritchie Centre, MIMR-PHI Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
| | - Marcel F Nold
- The Ritchie Centre, MIMR-PHI Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
| | - Peter J Meikle
- Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Benjamin T Kile
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia and
| | - Ian M Smyth
- Department of Biochemistry and Molecular Biology Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia
| |
Collapse
|
232
|
Ballak DB, van Diepen JA, Moschen AR, Jansen HJ, Hijmans A, Groenhof GJ, Leenders F, Bufler P, Boekschoten MV, Müller M, Kersten S, Li S, Kim S, Eini H, Lewis EC, Joosten LAB, Tilg H, Netea MG, Tack CJ, Dinarello CA, Stienstra R. IL-37 protects against obesity-induced inflammation and insulin resistance. Nat Commun 2014; 5:4711. [DOI: 10.1038/ncomms5711] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/16/2014] [Indexed: 12/16/2022] Open
|
233
|
Anti-inflammatory effect of IL-37b in children with allergic rhinitis. Mediators Inflamm 2014; 2014:746846. [PMID: 25177111 PMCID: PMC4142748 DOI: 10.1155/2014/746846] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 07/04/2014] [Accepted: 07/14/2014] [Indexed: 11/17/2022] Open
Abstract
Background. Interleukin-37 (IL-37), a newly described member of IL-1family, functioned as a fundamental inhibitor of innate inflammatory and immune responses, especially its isoform IL-37b. Objective. This study was undertaken to evaluate the expression and regulation of IL-37b in children with allergic rhinitis (AR). Methods. Forty children with AR and twenty-five normal controls were included. The relationship between IL-37b and Th1/2 cytokines production in serum and nasal lavage was examined by enzyme-linked immunosorbent assay (ELISA). Peripheral blood mononuclear cells (PBMCs) were purified for in vitro regulation experiment of IL-37b. Intranasal mometasone furoate was given in AR children and IL-37b change after one-month treatment was detected using ELISA. Results. We observed significantly decreased IL-37b expression levels in both serum and nasal lavage compared to controls. IL-37b was negatively correlated with Th2 cytokines. Our results also showed that IL-37b downregulated Th2 cytokine expressed by PBMCs and this modulation was through mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) pathway. We also found that intranasal mometasone furoate therapy can promote nasal IL-37b expression. Conclusion. IL-37b may be involved in Th2 cytokine regulation in AR and its expression was related to the efficacy of intranasal steroid therapy.
Collapse
|
234
|
Intestinal expression of the anti-inflammatory interleukin-1 homologue IL-37 in pediatric inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2014; 59:e18-26. [PMID: 24732025 DOI: 10.1097/mpg.0000000000000387] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The function of interleukin (IL)-37 has not been resolved. We recently showed that IL-37 suppresses colonic inflammation in mice. To gain more insight into its relevance in human disease, we investigated the expression of IL-37 in the intestine of pediatric patients with chronic inflammatory bowel disease (IBD). METHODS Intestinal biopsies were obtained from children with IBD (18 Crohn disease [CD], 14 ulcerative colitis [UC] and 11 controls) during endoscopy and analyzed for IL-37 expression by immunohistochemistry and real-time polymerase chain reaction. Results were correlated with immunostaining for IL-18 and IL-17, messenger RNA (mRNA) levels of pro- and anti-inflammatory cytokines, and clinical parameters. RESULTS IL-37 protein was detected in epithelial cells and submucosal lymphoid cells of patients with CD and UC as well as healthy controls. IL-37 protein expression tended to be higher with submucosal lymphoid cell infiltration of patients with CD and UC and correlated with histological severity score of inflammation. IL-18 showed a staining pattern similar to that of IL-37, whereas staining for IL-17 revealed distinct positive cells scattered in the submucosal layer. mRNA expression of IL-8, IL-17, and IL-10 was upregulated in patients with CD and UC. mRNA levels of IL-18 and IL-37 were not significantly elevated compared with controls. Levels of IL-37 and IL-18 mRNA showed a positive correlation in the CD group. CONCLUSIONS IL-37 protein is expressed in healthy and diseased bowel tissue. IL-37 and IL-18 show a similar expression pattern and correlate at mRNA levels. Future studies are warranted to delineate the specific contribution of IL-37 to modulate chronic bowel inflammation in humans.
