1
|
Nishioka K, Ogino H, Ihara E, Chinen T, Kimura Y, Esaki M, Bai X, Minoda Y, Tanaka Y, Wada M, Hata Y, Ambrosini YM, Ogawa Y. Importance of rectal over colon status in ulcerative colitis remission: the role of microinflammation and mucosal barrier dysfunction in relapse. J Gastroenterol 2025; 60:416-429. [PMID: 39672976 DOI: 10.1007/s00535-024-02199-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a refractory inflammatory disease that affects the rectum and colon, with pivotal involvement of the rectal environment in relapse initiation. This study was conducted in two phases to examine the differences in gene expression between the rectum and colon and to identify relapse factors. METHODS In ***Study 1, RNA sequencing was performed on biopsies from the colon and rectum of patients with active UC, those with remission UC, and controls. In Study 2, the mucosal impedance (MI) values reflecting mucosal barrier function and the mRNA expression of tight junction proteins and inflammatory cytokines were examined in 32 patients with remission UC and 22 controls. Relapse was monitored prospectively. RESULTS In Study 1, comprehensive genetic analysis using RNA sequencing revealed distinct gene profiles in the rectum and sigmoid colon of patients with remission UC. The rectum of these patients exhibited an enriched immune response and apical junction phenotype with persistent upregulation of CLDN2 gene expression. In Study 2, even in patients with remission UC, the MI values in the rectum, but not in the sigmoid colon, were significantly decreased, whereas they were negatively correlated with CLDN2, IL-1β, and IL-6 expressions. CONCLUSION The status of the rectum in patients with remission UC differs from that of the colon, with microinflammation and impaired mucosal barrier function, which are associated with the upregulation of CLDN2, playing a role in relapse.
Collapse
Affiliation(s)
- Kei Nishioka
- Department of Gastroenterology, Saiseikai Futsukaichi Hospital, Chikushino, Japan
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Haruei Ogino
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eikichi Ihara
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Takatoshi Chinen
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Kimura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mitsuru Esaki
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Gastroenterology and Hepatology, Mayo Clinic, Scottsdale, USA
| | - Xiaopeng Bai
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yosuke Minoda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshimasa Tanaka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Wada
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshitaka Hata
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoko M Ambrosini
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, USA
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
2
|
Zhu C, Boucheron N, Al-Rubaye O, Chung BK, Thorbjørnsen LW, Köcher T, Schuster M, Claudel T, Halilbasic E, Kunczer V, Muscate F, Cavanagh LL, Waltenberger D, Lercher A, Ohradanova-Repic A, Schatzlmaier P, Stojakovic T, Scharnagl H, Bergthaler A, Stockinger H, Huber S, Bock C, Kenner L, Karlsen TH, Ellmeier W, Trauner M. 24-Nor-ursodeoxycholic acid improves intestinal inflammation by targeting T H17 pathogenicity and transdifferentiation. Gut 2025:gutjnl-2024-333297. [PMID: 40032499 DOI: 10.1136/gutjnl-2024-333297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND 24-Nor-ursodeoxycholic acid (NorUDCA) is a novel therapeutic bile acid for treating immune-mediated cholestatic liver diseases, such as primary sclerosing cholangitis (PSC). OBJECTIVE Since PSC strongly associates with T helper-type-like 17 (TH17)-mediated intestinal inflammation, we explored NorUDCA's immunomodulatory potential on TH17 cells. DESIGN NorUDCA's impact on TH17 differentiation was assessed using a CD4+TNaive adoptive transfer mouse model, and on intraepithelial TH17 pathogenicity and transdifferentiation using an αCD3 stimulation model combined with interleukin-17A-fate-mapping. Mechanistic studies used molecular and multiomics approaches, flow cytometry and metabolic assays with pathogenic (p) TH17. Pathogenicity of pTH17 exposed to NorUDCA in vitro was evaluated following adoptive transfer in intestinal tissues or the central nervous system (CNS). Key findings were validated in an αCD3-stimulated humanised NSG mouse model reconstituted with peripheral blood mononuclear cells from patients with PSC. RESULTS NorUDCA suppressed TH17 effector function and enriched regulatory T cell (Treg) abundance upon CD4+TNaive cell transfer. NorUDCA mitigated intraepithelial TH17 pathogenicity and decreased the generation of proinflammatory 'TH1-like-TH17' cells, and enhanced TH17 transdifferentiation into Treg and Tr1 (regulatory type 1) cells in the αCD3-model. In vivo ablation revealed that Treg induction is crucial for NorUDCA's anti-inflammatory effect on TH17 pathogenicity. Mechanistically, NorUDCA restrained pTH17 effector function and simultaneously promoted functional Treg formation in vitro, by attenuating a glutamine-mTORC1-glycolysis signalling axis. Exposure of pTH17 to NorUDCA dampened their pathogenicity and expansion in the intestine or CNS upon transfer. NorUDCA's impact on TH17 inflammation was corroborated in the humanised NSG mouse model. CONCLUSION NorUDCA restricts TH17 inflammation in multiple mouse models, potentiating future clinical applications for treating TH17-mediated intestinal diseases and beyond.
Collapse
Affiliation(s)
- Ci Zhu
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Nicole Boucheron
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Osamah Al-Rubaye
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Brian K Chung
- Department of Transplantation Medicine, Clinic of Surgery and Specialized Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Liv Wenche Thorbjørnsen
- Department of Transplantation Medicine, Clinic of Surgery and Specialized Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Thomas Köcher
- Vienna BioCenter Core Facilities, Metabolomics, Vienna BioCenter, Vienna, Austria
| | - Michael Schuster
- Biomedical Sequencing Facility, Cemm, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Emina Halilbasic
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Victoria Kunczer
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Fanziska Muscate
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Lois L Cavanagh
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Darina Waltenberger
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Alexander Lercher
- Cemm, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Anna Ohradanova-Repic
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Philipp Schatzlmaier
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Graz, Graz, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Andreas Bergthaler
- Cemm, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Hannes Stockinger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Bock
- Biomedical Sequencing Facility, Cemm, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Lukas Kenner
- Division of Experimental and Translational Pathology, Department of Pathology, Medical University of Vienna, Vienna, Austria
- Unit of Laboratory Animal Pathology, Department for Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Tom H Karlsen
- Department of Transplantation Medicine, Clinic of Surgery and Specialized Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Wilfried Ellmeier
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Chen G, Ouyang X, Mu Y, Chen Y. Human breast milk-derived exosomes and their positive role on neonatal intestinal health. Pediatr Res 2025:10.1038/s41390-025-03813-8. [PMID: 39865171 DOI: 10.1038/s41390-025-03813-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/24/2024] [Accepted: 12/09/2024] [Indexed: 01/28/2025]
Abstract
Although the role of breast milk in promoting neonatal growth and maintaining intestinal homeostasis is well established, underlying mechanisms by which it protects the intestine from damage remain to be elucidated. Human breast milk-derived exosomes (HMDEs) are newly discovered active signaling vesicles with a diameter of 30-150 nm, which are key carriers of biological information exchange between mother and child. In addition, due to their ability to cross the gastrointestinal barrier, low immunogenicity, good biocompatibility and stability, HMDEs play an important role in regulating intestinal barrier integrity in newborns. In addition, HMDEs possess specific properties that are reformable and modifiable, offering promising strategies for the prevention and treatment of neonatal intestinal diseases. However, challenges such as purification, complex content, and quality control hinder their clinical application. This paper provides a comprehensive review of the biogenesis and properties of HMDEs, their isolation and purification, composition, and effects on neonatal intestinal barrier function, and further explores their potential biomedical applications. IMPACT: Breast milk helps maintain intestinal homeostasis in newborns and can prevent diseases, especially necrotizing enterocolitis (NEC). Breast milk contains abundant exosomes, which are important carriers of maternal and infant biological information exchange. Breast milk have the advantages of low immunogenicity, good biocompatibility and good stability, which helps to maintain the integrity of the intestinal barrier. Exosomes can be modified, which is expected to provide a more effective strategy for the prevention and treatment of intestinal diseases.
