1
|
Chen L, Gao C, Yin X, Mo L, Cheng X, Chen H, Jiang C, Wu B, Zhao Y, Li H, Li Y, Li J, Chen L, Deng Q, Yao P, Tang Y. Partial reduction of interleukin-33 signaling improves senescence and renal injury in diabetic nephropathy. MedComm (Beijing) 2024; 5:e742. [PMID: 39465143 PMCID: PMC11502718 DOI: 10.1002/mco2.742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 10/29/2024] Open
Abstract
Diabetic nephropathy (DN) is a frequent and costly complication of diabetes with limited understandings of mechanisms and therapies. Emerging evidence points to the important roles of interleukin-33 (IL-33) in acute kidney injury, yet its contribution to DN is still unclear. We here found a ubiquitous increase of IL-33 and its receptor (ST2) in murine models and patients with DN. Surprisingly, both IL-33 and ST2 knockdown aggravated renal lesions in DN, while overexpression of IL-33 also exacerbated the condition. Further population-based analyses revealed a positive correlation of IL-33 expression with renal dysfunction in DN patients. Individuals with high IL-33 expression-related polygenic risk score had a higher DN risk. These findings confirmed the harmful effects of IL-33 on DN. Conversely, endogenous and exogenous partial reduction of IL-33 signaling conferred renoprotective effects in vivo and in vitro. Mechanistically, IL-33 induced senescence by regulating cell cycle factors in HK-2 cells, and accordingly senescence led to renal cell damage through the secretion of senescence-related secretory phenotype (SASP) including IL-33 and prostaglandins. Together, elevated IL-33 accelerates cellular senescence to drive DN possibly by SASP production, while a partial blockage improves renal injury and senescence. Our findings pinpoint a possible and new avenue for DN interventions.
Collapse
Affiliation(s)
- Li Chen
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Oil Crops Research Institute of the Chinese Academy of Agricultural SciencesHubei Key Laboratory of Lipid Chemistry and Nutritionand Key Laboratory of Oilseeds ProcessingMinistry of AgricultureOil Crops and Lipids Process Technology National & Local Joint Engineering LaboratoryWuhanHubeiChina
| | - Chao Gao
- National Institute for Nutrition and HealthChinese Center for Disease Control and Prevention BeijingBeijingChina
| | - Xingzhu Yin
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Li Mo
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xueer Cheng
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Huimin Chen
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chunjie Jiang
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Bangfu Wu
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ying Zhao
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hongxia Li
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yanyan Li
- Shenzhen Center for Chronic Disease ControlShenzhenChina
| | - Jiansha Li
- Institute of PathologyTongji HospitalWuhanChina
- Department of PathologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Liangkai Chen
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qianchun Deng
- Oil Crops Research Institute of the Chinese Academy of Agricultural SciencesHubei Key Laboratory of Lipid Chemistry and Nutritionand Key Laboratory of Oilseeds ProcessingMinistry of AgricultureOil Crops and Lipids Process Technology National & Local Joint Engineering LaboratoryWuhanHubeiChina
| | - Ping Yao
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuhan Tang
- Department of Nutrition and Food HygieneHubei Key Laboratory of Food Nutrition and SafetyMinistry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and HealthKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionState Key Laboratory of Environment Health (Incubation)School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
2
|
Mohammadzadeh M, Athari SZ, Ghiasi F, Keyhanmanesh R, Ghaffari-Nasab A, Roshangar L, Korjan ES, Delkhosh A, Bavil FM. Bone Marrow-Derived C-Kit + Cells Improved Inflammatory IL-33/ST-2/ILC2 Axis in the Lung Tissue of Type 2 Diabetic Rats. Appl Biochem Biotechnol 2024; 196:7074-7088. [PMID: 38478319 DOI: 10.1007/s12010-024-04870-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 11/21/2024]
Abstract
Inflammation is an essential factor in pulmonary complications of diabetes. Bone marrow (BM)-derived C-kit+ cells have immunomodulatory properties and their transplantation is suggested as a promising strategy for ameliorating diabetes complications. This study evaluated the effect of BM-derived C-kit+ cells on the inflammation signaling pathway in lung tissue of type 2 diabetic male rats. Ten rats were used to extract C-kit cells, and 48 male Wistar rats weighing 180 ± 20 g were randomly divided into four equal groups: (1) Control (Cont), (2) Diabetic (D), (3) Diabetic + C-kit+ cells (D + C-kit pos) intravenously injected 50-µl phosphate buffer saline (PBS) containing 300,000 C-kit+ cells, and (4) Diabetic + C-kit- cells (D + C-kit neg), intravenously injected C-kit- cells. Diabetes induction increased IL-33, ST-2, CD127, and IL-2 levels and decreased IL-10. C-kit+ cell therapy significantly decreased IL-33 and CD127 and increased IL-10. In addition, lung histopathological changes significantly improved in the C-kit+ group compared to the diabetic group. These findings suggest that C-kit+ cells may have a potential therapeutic role in mitigating diabetes-induced respiratory complications via ameliorating the inflammation and histopathological changes in lung tissue.