Collapse
|
235
|
Lee KH, Park M, Ji KY, Lee HY, Jang JH, Yoon IJ, Oh SS, Kim SM, Jeong YH, Yun CH, Kim MK, Lee IY, Choi HR, Ko KS, Kang HS. Bacterial β-(1,3)-glucan prevents DSS-induced IBD by restoring the reduced population of regulatory T cells. Immunobiology 2014; 219:802-12. [PMID: 25092569 DOI: 10.1016/j.imbio.2014.07.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 05/06/2014] [Accepted: 07/12/2014] [Indexed: 02/07/2023]
Abstract
Bacterial β-(1,3)-glucan has more advantages in terms of cost, yield and efficiency than that derived from mushrooms, plants, yeasts and fungi. We have previously developed a novel and high-yield β-(1,3)-glucan produced by Agrobacterium sp. R259. This study aimed to elucidate the functional mechanism and therapeutic efficacy of bacterial β-(1,3)-glucan in dextran sulfate sodium (DSS)-induced inflammatory bowel disease (IBD).Mice were orally pretreated with bacterial β-(1,3)-glucan at daily doses of 2.5 or 5mg/kg for 2 weeks. After 6 days of DSS treatment, clinical assessment of IBD severity and expression of pro-inflammatory cytokines were evaluated. In vivo cell proliferation was examined by immunohistochemistry using Ki-67 and ER-TR7 antibodies. The frequency of regulatory T cells (Tregs) was analyzed by flow cytometry. Natural killer (NK) activity and IgA level were evaluated using NK cytotoxicity assay and ELISA.The deterioration of body weight gain, colonic architecture, disease score and histological score was recovered in DSS-induced IBD mice when pretreated with bacterial β-(1,3)-glucan. The recruitment of macrophages and the gene expression of proinflammatory cytokines, such as IL-1β, IL-6 and IL-17A/F, were markedly decreased in the colon of β-(1,3)-glucan-pretreated mice. β-(1,3)-Glucan induced the recovery of Tregs in terms of their frequency in DSS-induced IBD mice. Intriguingly, β-(1,3)-glucan reversed the functional defects of NK cells and excessive IgA production in DSS-induced IBD mice.We conclude that bacterial β-(1,3)-glucan prevented the progression of DSS-induced IBD by recovering the reduction of Tregs, functional defect of NK cells and excessive IgA production.
Collapse
Affiliation(s)
- Kwang-Ho Lee
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-75, Republic of Korea
| | - Min Park
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-75, Republic of Korea
| | - Kon-Young Ji
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-75, Republic of Korea
| | - Hwa-Youn Lee
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-75, Republic of Korea
| | - Ji-Hun Jang
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-75, Republic of Korea
| | - Il-Joo Yoon
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-75, Republic of Korea
| | - Seung-Su Oh
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-75, Republic of Korea
| | - Su-Man Kim
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-75, Republic of Korea
| | - Yun-Hwa Jeong
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-75, Republic of Korea
| | - Chul-Ho Yun
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-75, Republic of Korea
| | - Mi-Kyoung Kim
- Naturence Co., Ltd. Tanchun Industrial Complex, Road 69, Tanchun-myeon, Gongju-City, Chungcheongnam-do, Republic of Korea
| | - In-Young Lee
- Naturence Co., Ltd. Tanchun Industrial Complex, Road 69, Tanchun-myeon, Gongju-City, Chungcheongnam-do, Republic of Korea
| | - Ha-Rim Choi
- Department of Food and Nutrition, Nambu University, Gwangju 506-706, Republic of Korea
| | - Ki-sung Ko
- Department of Medicine, Medical Research Institute, College of Medicine, Chung-Ang University, Republic of Korea
| | - Hyung-Sik Kang
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 500-75, Republic of Korea.
| |
Collapse
|
236
|
Wu B, Meng K, Ji Q, Cheng M, Yu K, Zhao X, Tony H, Liu Y, Zhou Y, Chang C, Zhong Y, Zhu Z, Zhang W, Mao X, Zeng Q. Interleukin-37 ameliorates myocardial ischaemia/reperfusion injury in mice. Clin Exp Immunol 2014; 176:438-51. [PMID: 24527881 DOI: 10.1111/cei.12284] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2014] [Indexed: 01/04/2023] Open
Abstract
Innate immune and inflammatory responses are involved in myocardial ischaemia/reperfusion (I/R) injury. Interleukin (IL)-37 is a newly identified member of the IL-1 family, and functions as a fundamental inhibitor of innate immunity and inflammation. However, its role in myocardial I/R injury remains unknown. I/R or sham operations were performed on male C57BL/6J mice. I/R mice received an injection of recombinant human IL-37 or vehicle, immediately before reperfusion. Compared with vehicle treatment, mice treated with IL-37 showed an obvious amelioration of the I/R injury, as demonstrated by reduced infarct size, decreased cardiac troponin T level and improved cardiac function. This protective effect was associated with the ability of IL-37 to suppress production of proinflammatory cytokines, chemokines and neutrophil infiltration, which together contributed to a decrease in cardiomyocyte apoptosis and reactive oxygen species (ROS) generation. In addition, we found that IL-37 inhibited the up-regulation of Toll-like receptor (TLR)-4 expression and nuclear factor kappa B (NF-kB) activation after I/R, while increasing the anti-inflammatory IL-10 level. Moreover, the administration of anti-IL-10R antibody abolished the protective effects of IL-37 in I/R injury. In-vitro experiments further demonstrated that IL-37 protected cardiomyocytes from apoptosis under I/R condition, and suppressed the migration ability of neutrophils towards the chemokine LIX. In conclusion, IL-37 plays a protective role against mouse myocardial I/R injury, offering a promising therapeutic medium for myocardial I/R injury.