Collapse
Affiliation(s)
- Gen Chen
- Department of Pediatrics, The First People's Hospital of Chenzhou, Chenzhou, Hunan, 423000, China
| | - Xiangdong Ouyang
- Department of Pediatrics, The First People's Hospital of Chenzhou, Chenzhou, Hunan, 423000, China
| | - Yide Mu
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510000, China
| | - Yuqiong Chen
- Department of Pediatrics, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
4
|
Chen S, Zhang D, Li D, Zeng F, Chen C, Bai F. Microbiome characterization of patients with Crohn disease and the use of fecal microbiota transplantation: A review. Medicine (Baltimore) 2025; 104:e41262. [PMID: 39854760 PMCID: PMC11771716 DOI: 10.1097/md.0000000000041262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/19/2024] [Accepted: 11/15/2024] [Indexed: 01/26/2025] Open
Abstract
Inflammatory bowel disease is a chronic inflammatory condition predominantly affecting the intestines, encompassing both ulcerative colitis and Crohn disease (CD). As one of the most common gastrointestinal disorders, CD's pathogenesis is closely linked with the intestinal microbiota. Recently, fecal microbiota transplantation (FMT) has gained attention as a potential treatment for CD, with the effective reestablishment of intestinal microecology considered a crucial mechanism of FMT therapy. This article synthesizes the findings of population-based cohort studies to enhance our understanding of gut microbial characteristics in patients with CD. It delves into the roles of "beneficial" and "pathogenic" bacteria in CD's development. This article systematically reviews and compares data on clinical response rates, remission rates, adverse events, and shifts in bacterial microbiota. Among these studies, gut microbiome analysis was conducted in only 7, and a single study examined the metabolome. Overall, FMT has demonstrated a partial restoration of typical CD-associated microbiological alterations, leading to increased α-diversity in responders and a moderate shift in patient microbiota toward the donor profile. Several factors, including donor selection, delivery route, microbial state (fresh or frozen), and recipient condition, are identified as pivotal in influencing FMT's effectiveness. Future prospective clinical studies with larger patient cohorts and improved methodologies are imperative. In addition, standardization of FMT procedures, coupled with advanced genomic techniques such as macroproteomics and culture genomics, is necessary. These advancements will further clarify the bacterial microbiota alterations that significantly contribute to FMT's therapeutic effects in CD treatment, as well as elucidate the underlying mechanisms of action.
Collapse
Affiliation(s)
- Shiju Chen
- Graduate School, Hainan Medical University, Haikou, China
| | - Daya Zhang
- Graduate School, Hainan Medical University, Haikou, China
| | - Da Li
- Graduate School, Hainan Medical University, Haikou, China
| | - Fan Zeng
- Graduate School, Hainan Medical University, Haikou, China
| | - Chen Chen
- Graduate School, Hainan Medical University, Haikou, China
| | - Feihu Bai
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- The Gastroenterology Clinical Medical Center of Hainan Province, Haikou, China
| |
Collapse
|
5
|
Lu Y, Xiong Y, Zhang S, Wang B, Feng Y, Pu Z, Wei K, Chen J, Chen D, Zhang P. D-mannose reduces oxidative stress, inhibits inflammation, and increases treg cell proportions in mice with ulcerative colitis. Front Pharmacol 2024; 15:1454713. [PMID: 39555100 PMCID: PMC11563948 DOI: 10.3389/fphar.2024.1454713] [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: 06/25/2024] [Accepted: 10/18/2024] [Indexed: 11/19/2024] Open
Abstract
Background Regulatory T (Treg) cells is required to dampen immune responses against intestinal microbiota, which aid in a healthy body to promise that the resident gut microbiota should not attract the attention of the immune system. Inflammation and inflammatory bowel disease (IBD) can be induced if the immune system fails to ignore the resident gut microbiota and targets them instead. D-mannose, a common monosaccharide in nature, has been shown to ameliorate multiple autoimmune diseases. This study aimed to investigate the therapeutic effect of D-mannose on mice ulcerative colitis (UC) induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS), and elucidate its underlying mechanisms. Methods To simulate human IBD, we constructed a mouse model of UC by injecting TNBS into the colon. Results Our results demonstrated that D-mannose treatment effectively alleviated TNBS-induced UC in mice, as evidenced by the amelioration of UC symptoms. D-mannose treatment significantly reduced inflammation by decreasing the expression of proinflammatory cytokines and inflammation mediators. D-mannose treatment also significantly inhibited oxidative stress, promoted the expression of GSH and SOD, decreased the expression of MDA. Mechanistically, D-mannose upregulated the proportion of both CD4(+) Tregs and CD8(+) Tregs. Conclusion In summary, our study provides the first evidence of the therapeutic effect of D-mannose on mice with UC, which is likely mediated by upregulating Treg proportions.