Collapse
Affiliation(s)
- Milad Mohammadzadeh
- Stem Cell Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 51666-14766, Iran
- Department of Basic Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Seyed Zanyar Athari
- Stem Cell Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 51666-14766, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Ghiasi
- Stem Cell Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 51666-14766, Iran
| | - Rana Keyhanmanesh
- Stem Cell Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 51666-14766, Iran
| | - Arshad Ghaffari-Nasab
- Stem Cell Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 51666-14766, Iran
| | - Leila Roshangar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elnaz Salmani Korjan
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aref Delkhosh
- Stem Cell Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, 51666-14766, Iran
| | - Fariba Mirzaei Bavil
- Department of Basic Sciences, Maragheh University of Medical Sciences, Maragheh, Iran.
| |
Collapse
|
3
|
Yadav M, Uikey BN, Rathore SS, Gupta P, Kashyap D, Kumar C, Shukla D, Vijayamahantesh, Chandel AS, Ahirwar B, Singh AK, Suman SS, Priyadarshi A, Amit A. Role of cytokine in malignant T-cell metabolism and subsequent alternation in T-cell tumor microenvironment. Front Oncol 2023; 13:1235711. [PMID: 37746258 PMCID: PMC10513393 DOI: 10.3389/fonc.2023.1235711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
T cells are an important component of adaptive immunity and T-cell-derived lymphomas are very complex due to many functional sub-types and functional elasticity of T-cells. As with other tumors, tissues specific factors are crucial in the development of T-cell lymphomas. In addition to neoplastic cells, T- cell lymphomas consist of a tumor micro-environment composed of normal cells and stroma. Numerous studies established the qualitative and quantitative differences between the tumor microenvironment and normal cell surroundings. Interaction between the various component of the tumor microenvironment is crucial since tumor cells can change the microenvironment and vice versa. In normal T-cell development, T-cells must respond to various stimulants deferentially and during these courses of adaptation. T-cells undergo various metabolic alterations. From the stage of quiescence to attention of fully active form T-cells undergoes various stage in terms of metabolic activity. Predominantly quiescent T-cells have ATP-generating metabolism while during the proliferative stage, their metabolism tilted towards the growth-promoting pathways. In addition to this, a functionally different subset of T-cells requires to activate the different metabolic pathways, and consequently, this regulation of the metabolic pathway control activation and function of T-cells. So, it is obvious that dynamic, and well-regulated metabolic pathways are important for the normal functioning of T-cells and their interaction with the microenvironment. There are various cell signaling mechanisms of metabolism are involved in this regulation and more and more studies have suggested the involvement of additional signaling in the development of the overall metabolic phenotype of T cells. These important signaling mediators include cytokines and hormones. The impact and role of these mediators especially the cytokines on the interplay between T-cell metabolism and the interaction of T-cells with their micro-environments in the context of T-cells lymphomas are discussed in this review article.