Collapse
Affiliation(s)
- B Wu
- The Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Zhao JJ, Pan QZ, Pan K, Weng DS, Wang QJ, Li JJ, Lv L, Wang DD, Zheng HX, Jiang SS, Zhang XF, Xia JC. Interleukin-37 mediates the antitumor activity in hepatocellular carcinoma: role for CD57+ NK cells. Sci Rep 2014; 4:5177. [PMID: 24898887 PMCID: PMC4046124 DOI: 10.1038/srep05177] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 05/02/2014] [Indexed: 12/19/2022] Open
Abstract
The biological role of interleukin-37 (IL-37) in cancer is large unknown. Through immunohistochemical detection using 163 primary hepatocellular carcinoma (HCC) clinical specimens, we found the expression of IL-37 was decreased in tumor tissues, and the expression level was negatively correlated with tumor size. High expression of IL-37 in HCC tumor tissues was associated with better overall survival (OS) and disease-free survival (DFS). IL-37 expression in tumor tissues was positively associated with the density of tumor-infiltrating CD57+ natural killer (NK) cells, but not with the CD3+ and CD8+ T cells. Consistently, in vitro chemotaxis analysis showed that IL-37- overexpressing HCC cells could recruit more NK cells. The in vivo mouse model experiments also revealed that overexpression IL-37 in HCC cells significantly delayed tumor growth and recruited more NK cells into tumors tissues. Our finding suggested that IL-37 might play an important role for the prognosis of HCC patients via regulating innate immune-action.
Collapse
Affiliation(s)
- Jing-Jing Zhao
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China [3]
| | - Qiu-Zhong Pan
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2]
| | - Ke Pan
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China [3]
| | - De-Sheng Weng
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Qi-Jing Wang
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Jian-Jun Li
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Lin Lv
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Dan-Dan Wang
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Hai-Xia Zheng
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Shan-Shan Jiang
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Xiao-Fei Zhang
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China
| | - Jian-Chuan Xia
- 1] State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, People's Republic of China [2] Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou 510060, People's Republic of China
| |
Collapse
|
238
|
Expression and Regulation of Interleukin-37 in Pathogenesis of Nasal Polyps. Indian J Otolaryngol Head Neck Surg 2014; 66:401-6. [PMID: 26396952 DOI: 10.1007/s12070-014-0725-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 04/24/2014] [Indexed: 10/25/2022] Open
Abstract
Interleukin-37 (IL-37) belongs to IL-1 family and is recently identified as a natural suppressor of innate inflammatory and immune responses. Its role in digestive system was well characterized, however, little is known about its function in respiratory diseases. This study is aimed to investigate the expression and regulation of IL-37 in patients with nasal polyps (NPs). Twenty-five patients with NPs and sixteen normal controls were included, and IL-37 production was determined by immunohistochemistry and enzyme-linked immuno sorbent assay, respectively. The relationship between IL-37 expression and Th1/Th2 cytokines was also evaluated. Besides, the effect of IL-37 on dispersed nasal polyp cells (DNPCs) was investigated. We observed significantly decreased IL-37 mRNA and protein levels expression in NPs compared with normal control. IL-37 was found negatively with Th2 cytokines and had no relation with Th1 cytokines. Furthermore, we provided the first evidence that IL-37 down-regulates Th2 cytokine expressed by DNPCs. Our results demonstrate that enhanced Th2 cytokine levels was related to decreased IL-37 expression in NPs, and provide a possible explanation for IL-37's regulatory role in the pathogenesis of NPs.
Collapse
|
239
|
Abstract
Cytokines have a crucial role in the pathogenesis of inflammatory bowel diseases (IBDs), such as Crohn's disease and ulcerative colitis, where they control multiple aspects of the inflammatory response. In particular, the imbalance between pro-inflammatory and anti-inflammatory cytokines that occurs in IBD impedes the resolution of inflammation and instead leads to disease perpetuation and tissue destruction. Recent studies suggest the existence of a network of regulatory cytokines that has important implications for disease progression. In this Review, we discuss the role of cytokines produced by innate and adaptive immune cells, as well as their relevance to the future therapy of IBD.