Collapse
Affiliation(s)
- Yuqing Lu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Yongjian Xiong
- Central Laboratory, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shuangshuang Zhang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Boya Wang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Yuntao Feng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhuonan Pu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Kun Wei
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Jun Chen
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Dapeng Chen
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Peng Zhang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
6
|
Xiang Z, Zhang B, Cao S, Cao L, Li L, Huang D, Li Q, Chen Y, Gong X, Zhang X, Li R, Wu J, Peng Y, Huo G, Xu L, Zhang Z, Li D, Xia G. SPH7854, a gut-limited RORγt antagonist, ameliorates TNBS-induced experimental colitis in rat. Int Immunopharmacol 2024; 140:112884. [PMID: 39133959 DOI: 10.1016/j.intimp.2024.112884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 09/01/2024]
Abstract
Multiple lines of evidence suggest that Retinoic Acid Related Orphan Nuclear Receptor gamma t (RORγt) is a potent therapeutic target for inflammatory bowel disease (IBD). However, systemic blockade of RORγt easily leads to thymic lymphoma and aberrant liver function. Therefore, the development of gut-limited RORγt antagonists may lead to the development of innovative IBD therapeutics that improve safety and retain effectiveness. We discovered SPH7854, a potent and selective RORγt antagonist. The effect of SPH7854 on the differentiation of T helper 1 (Th1)/Th17/regulatory T (Treg) cells was evaluated in mouse and human primary cells. SPH7854 (2-(4-(ethylsulfonyl)phenyl)-N- (6-(2-methyl-2-(pyridin-2-yl) propanoyl)pyridin-3-yl)acetamide) dose-dependently inhibited interleukin-17A (IL-17A) secretion from mouse CD4 + T cells and human peripheral blood mononuclear cells (PBMC). Additionally, SPH7854 strongly suppressed Th17 cell differentiation and considerably promoted Treg cell differentiation while slightly affected Th1 cell differentiation from mouse CD4 + T cells. The pharmacokinetic (PK) studies indicated that SPH7854 was restricted to the gut: the bioavailability and maximal plasma concentration of SPH7854 after oral administration (6 mg/kg) were 1.24 ± 0.33 % and 4.92 ± 11.81 nM, respectively, in rats. Strikingly, oral administration of SPH7854 (5 mg/kg and 15 mg/kg) twice daily significantly alleviated 2, 4, 6-trinitrobenzensulfonic acid (TNBS)-induced colitis in rats. SPH7854, especially at 15 mg/kg, significantly alleviated symptoms and improved macroscopic signs and microscopic structure in rat colitis, with decreased colonic mucosal levels of IL-17A, IL-6, tumor necrosis factor α (TNFα), monocyte chemoattractant protein-1 (MCP-1) and myeloperoxidase (MPO). These evidences indicated that blockade of RORγt activity via a gut-limited antagonist may be an effective and safe therapeutic strategy for IBD treatment.
Collapse
MESH Headings
- Animals
- Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Humans
- Trinitrobenzenesulfonic Acid
- Colitis/chemically induced
- Colitis/drug therapy
- Colitis/immunology
- Male
- Rats
- Mice
- Th17 Cells/immunology
- Th17 Cells/drug effects
- Rats, Sprague-Dawley
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/drug effects
- Cell Differentiation/drug effects
- Disease Models, Animal
- Interleukin-17/metabolism
- Interleukin-17/antagonists & inhibitors
- Acetamides/therapeutic use
- Acetamides/pharmacology
- Cells, Cultured
- Anti-Inflammatory Agents/therapeutic use
- Anti-Inflammatory Agents/pharmacology
- Colon/drug effects
- Colon/pathology
- Colon/immunology
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Zhijun Xiang
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Bingbin Zhang
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Shuangyi Cao
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Long Cao
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Lingwen Li
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Dehua Huang
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Qian Li
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Yuxiang Chen
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Xuelian Gong
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Xiaohong Zhang
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Ruizhi Li
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Jinmiao Wu
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Yayuan Peng
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Guoyong Huo
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Lixia Xu
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Zhihui Zhang
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Di Li
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| | - Guangxin Xia
- Central Research Institute, National Key Laboratory of Innovative Immunotherapy, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai 201203, PR China.
| |
Collapse
|
7
|
Chen X, Zhang M, Zhou F, Gu Z, Li Y, Yu T, Peng C, Zhou L, Li X, Zhu D, Zhang X, Yu C. SIRT3 Activator Honokiol Inhibits Th17 Cell Differentiation and Alleviates Colitis. Inflamm Bowel Dis 2023; 29:1929-1940. [PMID: 37335900 PMCID: PMC10697418 DOI: 10.1093/ibd/izad099] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Indexed: 06/21/2023]
Abstract
BACKGROUND Honokiol (HKL), a natural extract of the bark of the magnolia tree and an activator of the mitochondrial protein sirtuin-3 (SIRT3), has been proposed to possess anti-inflammatory effects. This study investigated the inhibitory effects of HKL on T helper (Th) 17 cell differentiation in colitis. METHODS Serum and biopsies from 20 participants with ulcerative colitis (UC) and 18 healthy volunteers were collected for the test of serum cytokines, flow cytometry analysis (FACS), and relative messenger RNA (mRNA) levels of T cell subsets, as well as the expression of SIRT3 and phosphorylated signal transducer and activator of transcription/retinoic acid-related orphan nuclear receptor γt (p-STAT3/RORγt) signal pathway in colon tissues. In vitro, naïve clusters of differentiation (CD) 4 + T cells isolated from the mouse spleen differentiated to subsets including Th1, Th2, Th17, and regulatory T (Treg) cells. Peripheral blood monocytes (PBMCs) from healthy volunteers were induced to the polarization of Th17 cells. After HKL treatment, changes in T cell subsets, related cytokines, and transcription factors were measured. The dextran sulfate sodium (DSS)-induced colitis and interleukin (IL)-10-deficient mice were intraperitoneally injected with HKL. These experiments were conducted to study the effect of HKL on the development, cytokines, and expression of signaling pathway proteins in colitis. RESULTS Patients with UC had higher serum IL-17 and a higher proportion of Th17 differentiation in blood compared with healthy participants; while IL-10 level and the proportion of Treg cells were lower. Higher relative mRNA levels of RORγt and a lower SIRT3 expression in colon tissues were observed. In vitro, HKL had little effect on the differentiation of naïve CD4+ T cells to Th1, Th2, or Treg cells, but it downregulated IL-17 levels and the Th17 cell ratio in CD4+ T cells from the mouse spleen and human PBMCs under Th17 polarization. Even with a STAT3 activator, HKL still significantly inhibited IL-17 levels. In DSS-induced colitis mice and IL-10 deficient mice treated with HKL, the length of the colon, weight loss, disease activity index, and histopathological scores were improved, IL-17 and IL-21 levels, and the proportion of Th17 cells were decreased. Sirtuin-3 expression was increased, whereas STAT3 phosphorylation and RORγt expression were inhibited in the colon tissue of mice after HKL treatment. CONCLUSIONS Our study demonstrated that HKL could partially protect against colitis by regulating Th17 differentiation through activating SIRT3, leading to inhibition of the STAT3/RORγt signaling pathway. These results provide new insights into the protective effects of HKL against colitis and may facilitate the research of new drugs for inflammatory bowel disease.