Collapse
Affiliation(s)
- Megha Yadav
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Blessi N. Uikey
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Priyanka Gupta
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Diksha Kashyap
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Chanchal Kumar
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Dhananjay Shukla
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Vijayamahantesh
- Department of Immunology and Microbiology, University of Missouri, Columbia, SC, United States
| | - Arvind Singh Chandel
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo, Japan
| | - Bharti Ahirwar
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Shashi Shekhar Suman
- Department of Zoology, Udayana Charya (UR) College, Lalit Narayan Mithila University, Darbhanga, India
| | - Amit Priyadarshi
- Department of Zoology, Veer Kunwar Singh University, Arrah, India
| | - Ajay Amit
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| |
Collapse
|
4
|
Xie JY, Wang WJ, Wang N, Dong Q, Han H, Feng YP, Yuan Y, Feng J, Chen K. A novel immune-related gene signature correlated with serum IL33 expression in acute myeloid leukemia prognosis. Am J Transl Res 2023; 15:4332-4344. [PMID: 37434810 PMCID: PMC10331686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/15/2023] [Indexed: 07/13/2023]
Abstract
PURPOSE To identify and validate the immune-related gene signature in patients with acute myeloid leukemia (AML). METHODS Differentially expressed genes (DEGs) profiles and survival data were obtained from The Cancer Genome Atlas (TCGA), following screened immune-associated genes from the InnateDB database. Subsequently, the weighted gene co-expression network analysis (WGCNA) was used to detect functional modules, and survival analysis was performed. The least absolute shrinkage and selection operator (LASSO) regression model combined with a partial likelihood-based Cox proportional hazard regression model was applied to select prognostic genes, and the ESTIMATE algorithm was used to construct an immune score-based risk assessment model. Finally, two independent datasets from the Gene Expression Omnibus (GEO) and our clinical data were used for external validation. Moreover, a subpopulation of the immune microenvironment cells was analyzed by the CIBERSORT algorithm, and its related serum indicator was identified by the enzyme-linked immunosorbent assay (ELISA) in clinical samples. RESULTS Finally, CTSD, GNB2, CDK6, and WAS were identified as the immune-related gene signature, and the risk stratification model was validated in both the GSE12417 database and our clinical cohort. Furthermore, the fraction of activated mast cells was identified. CIBERSORT algorithm showed that these cells have a positive association with prognosis. In addition, mast cell stimulator IL-33 was markedly decreased in AML patients with poor prognoses. CONCLUSION A novel immune-related gene signature (CTSD, GNB2, CDK6 and WAS) and its associated plasma indicator (mast cells activator, IL-33) were found to have prognostic value in AML patients.
Collapse
Affiliation(s)
- Jin-Ye Xie
- Department of Laboratory Medicine, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
| | - Wei-Jia Wang
- Department of Laboratory Medicine, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
- Department of Medical Research, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
| | - Nan Wang
- Department of Laboratory Medicine, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
| | - Qian Dong
- Department of Laboratory Medicine, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
| | - Hui Han
- Department of Laboratory Medicine, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
| | - Yan-Pin Feng
- Department of Laboratory Medicine, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
| | - Yong Yuan
- Department of Cardiovascular Medicine, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
- Department of Medical Research, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
| | - Juan Feng
- School of Medicine, Foshan UniversityFoshan 528225, Guangdong, China
| | - Kang Chen
- Department of Laboratory Medicine, Zhongshan City People’s HospitalZhongshan 528403, Guangdong, China
| |
Collapse
|
5
|
Lei S, Jin J, Zhao X, Zhou L, Qi G, Yang J. The role of IL-33/ST2 signaling in the tumor microenvironment and Treg immunotherapy. Exp Biol Med (Maywood) 2022; 247:1810-1818. [PMID: 35733343 PMCID: PMC9679353 DOI: 10.1177/15353702221102094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Interleukin (IL)-33 is a tissue-derived nuclear cytokine belonging to the IL-1 family. Stimulation-2 (ST2) is the only known IL-33 receptor. ST2 signals mostly on immune cells found within tissues, such as regulatory T cells (Treg cells), CD8+ T cells, and natural killer (NK) cells. Therefore, the IL-33/ST2 signaling pathway is important in the immune system. IL-33 deficiency impairs Treg cell function. ST2 signaling is also increased in active Treg cells, providing a new approach for Treg-related immunotherapy. The IL-33/ST2 signaling pathway regulates multiple immune-related cells by activating various intracellular kinases and factors in the tumor microenvironment (TME). Here, we review the latest studies on the role of the IL-33/ST2 signaling pathway in TME and Treg immunotherapy.