Collapse
Affiliation(s)
- Markus F Neurath
- Department of Medicine 1, University of Erlangen-Nürnberg, Kussmaul Campus for Medical Research, 91054 Erlangen, Germany
| |
Collapse
|
240
|
Jartti T, Palomares O, Waris M, Tastan O, Nieminen R, Puhakka T, Rückert B, Aab A, Vuorinen T, Allander T, Vahlberg T, Ruuskanen O, Akdis M, Akdis CA. Distinct regulation of tonsillar immune response in virus infection. Allergy 2014; 69:658-67. [PMID: 24684577 PMCID: PMC7159333 DOI: 10.1111/all.12396] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2014] [Indexed: 01/05/2023]
Abstract
Background The relationships between tonsillar immune responses, and viral infection and allergy are incompletely known. Objective To study intratonsillar/nasopharyngeal virus detections and in vivo expressions of T‐cell‐ and innate immune response‐specific cytokines, transcription factors, and type I/II/III interferons in human tonsils. Methods Palatine tonsil samples were obtained from 143 elective tonsillectomy patients. Adenovirus, bocavirus‐1, coronavirus, enteroviruses, influenza virus, metapneumovirus, parainfluenza virus, rhinovirus, and respiratory syncytial virus were detected using PCR. The mRNA expression levels of IFN‐α, IFN‐β, IFN‐γ, IL‐10, IL‐13, IL‐17, IL‐28, IL‐29, IL‐37, TGF‐β, FOXP3, GATA3, RORC2, and Tbet were directly analyzed by quantitative RT‐PCR. Results Fifty percentage of subjects reported allergy, 59% had ≥1 nasopharyngeal viruses, and 24% had ≥1 intratonsillar viruses. Tonsillar virus detection showed a strong negative association with age; especially rhinovirus or parainfluenza virus detection showed positive association with IFN‐γ and Tbet expressions. IL‐37 expression was positively associated with atopic dermatitis, whereas IFN‐α, IL‐13, IL‐28, and Tbet expressions were negatively associated with allergic diseases. Network analyses demonstrated strongly polarized clusters of immune regulatory (IL‐10, IL‐17, TGF‐β, FOXP3, GATA3, RORC2, Tbet) and antiviral (IFN‐α, IFN‐β, IL‐28, IL‐29) genes. These two clusters became more distinctive in the presence of viral infection or allergy. A negative correlation between antiviral cytokines and IL‐10, IL‐17, IL‐37, FOXP3, and RORC2 was observed only in the presence of viruses, and interestingly, IL‐13 strongly correlated with antiviral cytokines. Conclusions Tonsillar cytokine expression is closely related to existing viral infections, age, and allergic illnesses and shows distinct clusters between antiviral and immune regulatory genes.
Collapse
Affiliation(s)
- T. Jartti
- Department of Pediatrics; Turku University Hospital; Turku Finland
| | - O. Palomares
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Christine Kühne-Center for Allergy Research and Education (CK-CARE); Davos Switzerland
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid (UCM); Madrid Spain
| | - M. Waris
- Department of Virology; University of Turku; Turku Finland
| | - O. Tastan
- Department of Computer Engineering; Bilkent University; Ankara Turkey
| | - R. Nieminen
- Department of Pediatrics; Turku University Hospital; Turku Finland
| | - T. Puhakka
- Department of Otorhinolaryngology; Turku University Hospital; Turku Finland
- Department of Otorhinolaryngology; Satakunta Central Hospital; Pori Finland
| | - B. Rückert
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Christine Kühne-Center for Allergy Research and Education (CK-CARE); Davos Switzerland
| | - A. Aab
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Christine Kühne-Center for Allergy Research and Education (CK-CARE); Davos Switzerland
| | - T. Vuorinen
- Department of Virology; University of Turku; Turku Finland
| | - T. Allander
- Department of Clinical Microbiology; Karolinska University Hospital; Stockholm Sweden
| | - T. Vahlberg
- Department of Biostatistics; University of Turku; Turku Finland
| | - O. Ruuskanen
- Department of Pediatrics; Turku University Hospital; Turku Finland
| | - M. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Christine Kühne-Center for Allergy Research and Education (CK-CARE); Davos Switzerland
| | - C. A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Christine Kühne-Center for Allergy Research and Education (CK-CARE); Davos Switzerland
| |
Collapse
|
241
|
Zhao PW, Jiang WG, Wang L, Jiang ZY, Shan YX, Jiang YF. Plasma levels of IL-37 and correlation with TNF-α, IL-17A, and disease activity during DMARD treatment of rheumatoid arthritis. PLoS One 2014; 9:e95346. [PMID: 24788826 PMCID: PMC4006923 DOI: 10.1371/journal.pone.0095346] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 03/25/2014] [Indexed: 01/26/2023] Open
Abstract
The aim of this study was to assess the change of IL-37 concentrations in rheumatoid arthritis (RA) patients under Disease-modifying anti-rheumatic drug (DMARD) therapy, and to establish a correlation between Interleukin-37 and pro-inflammatory cytokines in plasma and disease activity. The plasma level of IL-37 was determined using ELISA in 50 newly diagnosed RA patients and 30 healthy controls (HC). Plasma levels of IL-17A, IL-6 and TNF-α were measured using flow a cytometric bead array assay. We found that the concentrations of IL-37, as well as IL-17A, IL-6 and TNF-α, were higher in plasma of RA patients compared to HCs. Compared to patients who did not respond to DMARD treatment, treatment of patients responsive to DMARDs resulted in down-regulation of IL-17A, IL-6 and TNF-α expression. The plasma level of the anti-inflammatory cytokine IL-37 was also decreased in drug responders after DMARD treatment. The plasma level of IL-37 in RA patients was positively correlated with pro-inflammatory cytokines (IL-17A, TNF-α) and disease activity (CRP, DAS28) in RA patients. IL-37 expression in RA and during DMARD treatment appears to be controlled by the level of pro-inflammatory cytokines. This results in a strong correlation between plasma levels of IL-37 and disease activity in RA patients.