Collapse
Affiliation(s)
- Xiaotian Chen
- Department of Clinical Nutrition, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, P.R. China
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, P.R. China
| | - Mingming Zhang
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai 200001, P.R. China
| | - Fan Zhou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Zhengrong Gu
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210017, P.R. China
| | - Yuan Li
- Department of Clinical Nutrition, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, P.R. China
| | - Ting Yu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Chunyan Peng
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Lixing Zhou
- The Center of Gerontology and Geriatrics/National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Xiangrui Li
- Department of Clinical Nutrition, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, P.R. China
| | - Dandan Zhu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Xiaoqi Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Chenggong Yu
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, P.R. China
| |
Collapse
|
8
|
Niu Y, Zhang J, Shi D, Zang W, Niu J. Glycosides as Potential Medicinal Components for Ulcerative Colitis: A Review. Molecules 2023; 28:5210. [PMID: 37446872 DOI: 10.3390/molecules28135210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic, non-specific disease of unknown etiology. The disease develops mainly in the rectum or colon, and the main clinical symptoms include abdominal pain, diarrhea, and purulent bloody stools, with a wide variation in severity. The specific causative factors and pathogenesis of the disease are not yet clear, but most scholars believe that the disease is caused by the interaction of genetic, environmental, infectious, immune, and intestinal flora factors. As for the treatment of UC, medications are commonly used in clinical practice, mainly including aminosalicylates, glucocorticoids, and immunosuppressive drugs. However, due to the many complications associated with conventional drug therapy and the tendency for UC to recur, there is an urgent need to discover new, safer, and more effective drugs. Natural compounds with biodiversity and chemical structure diversity from medicinal plants are the most reliable source for the development of new drug precursors. Evidence suggests that glycosides may reduce the development and progression of UC by modulating anti-inflammatory responses, inhibiting oxidative stress, suppressing abnormal immune responses, and regulating signal transduction. In this manuscript, we provide a review of the epidemiology of UC and the available drugs for disease prevention and treatment. In addition, we demonstrate the protective or therapeutic role of glycosides in UC and describe the possible mechanisms of action to provide a theoretical basis for preclinical studies in drug development.
Collapse
Affiliation(s)
- Yating Niu
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| | - Jun Zhang
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Dianhua Shi
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Weibiao Zang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Jianguo Niu
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
9
|
Two cases of ulcerative colitis that developed while using abatacept. Clin J Gastroenterol 2022; 15:924-928. [DOI: 10.1007/s12328-022-01653-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023]
|
10
|
Eiro N, Fraile M, González-Jubete A, González LO, Vizoso FJ. Mesenchymal (Stem) Stromal Cells Based as New Therapeutic Alternative in Inflammatory Bowel Disease: Basic Mechanisms, Experimental and Clinical Evidence, and Challenges. Int J Mol Sci 2022; 23:ijms23168905. [PMID: 36012170 PMCID: PMC9408403 DOI: 10.3390/ijms23168905] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are an example of chronic diseases affecting 40% of the population, which involved tissue damage and an inflammatory process not satisfactorily controlled with current therapies. Data suggest that mesenchymal stem cells (MSC) may be a therapeutic option for these processes, and especially for IBD, due to their multifactorial approaches such as anti-inflammatory, anti-oxidative stress, anti-apoptotic, anti-fibrotic, regenerative, angiogenic, anti-tumor, or anti-microbial. However, MSC therapy is associated with important limitations as safety issues, handling difficulties for therapeutic purposes, and high economic cost. MSC-derived secretome products (conditioned medium or extracellular vesicles) are therefore a therapeutic option in IBD as they exhibit similar effects to their parent cells and avoid the issues of cell therapy. In this review, we proposed further studies to choose the ideal tissue source of MSC to treat IBD, the implementation of new standardized production strategies, quality controls and the integration of other technologies, such as hydrogels, which may improve the therapeutic effects of derived-MSC secretome products in IBD.
Collapse
Affiliation(s)
- Noemi Eiro
- Research Unit, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-98-5320050 (ext. 84216) (N.E.); Fax: +34-98-531570 (N.E.)
| | - Maria Fraile
- Research Unit, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
| | | | - Luis O. González
- Department of Anatomical Pathology, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
| | - Francisco J. Vizoso
- Research Unit, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
- Department of Surgery, Fundación Hospital de Jove, Av. de Eduardo Castro, 161, 33290 Gijón, Spain
- Correspondence: (N.E.); (F.J.V.); Tel.: +34-98-5320050 (ext. 84216) (N.E.); Fax: +34-98-531570 (N.E.)
| |
Collapse
|
11
|
Sato M, Fujii K, Takagi H, Shibuya I, Oka D, Yamaya N, Hagita H, Matsumoto M, Inagaki K. Antibacterial and Immunosuppressive Effects of OPS-2071, a Candidate Therapy for Inflammatory Bowel Disease. Dig Dis Sci 2022; 67:3993-4007. [PMID: 34463880 DOI: 10.1007/s10620-021-07237-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/19/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the gastrointestinal tract. Although many types of drug are used, clinical outcomes are still unsatisfactory. Previous studies have suggested that intestinal bacteria are involved in the pathogenesis of IBD. Accordingly, in an IBD model we evaluated the therapeutic effects of OPS-2071, a low-absorption quinolone antibacterial agent indicated for intestinal infection, and investigated its mechanism of action. METHODS The therapeutic effects of OPS-2071 and comparison therapies were evaluated using naive CD4 + T cell-transfer IBD model mice. In vitro inhibition of LPS-induced TNF-α production and inhibitory effects on T cell responses stimulated using anti-CD3/CD28 antibody-loaded beads were evaluated using mouse splenocytes and human peripheral blood mononuclear cells. In addition, in vitro activities against bacteria implicated in IBD pathogenesis were tested. RESULTS OPS-2071 dose-dependently decreased both colonic weight/length ratio and the colitis histological score as compared with the vehicle group. The therapeutic effect of OPS-2071 was equivalent to that of anti-IL-12/23 (p40) antibody. In vitro, OPS-2071 suppressed TNF-α production induced by LPS stimulation and T cell responses in a dose-dependent manner. At high concentrations, these effects were comparable to those of existing immunosuppressive agents, such as prednisolone, in both mouse and human cells. OPS-2071 also showed antibacterial activity against IBD-related bacteria. CONCLUSIONS Our results suggest that OPS-2071 had both immunosuppressive and antibacterial effects. This dual effect makes OPS-2071 a unique and promising candidate for IBD.