Collapse
Affiliation(s)
- Shangbo Lei
- Department of Immunology, Guilin Medical University, Guilin 541199, Guangxi, China,Department of Pathophysiology, Guilin Medical University, Guilin 541199, Guangxi, China,Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Jiamin Jin
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Xiangfeng Zhao
- Department of Immunology, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Lihua Zhou
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Guangying Qi
- Department of Pathophysiology, Guilin Medical University, Guilin 541199, Guangxi, China,Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Jinfeng Yang
- Department of Immunology, Guilin Medical University, Guilin 541199, Guangxi, China,Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, Guangxi, China,Jinfeng Yang.
| |
Collapse
|
6
|
Shakerian L, Kolahdooz H, Garousi M, Keyvani V, Kamal Kheder R, Abdulsattar Faraj T, Yazdanpanah E, Esmaeili SA. IL-33/ST2 axis in autoimmune disease. Cytokine 2022; 158:156015. [PMID: 36041312 DOI: 10.1016/j.cyto.2022.156015] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/03/2022]
Abstract
Interleukin-33 (IL-33) is a member of the IL-1 family and plays an ambivalent role in autoimmune diseases. IL-33 signals via the ST2 receptor and drives cytokine production in mast cells, basophils, eosinophils, NK cells, and T lymphocyte cells. The vital role of IL-33 as an active component gives rise to aberrant local and systemic damage which has been demonstrated in numerous inflammatory disorders and immune-mediated pathological conditions including multiple sclerosis (MS), rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), psoriasis, Sjogren's syndrome, inflammatory bowel disease (IBD), etc. IL-33/ST2 axis can up-regulate pro-inflammatory cytokine release in autoimmune disease, however, in some metabolic diseases like diabetes mellitus type 1 IL-33 can be considered an anti-inflammatory cytokine. The purpose of this review is to discuss selected studies on IL-33/ST2 axis in autoimmune diseases and its potential role as a pathogenic or protective cytokine.
Collapse
Affiliation(s)
- Leila Shakerian
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanieh Kolahdooz
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mitra Garousi
- Department of Internal Medicine, Faculty of Medical Sciences, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Vahideh Keyvani
- Molecular Genetics, Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Ramiar Kamal Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Rania 46012, Sulaymaniyah, Iraq; Department of Basic Sciences, College of Medicine, Hawler Medical University, Erbil, Iraq
| | - Tola Abdulsattar Faraj
- Department of Basic Sciences, College of Medicine, Hawler Medical University, Erbil, Iraq; Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Yeoh WJ, Vu VP, Krebs P. IL-33 biology in cancer: An update and future perspectives. Cytokine 2022; 157:155961. [PMID: 35843125 DOI: 10.1016/j.cyto.2022.155961] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/03/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
Interleukin-33 (IL-33) is a member of the IL-1 family of cytokines that is constitutively expressed in the nucleus of epithelial, endothelial and fibroblast-like cells. Upon cell stress, damage or necrosis, IL-33 is released into the cytoplasm to exert its prime role as an alarmin by binding to its specific receptor moiety, ST2. IL-33 exhibits pleiotropic function in inflammatory diseases and particularly in cancer. IL-33 may play a dual role as both a pro-tumorigenic and anti-tumorigenic cytokine, dependent on tumor and cellular context, expression levels, bioactivity and the nature of the inflammatory environment. In this review, we discuss the differential contribution of IL-33 to malignant or inflammatory conditions, its multifaceted effects on the tumor microenvironment, while providing possible explanations for the discrepant findings described in the literature. Additionally, we examine the emerging and divergent functions of IL-33 in the nucleus, and aspects of IL-33 biology that are currently under-addressed.
Collapse
Affiliation(s)
- Wen Jie Yeoh
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Vivian P Vu
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Philippe Krebs
- Institute of Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
8
|
Tan XY, Jing HY, Ma YR. Interleukin-33/ Suppression of Tumorigenicity 2 in Renal Fibrosis: Emerging Roles in Prognosis and Treatment. Front Physiol 2022; 12:792897. [PMID: 35046838 PMCID: PMC8761767 DOI: 10.3389/fphys.2021.792897] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/02/2021] [Indexed: 12/20/2022] Open
Abstract
Chronic kidney disease (CKD) is a major public health problem that affects more than 10% of the population worldwide and has a high mortality rate. Therefore, it is necessary to identify novel treatment strategies for CKD. Incidentally, renal fibrosis plays a central role in the progression of CKD to end-stage renal disease (ESRD). The activation of inflammatory pathways leads to the development of renal fibrosis. In fact, interleukin-33 (IL-33), a newly discovered member of the interleukin 1 (IL-1) cytokine family, is a crucial regulator of the inflammatory process. It exerts pro-inflammatory and pro-fibrotic effects via the suppression of tumorigenicity 2 (ST2) receptor, which, in turn, activates other inflammatory pathways. Although the role of this pathway in cardiac, pulmonary, and hepatic fibrotic diseases has been extensively studied, its precise role in renal fibrosis has not yet been completely elucidated. Recent studies have shown that a sustained activation of IL-33/ST2 pathway promotes the development of renal fibrosis. However, with prolonged research in this field, it is expected that the IL-33/ST2 pathway will be used as a diagnostic and prognostic tool for renal diseases. In addition, the IL-33/ST2 pathway seems to be a new target for the future treatment of CKD. Here, we review the mechanisms and potential applications of the IL-33/ST2 pathway in renal fibrosis; such that it can help clinicians and researchers to explore effective treatment options and develop novel medicines for CKD patients.