Collapse
Affiliation(s)
- Ping-Wei Zhao
- Key Laboratory of Zoonosis Research, Ministry of Education, The First Hospital, Jilin University, Changchun, China
| | - Wei-Guang Jiang
- Key Laboratory of Zoonosis Research, Ministry of Education, The First Hospital, Jilin University, Changchun, China
| | - Li Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, The First Hospital, Jilin University, Changchun, China
| | - Zhen-Yu Jiang
- Department of Rheumatism, The First Hospital, Jilin University, Changchun, China
| | - Yu-Xing Shan
- Key Laboratory of Zoonosis Research, Ministry of Education, The First Hospital, Jilin University, Changchun, China
| | - Yan-Fang Jiang
- Key Laboratory of Zoonosis Research, Ministry of Education, The First Hospital, Jilin University, Changchun, China
- * E-mail:
| |
Collapse
|
242
|
Bersudsky M, Luski L, Fishman D, White RM, Ziv-Sokolovskaya N, Dotan S, Rider P, Kaplanov I, Aychek T, Dinarello CA, Apte RN, Voronov E. Non-redundant properties of IL-1α and IL-1β during acute colon inflammation in mice. Gut 2014; 63:598-609. [PMID: 23793223 DOI: 10.1136/gutjnl-2012-303329] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The differential role of the IL-1 agonists, IL-1α, which is mainly cell-associated versus IL-1β, which is mostly secreted, was studied in colon inflammation. DESIGN Dextran sodium sulfate (DSS) colitis was induced in mice globally deficient in either IL-1α or IL-1β, and in wild-type mice, or in mice with conditional deletion of IL-1α in intestinal epithelial cells (IECs). Bone marrow transplantation experiments were performed to assess the role of IL-1α or IL-1β of myeloid versus colon non-hematopoietic cells in inflammation and repair in acute colitis. RESULTS IL-1α released from damaged IECs acts as an alarmin by initiating and propagating colon inflammation, as IL-1α deficient mice exhibited mild disease symptoms with improved recovery. IL-1β is involved in repair of IECs and reconstitution of the epithelial barrier during the resolution of colitis; its deficiency correlates with disease exacerbation. Neutralisation of IL-1α in control mice during acute colitis led to alleviation of clinical and histological manifestations, whereas treatment with rIL-1Ra or anti-IL-1β antibodies was not effective. Repair after colitis correlated with accumulation of CD8 and regulatory T cells in damaged crypts. CONCLUSIONS The role of IL-1α and IL-1β differs in DSS-induced colitis in that IL-1α, mainly of colon epithelial cells is inflammatory, whereas IL-1β, mainly of myeloid cell origin, promotes healing and repair. Given the dissimilar functions of each IL-1 agonistic molecule, an IL-1 receptor blockade would not be as therapeutically effective as specific neutralising of IL-1α, which leaves IL-1β function intact.
Collapse
Affiliation(s)
- Marina Bersudsky
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology and Immunology and The Cancer Research Center, Ben-Gurion University of the Negev, , Beer-Sheva, Israel
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
IL-37 inhibits the production of inflammatory cytokines in peripheral blood mononuclear cells of patients with systemic lupus erythematosus: its correlation with disease activity. J Transl Med 2014; 12:69. [PMID: 24629023 PMCID: PMC4003851 DOI: 10.1186/1479-5876-12-69] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 03/12/2014] [Indexed: 12/15/2022] Open
Abstract
Background Interleukin-37 (IL-37), a new member of IL-1 family cytokine, is recently identified as a natural inhibitor of innate immunity. This study aimed to measure the peripheral blood mononuclear cells (PBMCs) and serum levels of IL-37 in patients with systemic lupus erythematosus (SLE) and to investigate its role in SLE, including its correlation with disease activity, organ disorder and the regulation of inflammatory cytokines. Methods The expressions of IL-37 mRNAs in PBMCs and serum IL-37 levels in 66 SLE patients were measured by real-time polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). SLE patients PBMCs were stimulated with recombinant IL-37, levels of cytokines TNF-α, IL-1β, IL-6 and IL-10 were detected by RT-PCR and ELISA. Results IL-37 mRNAs and serum protein levels were higher in patients with SLE compared with healthy controls. Patients with active disease showed higher IL-37 mRNAs and serum protein levels compared with those with inactive disease as well as healthy controls. Serum IL-37 levels correlated with SLEDAI and inversely with C3 and C4. Serum IL-37 levels were higher in SLE patients with renal involvement compared with those without renal disease. In vitro, IL-37 inhibited the production of TNF-α, IL-1β and IL-6 in PBMCs of patients with SLE, whereas the production of IL-10 was unaffected. Conclusions IL-37 associated with SLE disease activity, especially related with SLE renal disease activity. IL-37 is an important cytokine in the control of SLE pathogenesis by suppressing the production of inflammatory cytokines. Thus, IL-37 may provide a novel research target for the pathogenesis and therapy of SLE.