Collapse
Affiliation(s)
- Masayoshi Sato
- Infectious Diseases Unit, Department of Medical Innovations, New Drug Research Division, Otsuka Pharmaceutical Co., Ltd, 463-10, Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima, 771-0192, Japan
| | - Kazuyuki Fujii
- Infectious Diseases Unit, Department of Medical Innovations, New Drug Research Division, Otsuka Pharmaceutical Co., Ltd, 463-10, Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima, 771-0192, Japan
| | - Hiroko Takagi
- Infectious Diseases Unit, Department of Medical Innovations, New Drug Research Division, Otsuka Pharmaceutical Co., Ltd, 463-10, Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima, 771-0192, Japan.
| | - Isao Shibuya
- Infectious Diseases Unit, Department of Medical Innovations, New Drug Research Division, Otsuka Pharmaceutical Co., Ltd, 463-10, Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima, 771-0192, Japan
| | - Daisuke Oka
- Infectious Diseases Unit, Department of Medical Innovations, New Drug Research Division, Otsuka Pharmaceutical Co., Ltd, 463-10, Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima, 771-0192, Japan
| | - Naomitsu Yamaya
- Department of Drug Metabolism and Pharmacokinetics, Otsuka Pharmaceutical Co., Ltd, 463-10, Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima, 771-0192, Japan
| | - Hiraku Hagita
- Department of Drug Metabolism and Pharmacokinetics, Otsuka Pharmaceutical Co., Ltd, 463-10, Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima, 771-0192, Japan
| | - Makoto Matsumoto
- Pharmaceutical Business Division, Otsuka Pharmaceutical Co., Ltd, Shinagawa Grand Central Tower, 2-16-4 Konan, Minato-ku, Tokyo, 108-8242, Japan
| | - Katsuya Inagaki
- Infectious Diseases Unit, Department of Medical Innovations, New Drug Research Division, Otsuka Pharmaceutical Co., Ltd, 463-10, Kagasuno, Kawauchi-cho, Tokushima-shi, Tokushima, 771-0192, Japan
| |
Collapse
|
12
|
Duncan K, Carey-Ewend K, Vaishnava S. Spatial analysis of gut microbiome reveals a distinct ecological niche associated with the mucus layer. Gut Microbes 2022; 13:1874815. [PMID: 33567985 PMCID: PMC8253138 DOI: 10.1080/19490976.2021.1874815] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Mucus-associated bacterial communities are critical for determining disease pathology and promoting colonization resistance. Yet the key ecological properties of mucus resident communities remain poorly defined. Using an approach that combines in situ hybridization, laser microdissection and 16s rRNA sequencing of spatially distinct regions of the mouse gut lumen, we discovered that a dense microbial community resembling a biofilm is embedded in the mucus layer. The mucus-associated biofilm-like community excluded bacteria belonging to phylum Proteobacteria. Additionally, it was significantly more diverse and consisted of bacterial species that were unique to it. By employing germ-free mice deficient in T and B lymphocytes we found that formation of biofilm-like structure was independent of adaptive immunity. Instead the integrity of biofilm-like community depended on Gram-positive commensals such as Clostridia. Additionally, biofilm-like community in the mucus lost fewer Clostridia and showed smaller bloom of Proteobacteria compared to the lumen upon antibiotic treatment. When subjected to time-restricted feeding biofilm-like structure significantly enhanced in size and showed enrichment of Clostridia. Taken together our work discloses that mucus-associated biofilm-like community represents a specialized community that is structurally and compositionally distinct that excludes aerobic bacteria while enriching for anaerobic bacteria such as Clostridia, exhibits enhanced stability to antibiotic treatment and that can be modulated by dietary changes.
Collapse
Affiliation(s)
- Kellyanne Duncan
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Kelly Carey-Ewend
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Shipra Vaishnava
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States,CONTACT Shipra Vaishnava Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, United States
| |
Collapse
|
13
|
Noda M, Danshiitsoodol N, Kanno K, Uchida T, Sugiyama M. The Exopolysaccharide Produced by Lactobacillus paracasei IJH-SONE68 Prevents and Ameliorates Inflammatory Responses in DSS-Induced Ulcerative Colitis. Microorganisms 2021; 9:microorganisms9112243. [PMID: 34835369 PMCID: PMC8621803 DOI: 10.3390/microorganisms9112243] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory bowel disease (IBD) is an autoimmune disease characterized by chronic inflammation of the gastrointestinal tract. IBD includes Crohn’s disease (CD) and ulcerative colitis (UC). CD can occur in any part of the gastrointestinal tract, whereas UC mainly occurs in the colon and rectum. We previously demonstrated that a novel exopolysaccharide (EPS) produced by a plant-derived bacterium, Lactobacillus paracasei IJH-SONE68, prevents and improves the inflammation in contact dermatitis model mice via oral administration. To evaluate the preventive effect of the EPS against other inflammatory diseases, in the present study, we employed dextran sulfate sodium (DSS)-induced UC model mice. The stool consistency, hematochezia, and colonic atrophy of the mice were improved by the orally administered EPS. We also evaluated the cytokine transcription. Overexpression of the mouse macrophage inflammatory protein 2 mRNA in the colon as a functional homolog of human interleukin-8 was decreased by the orally administered EPS. However, the expression of interleukin-10, which is known as an anti-inflammatory cytokine, was stimulated in the EPS-administrated group. Based on these results, we conclude that the IJH-SONE68-derived EPS is a promising lead material for the development of drugs useful in treating inflammatory diseases such as UC.
Collapse
Affiliation(s)
- Masafumi Noda
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.N.); (N.D.)
| | - Narandalai Danshiitsoodol
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.N.); (N.D.)
| | - Keishi Kanno
- Department of General Internal Medicine, Hiroshima University Hospital, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan;
- Department of Clinical Pharmaceutical and Therapeutics, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan;
| | - Tomoyuki Uchida
- Department of Clinical Pharmaceutical and Therapeutics, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan;
- Sone Farm Co., Ltd., Shinjuku, Shinjuku-ku, Tokyo 160-0022, Japan
| | - Masanori Sugiyama
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.N.); (N.D.)
- Correspondence: ; Tel.: +81-82-257-5280
| |
Collapse
|
14
|
Aarts J, Boleij A, Pieters BCH, Feitsma AL, van Neerven RJJ, Ten Klooster JP, M'Rabet L, Arntz OJ, Koenders MI, van de Loo FAJ. Flood Control: How Milk-Derived Extracellular Vesicles Can Help to Improve the Intestinal Barrier Function and Break the Gut-Joint Axis in Rheumatoid Arthritis. Front Immunol 2021; 12:703277. [PMID: 34394100 PMCID: PMC8356634 DOI: 10.3389/fimmu.2021.703277] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Many studies provided compelling evidence that extracellular vesicles (EVs) are involved in the regulation of the immune response, acting as both enhancers and dampeners of the immune system, depending on the source and type of vesicle. Research, including ours, has shown anti-inflammatory effects of milk-derived EVs, using human breast milk as well as bovine colostrum and store-bought pasteurized cow milk, in in vitro systems as well as therapeutically in animal models. Although it is not completely elucidated which proteins and miRNAs within the milk-derived EVs contribute to these immunosuppressive capacities, one proposed mechanism of action of the EVs is via the modulation of the crosstalk between the (intestinal) microbiome and their host health. There is increasing awareness that the gut plays an important role in many inflammatory diseases. Enhanced intestinal leakiness, dysbiosis of the gut microbiome, and bowel inflammation are not only associated with intestinal diseases like colitis and Crohn's disease, but also characteristic for systemic inflammatory diseases such as lupus, multiple sclerosis, and rheumatoid arthritis (RA). Strategies to target the gut, and especially its microbiome, are under investigation and hold a promise as a therapeutic intervention for these diseases. The use of milk-derived EVs, either as stand-alone drug or as a drug carrier, is often suggested in recent years. Several research groups have studied the tolerance and safety of using milk-derived EVs in animal models. Due to its composition, milk-derived EVs are highly biocompatible and have limited immunogenicity even cross species. Furthermore, it has been demonstrated that milk-derived EVs, when taken up in the gastro-intestinal tract, stay intact after absorption, indicating excellent stability. These characteristics make milk-derived EVs very suitable as drug carriers, but also by themselves, these EVs already have a substantial immunoregulatory function, and even without loading, these vesicles can act as therapeutics. In this review, we will address the immunomodulating capacity of milk-derived EVs and discuss their potential as therapy for RA patients. Review criteria The search terms "extracellular vesicles", "exosomes", "microvesicles", "rheumatoid arthritis", "gut-joint axis", "milk", and "experimental arthritis" were used. English-language full text papers (published between 1980 and 2021) were identified from PubMed and Google Scholar databases. The reference list for each paper was further searched to identify additional relevant articles.