Collapse
Affiliation(s)
- Xiao-Yang Tan
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hao-Yue Jing
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue-Rong Ma
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
9
|
Plasma IL-33 levels are decreased in patients with high-risk myelodysplastic syndrome and show no correlation with pro-inflammatory IL-6 levels. Cytokine 2021; 148:155617. [PMID: 34130905 DOI: 10.1016/j.cyto.2021.155617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 11/21/2022]
Abstract
Aberrant inflammatory signaling has been shown to be a key pathogenic driver in myelodysplastic syndromes (MDS). Abnormal IL-33 expression has been implicated in inflammatory, immune-related disorders and, some tumors. However, its role in MDS remains widely unknown. This study aimed to evaluate the relationship between plasma levels of IL-33, clinical and prognostic data and, IL-6 levels in 101 patients with MDS. A comparative group of 59 healthy individuals was also evaluated. Plasma levels of cytokines were determined by enzyme-linked immunosorbent assay. Lower levels of IL-33 were found in patients with MDS when compared to the control group (p = 0.001), mainly in patients with more advanced stages of the disease and worse prognosis. No significant correlation between the levels of IL-33 and IL-6 was observed (r = 0.175; p = 0.081). These results reinforce the close association between immunological disorders and the pathogenesis of MDS. A greater understanding of the role of inflammatory cytokines in the disease can potentially provide new diagnosis and prognosis markers and new therapeutic targets.
Collapse
|
10
|
De Martinis M, Ginaldi L, Sirufo MM, Bassino EM, De Pietro F, Pioggia G, Gangemi S. IL-33/Vitamin D Crosstalk in Psoriasis-Associated Osteoporosis. Front Immunol 2021; 11:604055. [PMID: 33488605 PMCID: PMC7819870 DOI: 10.3389/fimmu.2020.604055] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
Patients with psoriasis (Pso) and, in particular, psoriatic arthritis (PsoA) have an increased risk of developing osteoporosis (OP). It has been shown that OP is among the more common pathologies associated with Pso, mainly due to the well-known osteopenizing conditions coexisting in these patients. Pso and OP share common risk factors, such as vitamin D deficiency and chronic inflammation. Interestingly, the interleukin (IL)-33/ST2 axis, together with vitamin D, is closely related to both Pso and OP. Vitamin D and the IL-33/ST2 signaling pathways are closely involved in bone remodeling, as well as in skin barrier pathophysiology. The production of anti-osteoclastogenic cytokines, e.g., IL-4 and IL-10, is promoted by IL-33 and vitamin D, which are stimulators of both regulatory and Th2 cells. IL-33, together with other Th2 cytokines, shifts osteoclast precursor differentiation towards macrophage and dendritic cells and inhibits receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastogenesis by regulating the expression of anti-osteoclastic genes. However, while the vitamin D protective functions in OP and Pso have been definitively ascertained, the overall effect of IL-33 on bone and skin homeostasis, because of its pleiotropic action, is still controversial. Emerging evidence suggests a functional link between vitamin D and the IL-33/ST2 axis, which acts through hormonal influences and immune-mediated effects, as well as cellular and metabolic functions. Based on the actions of vitamin D and IL-33 in Pso and OP, here, we hypothesize the role of their crosstalk in the pathogenesis of both these pathologies.