Collapse
|
244
|
Elevated plasma IL-37, IL-18, and IL-18BP concentrations in patients with acute coronary syndrome. Mediators Inflamm 2014; 2014:165742. [PMID: 24733959 PMCID: PMC3964842 DOI: 10.1155/2014/165742] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/19/2014] [Accepted: 01/20/2014] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE More recently, evidence showed that the novel anti-inflammatory cytokine interleukin- (IL-) 37 was expressed in the foam-like cells of atherosclerotic coronary and carotid artery plaques, suggesting that IL-37 is involved in atherosclerosis-related diseases. However, the plasma levels of IL-37 in patients with acute coronary syndrome (ACS, including unstable angina pectoris and acute myocardial infarction) have yet to be investigated. METHODS Plasma IL-37, IL-18, and IL-18BP levels were measured in 50 patients with stable angina pectoris (SAP), 75 patients with unstable angina pectoris (UAP), 67 patients with acute myocardial infarction (AMI), and 65 control patients. RESULTS The plasma IL-37, IL-18, and IL-18BP levels were significantly increased in ACS patients compared to SAP and control patients. A correlation analysis showed that the plasma biomarker levels were positively correlated with each other and with the levels of C-reactive protein (CRP), N-terminal probrain natriuretic peptide (NT-proBNP), and left ventricular end-diastolic dimension (LVEDD) but negatively correlated with left ventricular ejection fraction (LVEF). Furthermore, the plasma IL-37, IL-18, and IL-18BP had no correlation with the severity of the coronary artery stenosis. CONCLUSIONS The results indicate that the plasma IL-37 levels are associated with the onset of ACS.
Collapse
|
245
|
Garlanda C, Dinarello CA, Mantovani A. The interleukin-1 family: back to the future. Immunity 2014; 39:1003-18. [PMID: 24332029 DOI: 10.1016/j.immuni.2013.11.010] [Citation(s) in RCA: 1436] [Impact Index Per Article: 130.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 11/20/2013] [Indexed: 12/14/2022]
Abstract
Interleukin-1 (IL-1) is a central mediator of innate immunity and inflammation. The IL-1 family includes seven ligands with agonist activity (IL-1α and IL-1β, IL-18, IL-33, IL-36α, IL-36β, IL-36γ), three receptor antagonists (IL-1Ra, IL-36Ra, IL-38), and an anti-inflammatory cytokine (IL-37). Members of the IL-1 Receptor (IL-1R) family include six receptor chains forming four signaling receptor complexes, two decoy receptors (IL-1R2, IL-18BP), and two negative regulators (TIR8 or SIGIRR, IL-1RAcPb). A tight regulation via receptor antagonists, decoy receptors, and signaling inhibitors ensures a balance between amplification of innate immunity and uncontrolled inflammation. All cells of the innate immune system express and/or are affected by IL-1 family members. Moreover, IL-1 family members play a key role in the differentiation and function of polarized innate and adaptive lymphoid cells. Here we will review the key properties of IL-1 family members, with emphasis on pathways of negative regulation and orchestration of innate and adaptive immunity.
Collapse
Affiliation(s)
- Cecilia Garlanda
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Italy
| | - Charles A Dinarello
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Medicine, Radboud University Medical Center, Nijmegen 6500 HC, The Netherlands
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Italy; BIOMETRA Department, Università degli Studi di Milano, 20133 Milano, Italy.