Collapse
Affiliation(s)
- Joyce Aarts
- Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center (Radboudumc), Nijmegen, Netherlands
| | - Annemarie Boleij
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center (Radboudumc), Nijmegen, Netherlands
| | - Bartijn C H Pieters
- Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center (Radboudumc), Nijmegen, Netherlands
| | | | - R J Joost van Neerven
- FrieslandCampina, Amersfoort, Netherlands.,Cell Biology and Immunology, Wageningen University & Research, Wageningen, Netherlands
| | - Jean Paul Ten Klooster
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, Netherlands
| | - Laura M'Rabet
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, Netherlands
| | - Onno J Arntz
- Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center (Radboudumc), Nijmegen, Netherlands
| | - Marije I Koenders
- Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center (Radboudumc), Nijmegen, Netherlands
| | - Fons A J van de Loo
- Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center (Radboudumc), Nijmegen, Netherlands
| |
Collapse
|
15
|
Negi S, Saini S, Tandel N, Sahu K, Mishra RP, Tyagi RK. Translating Treg Therapy for Inflammatory Bowel Disease in Humanized Mice. Cells 2021; 10:1847. [PMID: 34440615 PMCID: PMC8393385 DOI: 10.3390/cells10081847] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
Crohn's disease and ulcerative colitis, two major forms of inflammatory bowel disease (IBD) in humans, afflicted in genetically predisposed individuals due to dysregulated immune response directed against constituents of gut flora. The defective immune responses mounted against the regulatory mechanisms amplify and maintain the IBD-induced mucosal inflammation. Therefore, restoring the balance between inflammatory and anti-inflammatory immunepathways in the gut may contribute to halting the IBD-associated tissue-damaging immune response. Phenotypic and functional characterization of various immune-suppressive T cells (regulatory T cells; Tregs) over the last decade has been used to optimize the procedures for in vitro expansion of these cells for developing therapeutic interventional strategies. In this paper, we review the mechanisms of action and functional importance of Tregs during the pathogenesis of IBD and modulating the disease induced inflammation as well as role of mouse models including humanized mice repopulated with the human immune system (HIS) to study the IBD. "Humanized" mouse models provide new tools to analyze human Treg ontogeny, immunobiology, and therapy and the role of Tregs in developing interventional strategies against IBD. Overall, humanized mouse models replicate the human conditions and prove a viable tool to study molecular functions of human Tregs to harness their therapeutic potential.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Colitis, Ulcerative/genetics
- Colitis, Ulcerative/immunology
- Colitis, Ulcerative/metabolism
- Colitis, Ulcerative/therapy
- Crohn Disease/genetics
- Crohn Disease/immunology
- Crohn Disease/metabolism
- Crohn Disease/therapy
- Disease Models, Animal
- Hematopoietic Stem Cell Transplantation
- Humans
- Mice, Transgenic
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Sushmita Negi
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
- BERPDC Department, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Sheetal Saini
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
| | - Nikunj Tandel
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India;
| | - Kiran Sahu
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
| | - Ravi P.N. Mishra
- BERPDC Department, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India
| | - Rajeev K. Tyagi
- Biomedical Parasitology and Nano-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; (S.N.); (S.S.); (K.S.)
| |
Collapse
|
16
|
Van Spaendonk H, Ceuleers H, Smet A, Berg M, Joossens J, Van der Veken P, Francque SM, Lambeir AM, De Man JG, De Meester I, Augustyns K, De Winter BY. The Effect of a Novel Serine Protease Inhibitor on Inflammation and Intestinal Permeability in a Murine Colitis Transfer Model. Front Pharmacol 2021; 12:682065. [PMID: 34248633 PMCID: PMC8264366 DOI: 10.3389/fphar.2021.682065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
Background: A protease/antiprotease disbalance is observed in inflammatory bowel diseases (IBD). We therefore studied the effect of the novel serine protease inhibitor UAMC-00050 on intestinal inflammation and permeability in a chronic colitis T cell transfer mouse model to get further insight into the regulation of T cell-mediated immunopathology. Methods: Colitis was induced in severe combined immunodeficient (SCID) mice, by the adoptive transfer of CD4+CD25-CD62L+ T cells. Animals were treated intraperitoneally (i.p.) 2x/day with vehicle or UAMC-00050 (5 mg/kg) from week 2 onwards. Colonic inflammation was assessed by clinical parameters, colonoscopy, macroscopy, microscopy, myeloperoxidase activity and cytokine expression levels. At week 4, 4 kDa FITC-dextran intestinal permeability was evaluated and T helper transcription factors, protease-activated receptors and junctional proteins were quantified by RT-qPCR. Results: Adoptive transfer of CD4+CD25-CD62L+ T cells resulted in colonic inflammation and an altered intestinal permeability. The serine protease inhibitor UAMC-00050 ameliorated both the inflammatory parameters and the intestinal barrier function. Furthermore, a decrease in colonic mRNA expression of Tbet and PAR4 was observed in colitis mice after UAMC-00050 treatment. Conclusion: The beneficial effect of UAMC-00050 on inflammation was apparent via a reduction of Tbet, IFN-γ, TNF-α, IL-1β and IL-6. Based on these results, we hypothesize a pivotal effect of serine protease inhibition on the Th1 inflammatory profile potentially mediated via PAR4.