Collapse
Affiliation(s)
- Massimo De Martinis
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Lia Ginaldi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Maria Maddalena Sirufo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Enrica Maria Bassino
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Francesca De Pietro
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), Messina, Italy
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
11
|
De Boeck A, Ahn BY, D'Mello C, Lun X, Menon SV, Alshehri MM, Szulzewsky F, Shen Y, Khan L, Dang NH, Reichardt E, Goring KA, King J, Grisdale CJ, Grinshtein N, Hambardzumyan D, Reilly KM, Blough MD, Cairncross JG, Yong VW, Marra MA, Jones SJM, Kaplan DR, McCoy KD, Holland EC, Bose P, Chan JA, Robbins SM, Senger DL. Glioma-derived IL-33 orchestrates an inflammatory brain tumor microenvironment that accelerates glioma progression. Nat Commun 2020; 11:4997. [PMID: 33020472 PMCID: PMC7536425 DOI: 10.1038/s41467-020-18569-4] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
Despite a deeper molecular understanding, human glioblastoma remains one of the most treatment refractory and fatal cancers. It is known that the presence of macrophages and microglia impact glioblastoma tumorigenesis and prevent durable response. Herein we identify the dual function cytokine IL-33 as an orchestrator of the glioblastoma microenvironment that contributes to tumorigenesis. We find that IL-33 expression in a large subset of human glioma specimens and murine models correlates with increased tumor-associated macrophages/monocytes/microglia. In addition, nuclear and secreted functions of IL-33 regulate chemokines that collectively recruit and activate circulating and resident innate immune cells creating a pro-tumorigenic environment. Conversely, loss of nuclear IL-33 cripples recruitment, dramatically suppresses glioma growth, and increases survival. Our data supports the paradigm that recruitment and activation of immune cells, when instructed appropriately, offer a therapeutic strategy that switches the focus from the cancer cell alone to one that includes the normal host environment.
Collapse
Affiliation(s)
- Astrid De Boeck
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bo Young Ahn
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Charlotte D'Mello
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Xueqing Lun
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shyam V Menon
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mana M Alshehri
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Frank Szulzewsky
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Yaoqing Shen
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Lubaba Khan
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ngoc Ha Dang
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Elliott Reichardt
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kimberly-Ann Goring
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jennifer King
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Cameron J Grisdale
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Natalie Grinshtein
- Department of Molecular Genetics, University of Toronto and Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute and the Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Karlyne M Reilly
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Michael D Blough
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - J Gregory Cairncross
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - David R Kaplan
- Department of Molecular Genetics, University of Toronto and Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Kathy D McCoy
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Eric C Holland
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Pinaki Bose
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jennifer A Chan
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Pathology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Stephen M Robbins
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Donna L Senger
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
12
|
Wang Y, Luo H, Wei M, Becker M, Hyde RK, Gong Q. IL-33/IL1RL1 axis regulates cell survival through the p38 MAPK pathway in acute myeloid leukemia. Leuk Res 2020; 96:106409. [PMID: 32652328 DOI: 10.1016/j.leukres.2020.106409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/13/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) is often characterized by the presence of specific and recurrent chromosomal abnormalities. Current treatments have greatly increased remission rate, but relapse still occurs. Therefore, novel therapeutic approaches are required. Previously, using a conditional Cbfb-MYH11 knockin mouse model, we showed that Cbfb-MYH11 induces the expression of a cytokine receptor, IL1RL1. Treatment with IL-33, the only known ligand of IL1RL1, promotes leukemia cell survival in vitro. We further found that IL1RL1+ cells survive better with chemotherapy than IL1RL1- population. However, the mechanism is not clear. Here, we show that IL-33 treatment decreased drug sensitivity in the human inv(16) AML cell line ME-1. By RT-PCR, we found that IL-33 increased the expression of IL-4 and IL-6 and led to the activation of both p38 MAPK and NF-κB. We also showed that IL-33 decreased apoptosis with increased phosphorylation of p38 MAPK. Moreover, pre-treatment with MAPK inhibitor attenuated the phosphorylation of p38 enhanced by IL-33 and reversed the anti-apoptotic effect by IL-33. Taken together, our findings give news insights into the potential mechanism of the anti-apoptotic effect by IL-33/IL1RL1 axis in AML which will help in future drug development.