| |
Collapse
|
246
|
Bulau AM, Nold MF, Li S, Nold-Petry CA, Fink M, Mansell A, Schwerd T, Hong J, Rubartelli A, Dinarello CA, Bufler P. Role of caspase-1 in nuclear translocation of IL-37, release of the cytokine, and IL-37 inhibition of innate immune responses. Proc Natl Acad Sci U S A 2014; 111:2650-5. [PMID: 24481253 PMCID: PMC3932872 DOI: 10.1073/pnas.1324140111] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
IL-37 is a fundamental inhibitor of innate immunity. Human IL-37 has a caspase-1 cleavage site and translocates to the nucleus upon LPS stimulation. Here, we investigated whether caspase-1 processing affects IL-37-mediated suppression of LPS-induced cytokines and the release from cells by analyzing a caspase-1 cleavage site mutant IL-37 (IL-37D20A). Nuclear translocation of IL-37D20A is significantly impaired compared with WT IL-37 in transfected cells. LPS-induced IL-6 was decreased in cells expressing WT IL-37 but not IL-37D20A. The function of IL-37 in transfected bone marrow-derived macrophages is nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome-dependent, because IL-37 transfection in apoptosis-associated speck-like protein containing a carboxyl-terminal caspase recruitment domain- and NLRP3-deficient cells does not reduce levels of IL-6 and IL-1β upon LPS stimulation. IL-37-expressing macrophages release both precursor and mature IL-37, but only the externalization of mature IL-37 was dependent on ATP. Precursor and mature IL-37 was also secreted from human dendritic cells and peripheral blood mononuclear cells. To determine whether IL-37 is active in the extracellular compartment, we pretreated IL-37 transgenic mice with IL-37-neutralizing antibodies before LPS challenge. In IL-37-expressing mice, neutralizing IL-37 antibodies reversed the suppression of LPS-induced serum IL-6. In contrast, the addition of neutralizing antibody did not reverse suppression of LPS-induced IL-6 in mouse macrophages transfected with IL-37. Although caspase-1 is required for nuclear translocation of intracellular IL-37 and for secretion of mature IL-37, the release of the IL-37 precursor is independent of caspase-1 activation. IL-37 now emerges as a dual-function cytokine with intra- and extracellular properties for suppressing innate inflammation.
Collapse
Affiliation(s)
- Ana-Maria Bulau
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, D-80337 Munich, Germany
| | | | - Suzhao Li
- University of Colorado Denver, Aurora, CO 80045
| | | | - Michaela Fink
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, D-80337 Munich, Germany
| | - Ashley Mansell
- Centre for Innate Immunity and Infectious Diseases, Monash Institute of Medical Research, Monash University, Melbourne, VIC 3168, Australia
| | - Tobias Schwerd
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, D-80337 Munich, Germany
| | - Jaewoo Hong
- Department of Biomedical Science and Technology, Konkuk University, Seoul 143-701, Korea
| | - Anna Rubartelli
- Cell Biology Unit, Istituto Di Ricovero e Cura a Carattere Scientifico, Azienda Ospedaliera Universitaria San Martino, Istituto Scientifica (per lo studio e la cura dei) Tumori, 16132 Genoa, Italy; and
| | - Charles A. Dinarello
- University of Colorado Denver, Aurora, CO 80045
- Department of Medicine, Radboud University Medical Centre, 6500 HB, Nijmegen, The Netherlands
| | - Philip Bufler
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, D-80337 Munich, Germany
| |
Collapse
|
247
|
Abstract
The immune system provides defenses against invading pathogens while maintaining immune tolerance to self-antigens. This immune homeostasis is harmonized by the direct interactions between immune cells and the cytokine environment in which immune cells develop and function. Herein, we discuss three non-redundant paradigms by which cytokines maintain or break immune tolerance. We firstly describe how anti-inflammatory cytokines exert direct inhibitory effects on immune cells to enforce immune tolerance, followed by discussing other cytokines that maintain immune tolerance through inducing CD4(+)Foxp3(+) regulatory T cells (Tregs), which negatively control immune cells. Interleukin (IL)-2 is the most potent cytokine in promoting the development and survival of Tregs, thereby mediating immune tolerance. IL-35 is mainly produced by Tregs, but its biology function remains to be defined. Finally, we discuss the actions of proinflammatory cytokines that breach immune tolerance and induce autoimmunity, which include IL-7, IL-12, IL-21, and IL-23. Recent genetic studies have revealed the role of these cytokines (or their cognate receptors) in susceptibility to autoimmune diseases. Taken together, we highlight in this review the cytokine regulation of immune tolerance, which will help in further understanding of human diseases that are caused by dysregulated immune system.