Collapse
Affiliation(s)
- Hanne Van Spaendonk
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Hannah Ceuleers
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Annemieke Smet
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Maya Berg
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Jurgen Joossens
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Pieter Van der Veken
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Sven M. Francque
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
| | - Anne-Marie Lambeir
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Joris G. De Man
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Ingrid De Meester
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Koen Augustyns
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Benedicte Y. De Winter
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
17
|
Extracellular Vesicles from Thapsigargin-Treated Mesenchymal Stem Cells Ameliorated Experimental Colitis via Enhanced Immunomodulatory Properties. Biomedicines 2021; 9:biomedicines9020209. [PMID: 33670708 PMCID: PMC7922639 DOI: 10.3390/biomedicines9020209] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 02/08/2023] Open
Abstract
Therapeutic applications of extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have attracted considerable attention because of their immunomodulatory properties against immune-mediated, inflammatory diseases. Here, we demonstrated enhanced immunomodulatory properties of EVs secreted from endoplasmic reticulum (ER) stress inducer thapsigargin (TSG)-primed human Wharton's jelly-derived MSCs (WJ-MSCs). EVs from TSG-primed WJ-MSCs (TSG-EV) showed increased yield and expression of immunomodulatory factors, such as transforming growth factor-β1 (TGFβ), cyclooxygenase-2 (COX2), and especially indoleamine 2,3-dioxygenase (IDO), compared to control EVs. TSG-EV showed a significantly enhanced immunosuppressive effect on human peripheral blood-derived T cell proliferation and Th1 and Th17 differentiation, whereas Treg and M2-type macrophage were enriched compared to a control EV-treated group. Furthermore, TSG-EV substantially mitigated mouse experimental colitis by reducing the inflammatory response and maintaining intestinal barrier integrity. A significant increase of Tregs and M2-type macrophages in colitic colons of a TSG-EV-treated mouse suggests an anti-inflammatory effect of TSG-EV in colitis model, possibly mediated by Treg and macrophage polarization. These data indicate that TSG treatment promoted immunomodulatory properties of EVs from WJ-MSCs, and TSG-EV may provide a new therapeutic approach for treatment of colitis.
Collapse
|
18
|
Leylabadlo HE, Ghotaslou R, Feizabadi MM, Farajnia S, Moaddab SY, Ganbarov K, Khodadadi E, Tanomand A, Sheykhsaran E, Yousefi B, Kafil HS. The critical role of Faecalibacterium prausnitzii in human health: An overview. Microb Pathog 2020; 149:104344. [PMID: 32534182 DOI: 10.1016/j.micpath.2020.104344] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022]
Abstract
Faecalibacterium prausnitzii (F. prausnitzii) is one of the most abundant bacterial species in the colon of healthy human adults and representing more than 5% of the total bacterial population. Recently, it has been known as a major actor in human intestinal health and a biosensor. Changes in this species population richness and quantity have been observed in many illnesses and several investigations have reported that abundance of F. prausnitzii is reduced in different intestinal disorders. In the current review, we aim to consider literature from various library databases and electronic searches (Science Direct, PubMed, and Google Scholar) which were randomly collected and serve as an overview of different features of F. prausnitzii including metabolites, anti-inflammatory action, and correlation of dysbiosis of this bacterium with various complications in human.
Collapse
Affiliation(s)
- Hamed Ebrahimzadeh Leylabadlo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Reza Ghotaslou
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Mehdi Feizabadi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Safar Farajnia
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Seyed Yaghoub Moaddab
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Ehsaneh Khodadadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Asghar Tanomand
- Department of Microbiology, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Elham Sheykhsaran
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Bahman Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hossein Samadi Kafil
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
19
|
Wei C, Wang JY, Xiong F, Wu BH, Luo MH, Yu ZC, Liu TT, Li DF, Tang Q, Li YX, Zhang DG, Xu ZL, Jin HT, Wang LS, Yao J. Curcumin ameliorates DSS‑induced colitis in mice by regulating the Treg/Th17 signaling pathway. Mol Med Rep 2020; 23:34. [PMID: 33179078 PMCID: PMC7684861 DOI: 10.3892/mmr.2020.11672] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 09/19/2019] [Indexed: 12/26/2022] Open
Abstract
Curcumin has a therapeutic effect on ulcerative colitis, but the underlying mechanism has yet to be elucidated. The aim of the present study was to clarify the possible mechanisms. Dextran sulfate sodium-induced colitis mice were treated with curcumin via gavage for 7 days. The effects of curcumin on disease activity index (DAI) and pathological changes of colonic tissue in mice were determined. Interleukin (IL)-6, IL-10, IL-17 and IL-23 expression levels were measured by ELISA. Flow cytometry was used to detect the ratio of mouse spleen regulatory T cells (Treg)/Th17 cells, and western blotting was used to measure the nuclear protein hypoxia inducible factor (HIF)-1α level. The results demonstrated that curcumin can significantly reduce DAI and spleen index scores and improve mucosal inflammation. Curcumin could also regulate the re-equilibration of Treg/Th17. IL-10 level in the colon was significantly increased, while inflammatory cytokines IL-6, IL-17 and IL-23 were significantly reduced following curcumin treatment. No significant difference in HIF-1α was observed between the colitis and the curcumin group. It was concluded that oral administration of curcumin can effectively treat experimental colitis by regulating the re-equilibration of Treg/Th17 and that the regulatory mechanism may be closely related to the IL-23/Th17 pathway. The results of the present study provided molecular insight into the mechanism by which curcumin treats ulcerative colitis.
Collapse
Affiliation(s)
- Cheng Wei
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Jian-Yao Wang
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, Guangdong 518026, P.R. China
| | - Feng Xiong
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Ben-Hua Wu
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Ming-Han Luo
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Zhi-Chao Yu
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Ting-Ting Liu
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - De-Feng Li
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Qi Tang
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Ying-Xue Li
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Ding-Guo Zhang
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Zheng-Lei Xu
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Hong-Tao Jin
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Li-Sheng Wang
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Jun Yao
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
20
|
Ogino H, Fukaura K, Iboshi Y, Nagamatsu Y, Okuno H, Nishioka K, Nishihara Y, Tanaka Y, Chinen T, Ihara E, Ogawa Y. Role of the IL-23-T-bet/GATA3 Axis for the Pathogenesis of Ulcerative Colitis. Inflammation 2020; 44:592-603. [PMID: 33040251 DOI: 10.1007/s10753-020-01358-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/02/2020] [Indexed: 11/28/2022]
Abstract
Ulcerative colitis (UC) has been considered a Th2- and Th17-related disease. However, anti-IL-12/23 p40 antibody, which blocks Th1 and Th17 cell induction and maintenance, has shown efficacy in treating UC, suggesting that UC might not be a prototypical Th2 and Th17 cell-mediated autoimmune disease. To verify how the immune responses in UC patients interact with each other, we analyzed the cytokine expression and transcription factors involved in the Th1, Th2, and Th17 responses. The mucosal expression of 19 cytokines and transcription factors related to Th1, Th2, and Th17 cells, as well as Tregs, were measured by quantitative polymerase chain reaction using endoscopic biopsy specimens from inflamed colons of UC patients. A correlation analysis between the cytokines and transcription factors was conducted. The characteristic cytokine profile in UC patients has two immune response clusters: Th17-related responses and Th1-/Th2-related responses. IL-23 showed a weaker association with Th17 cell-related cytokines and transcription factor RORC and a much stronger correlation with T-bet and GATA3. In the high-IL-23-expression group, the rate of chronic continuous type was higher and the remission rate lower than in the low-IL-23-expression group. IL-23 may be a very important cytokine for evaluating the UC disease condition, as the expression of IL-23 is associated with certain clinical characteristics of UC patients. A unique association between IL-23 and T-bet/GATA3 might play a key role in the pathogenesis of UC.