Collapse
Affiliation(s)
- Yiqian Wang
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, PR China
| | - Huanmin Luo
- Third Clinical School, Guangzhou Medical University, Guangzhou, PR China
| | - Mengyi Wei
- Nanshan School, Guangzhou Medical University, Guangzhou, PR China
| | - Michelle Becker
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - R Katherine Hyde
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Qing Gong
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, PR China.
| |
Collapse
|
13
|
Capitano ML, Griesenauer B, Guo B, Cooper S, Paczesny S, Broxmeyer HE. The IL-33 Receptor/ST2 acts as a positive regulator of functional mouse bone marrow hematopoietic stem and progenitor cells. Blood Cells Mol Dis 2020; 84:102435. [PMID: 32408242 PMCID: PMC7788514 DOI: 10.1016/j.bcmd.2020.102435] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/19/2022]
Abstract
There is a paucity of information on a potential role for the IL-33 receptor/ST2 in the regulation of mouse bone marrow (BM) hematopoietic stem (HSC) and progenitor (HPC) cells. Comparing the BM of st2−/− and wild type (WT) control mice using functional assays, it was found that st2−/− BM cells had poorer engrafting capacity than WT BM in a competitive repopulating assay using congenic mice, with no changes in reconstitution of B-, T- and myeloid cells following transplantation. The BM of st2−/− mice also had fewer granulocyte-macrophage, erythroid, and multipotential progenitors than that of WT BM and these st2−/− HPC were in a slow cycling state compared to that of the rapidly cycling HPC of the WT mice. While functional assessment of HSC and HPC demonstrated that ST2 has a positive influence on regulation of HSC, we could not pick up differences in st2−/− compared to WT BM using only phenotypic analysis of HSC and HPC populations prior to transplantation, again demonstrating that phenotypic analysis of HSC and HPC do not always recapitulate the functional assessments of these immature hematopoietic cells. ST2 is a positive modulator of hematopoiesis. ST2-/- is a positive modulator of hematopoiesis Immunophenotyping of st2-/- hematopoietic stem cell numbers does not recapitulate functional capability of these cells.
Collapse
Affiliation(s)
- Maegan L Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States of America.
| | - Brad Griesenauer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Bin Guo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Scott Cooper
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Sophie Paczesny
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States of America.
| |
Collapse
|
14
|
Ryan N, Anderson K, Volpedo G, Varikuti S, Satoskar M, Satoskar S, Oghumu S. The IL-33/ST2 Axis in Immune Responses Against Parasitic Disease: Potential Therapeutic Applications. Front Cell Infect Microbiol 2020; 10:153. [PMID: 32363166 PMCID: PMC7180392 DOI: 10.3389/fcimb.2020.00153] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022] Open
Abstract
Parasitic infections pose a wide and varying threat globally, impacting over 25% of the global population with many more at risk of infection. These infections are comprised of, but not limited to, toxoplasmosis, malaria, leishmaniasis and any one of a wide variety of helminthic infections. While a great deal is understood about the adaptive immune response to each of these parasites, there remains a need to further elucidate the early innate immune response. Interleukin-33 is being revealed as one of the earliest players in the cytokine milieu responding to parasitic invasion, and as such has been given the name "alarmin." A nuclear cytokine, interleukin-33 is housed primarily within epithelial and fibroblastic tissues and is released upon cellular damage or death. Evidence has shown that interleukin-33 seems to play a crucial role in priming the immune system toward a strong T helper type 2 immune response, necessary in the clearance of some parasites, while disease exacerbating in the context of others. With the possibility of being a double-edged sword, a great deal remains to be seen in how interleukin-33 and its receptor ST2 are involved in the immune response different parasites elicit, and how those parasites may manipulate or evade this host mechanism. In this review article we compile the current cutting-edge research into the interleukin-33 response to toxoplasmosis, malaria, leishmania, and helminthic infection. Furthermore, we provide insight into directions interleukin-33 research may take in the future, potential immunotherapeutic applications of interleukin-33 modulation and how a better clarity of early innate immune system responses involving interleukin-33/ST2 signaling may be applied in development of much needed treatment options against parasitic invaders.
Collapse
Affiliation(s)
- Nathan Ryan
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Division of Anatomy, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Kelvin Anderson
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Greta Volpedo
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Microbiology, The Ohio State University, Columbus, OH, United States
| | - Sanjay Varikuti
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Monika Satoskar
- Northeast Ohio Medical University, Rootstown, OH, United States
| | - Sanika Satoskar
- Northeast Ohio Medical University, Rootstown, OH, United States
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|