Collapse
Affiliation(s)
- Jie Wu
- Department of Surgery, Center for Immunobiology and Transplantation Research, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas USA
| | - Aini Xie
- Department of Surgery, Center for Immunobiology and Transplantation Research, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas USA
| | - Wenhao Chen
- Department of Surgery, Center for Immunobiology and Transplantation Research, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas USA
| |
Collapse
|
248
|
Teng X, Hu Z, Wei X, Wang Z, Guan T, Liu N, Liu X, Ye N, Deng G, Luo C, Huang N, Sun C, Xu M, Zhou X, Deng H, Edwards CK, Chen X, Wang X, Cui K, Wei Y, Li J. IL-37 Ameliorates the Inflammatory Process in Psoriasis by Suppressing Proinflammatory Cytokine Production. THE JOURNAL OF IMMUNOLOGY 2014; 192:1815-23. [DOI: 10.4049/jimmunol.1300047] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
249
|
Masterson JC, McNamee EN, Hosford L, Capocelli KE, Ruybal J, Fillon SA, Doyle AD, Eltzschig HK, Rustgi AK, Protheroe CA, Lee NA, Lee JJ, Furuta GT. Local hypersensitivity reaction in transgenic mice with squamous epithelial IL-5 overexpression provides a novel model of eosinophilic oesophagitis. Gut 2014; 63:43-53. [PMID: 23161496 PMCID: PMC3884674 DOI: 10.1136/gutjnl-2012-303631] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Eosinophilic oesophagitis (EoE) is a chronic inflammatory condition of the oesophagus with limited treatment options. No previous transgenic model has specifically targeted the oesophageal mucosa to induce oesophageal eosinophilia. DESIGN We developed a mouse model that closely resembles EoE by utilising oxazolone haptenation in mice with transgenic overexpression of an eosinophil poietic and survival factor (interleukin (IL)-5) in resident squamous oesophageal epithelia. RESULTS Overexpression of IL-5 in the healthy oesophagus was achieved in transgenic mice (L2-IL5) using the squamous epithelial promoter Epstein-Barr virus ED-L2. Oxazolone-challenged L2-IL5 mice developed dose-dependent pan-oesophageal eosinophilia, including eosinophil microabscess formation and degranulation as well as basal cell hyperplasia. Moreover, oesophagi expressed increased IL-13 and the eosinophil agonist chemokine eotaxin-1. Treatment of these mice with corticosteroids significantly reduced eosinophilia and epithelial inflammation. CONCLUSIONS L2-IL5 mice provide a novel experimental model that can potentially be used in preclinical testing of EoE-related therapeutics and mechanistic studies identifying pathogenetic features associated with mucosal eosinophilia.
Collapse
Affiliation(s)
- Joanne C Masterson
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Digestive Health Institute, Aurora, Colorado, USA,Gastrointestinal Eosinophilic Diseases Program, Aurora, Colorado, USA,Children’s Hospital Colorado, Aurora, Colorado, USA,Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, USA,University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Eóin N McNamee
- Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, USA,Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA,University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Lindsay Hosford
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Digestive Health Institute, Aurora, Colorado, USA,Gastrointestinal Eosinophilic Diseases Program, Aurora, Colorado, USA,Children’s Hospital Colorado, Aurora, Colorado, USA,Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, USA,University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Kelley E Capocelli
- Gastrointestinal Eosinophilic Diseases Program, Aurora, Colorado, USA,Children’s Hospital Colorado, Aurora, Colorado, USA,Department of Pathology, Children’s Hospital Colorado, Aurora, Colorado, USA,University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Joseph Ruybal
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Digestive Health Institute, Aurora, Colorado, USA,Gastrointestinal Eosinophilic Diseases Program, Aurora, Colorado, USA,Children’s Hospital Colorado, Aurora, Colorado, USA,Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, USA,University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Sophie A Fillon
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Digestive Health Institute, Aurora, Colorado, USA,Gastrointestinal Eosinophilic Diseases Program, Aurora, Colorado, USA,Children’s Hospital Colorado, Aurora, Colorado, USA,Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, USA,University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Alfred D Doyle
- Department of Biochemistry and Molecular Biology, Mayo Clinic Scottsdale, Arizona, USA
| | - Holger K Eltzschig
- Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, USA,Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA,University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Anil K Rustgi
- Division of Gastroenterology, Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, USA
| | - Cheryl A Protheroe
- Department of Biochemistry and Molecular Biology, Mayo Clinic Scottsdale, Arizona, USA
| | - Nancy A Lee
- Department of Biochemistry and Molecular Biology, Mayo Clinic Scottsdale, Arizona, USA
| | - James J Lee
- Department of Biochemistry and Molecular Biology, Mayo Clinic Scottsdale, Arizona, USA
| | - Glenn T Furuta
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Digestive Health Institute, Aurora, Colorado, USA,Gastrointestinal Eosinophilic Diseases Program, Aurora, Colorado, USA,Children’s Hospital Colorado, Aurora, Colorado, USA,Mucosal Inflammation Program, University of Colorado Denver, Aurora, Colorado, USA,University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
250
|
Wang X, Yin J, Zheng L, Wang L, Shi Y, Tang W, Ding G, Liu C, Liu R, Chen S, Gu H. The variant interleukin 1f7 rs3811047 G>A was associated with a decreased risk of gastric cardiac adenocarcinoma in a Chinese Han population. Tumour Biol 2013; 35:3509-15. [DOI: 10.1007/s13277-013-1463-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 11/22/2013] [Indexed: 01/28/2023] Open
|