Collapse
Affiliation(s)
- Haruei Ogino
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-city, 812-8582, Japan.
| | - Keita Fukaura
- Department of Gastroenterology, Saiseikai Futsukaichi Hospital, Fukuoka, Japan
| | - Yoichiro Iboshi
- Department of Gastroenterology, Clinical Research Institute, National Hospital Organization, Kyushu Medical Center, Fukuoka, Japan
| | - Yousuke Nagamatsu
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-city, 812-8582, Japan
| | - Hiroaki Okuno
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-city, 812-8582, Japan.,Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Kei Nishioka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-city, 812-8582, Japan
| | - Yuichiro Nishihara
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-city, 812-8582, Japan
| | - Yoshimasa Tanaka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-city, 812-8582, Japan
| | - Takatoshi Chinen
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-city, 812-8582, Japan
| | - Eikich Ihara
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-city, 812-8582, Japan.,Department of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-city, 812-8582, Japan
| |
Collapse
|
21
|
Interplay between Cytokine Circuitry and Transcriptional Regulation Shaping Helper T Cell Pathogenicity and Plasticity in Inflammatory Bowel Disease. Int J Mol Sci 2020; 21:ijms21093379. [PMID: 32403220 PMCID: PMC7247009 DOI: 10.3390/ijms21093379] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disorder manifested as Crohn’s disease (CD) and ulcerative colitis (UC) characterized by intestinal inflammation and involves a dysregulated immune response against commensal microbiota through the activation of CD4 T helper cells. T helper cell differentiation to effector or regulatory phenotypes is controlled by cytokine networks and transcriptional regulators. Distinct polarized T helper cells are able to alter their phenotypes to adapt to diverse and fluctuating physiological environments. T helper cells exhibit intrinsic instability and flexibility to express cytokines of other lineages or transdifferentiate from one T helper cell type to another in response to various perturbations from physiological cytokine milieu as a means of promoting local immunity in response to injury or ensure tissue homeostasis. Furthermore, functional plasticity and diversity of T helper cells are associated with pathogenicity and are critical for immune homeostasis and prevention of autoimmunity. In this review, we provide deeper insights into the combinatorial extrinsic and intrinsic signals that control plasticity and transdifferentiation of T helper cells and also highlight the potential of exploiting the genetic reprogramming plasticity of T helper cells in the treatment of IBD.
Collapse
|
22
|
A Comprehensive Review and Update on the Pathogenesis of Inflammatory Bowel Disease. J Immunol Res 2019; 2019:7247238. [PMID: 31886308 PMCID: PMC6914932 DOI: 10.1155/2019/7247238] [Citation(s) in RCA: 588] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/15/2019] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and life-threating inflammatory disease of gastroenteric tissue characterized by episodes of intestinal inflammation. The pathogenesis of IBD is complex. Recent studies have greatly improved our knowledge of the pathophysiology of IBD, leading to great advances in the treatment as well as diagnosis of IBD. In this review, we have systemically reviewed the pathogenesis of IBD and highlighted recent advances in host genetic factors, gut microbiota, and environmental factors and, especially, in abnormal innate and adaptive immune responses and their interactions, which may hold the keys to identify novel predictive or prognostic biomarkers and develop new therapies.
Collapse
|
23
|
Hsiao HB, Wu JB, Lin WC. Anti-arthritic and anti-inflammatory effects of (-)-Epicatechin-3-O-β-d-allopyranoside, a constituent of Davallia formosana. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 52:12-22. [PMID: 30599891 DOI: 10.1016/j.phymed.2018.09.192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 06/04/2018] [Accepted: 09/17/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND (-)-Epicatechin-3-O-β-d-allopyranoside (ECAP) is isolated from the popular Chinese herbal medicine Davallia formosana, which has been used to treat bone diseases including bone fracture, arthritis, and osteoporosis. PURPOSE To investigate the antiarthritic and the anti-inflammatory effect of ECAP on a mouse model of collagen-induced arthritis (CIA) and in vitro. METHODS Male DBA/1 J mice were immunized by administering an intradermal injection of 100 µg of type II collagen in Freund's complete adjuvant. The control groups (vehicle) and ECAP were administered orally at doses of 1 ml/kg (H2O), 50 and 100 mg/ml/kg once a day from Day 22 to Day 42 after primary immunization. Paw swelling, arthritis severity score, and histological changes were examined. Enzyme-linked immunosorbent assay was used to measure the levels of cytokines, including tumor necrosis factor alpha (TNF-α), interleukin (IL)-10, IL-17, IL-4, and interferon-γ (IFN-γ), in splenocytes. Furthermore, the anti-inflammatory activities of ECAP were investigated in vitro by measuring nitric oxide (NO) levels in lipopolysaccharide (LPS)-activated RAW264.7 macrophages. RESULTS In the CIA model, the oral administration of ECAP ameliorated paw edema and reduced the arthritis severity score and disease incidence. Histopathological examination demonstrated that ECAP treatment effectively protected the bone and cartilage of knee joints from erosion, lesion formation, and deformation compared with the vehicle treatment. ECAP also reduced IL-1β and MMP-9 expression in inflamed joints. Compared with the vehicle-treated mice with CIA, the reduced severity of the disease in ECAP-treated mice was associated with decreased levels of TNF-α and IL-17 and increased levels of IL-10 and IL-4 in the supernatants of splenocyte cultures. Flow cytometry analysis demonstrated that ECAP increased the population of CD4+CD25+ regulatory T cells, thereby inhibiting the B cell population. Anticollagen IgG1 and IgG2a levels decreased in the serum of ECAP-treated mice. ECAP suppressed LPS-induced NO production in RAW264.7 macrophages. CONCLUSION The administration of ECAP effectively suppressed inflammation and inflammatory pain and adjuvant-induced arthritis, indicating its therapeutic potential in the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Hung-Bo Hsiao
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Jin-Bin Wu
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Wen-Chuan Lin
- School of Pharmacy, China Medical University, Taichung, Taiwan.
| |
Collapse
|
24
|
|
25
|
Shen Z, Zhu C, Quan Y, Yang J, Yuan W, Yang Z, Wu S, Luo W, Tan B, Wang X. Insights into Roseburia intestinalis which alleviates experimental colitis pathology by inducing anti-inflammatory responses. J Gastroenterol Hepatol 2018. [PMID: 29532517 DOI: 10.1111/jgh.14144] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM The study aims to elucidate the anti-inflammatory effect and mechanism of Roseburia intestinalis (R. intestinalis) in Crohn's disease (CD). METHODS 16S-rRNA genome sequencing technique is used to detect the characteristics of intestinal microbiota in untreated CD patients and healthy controls. Then the study investigates the effects of R. intestinalis on disease activity index score, intestinal pathology, the differentiation of Treg cells, and the expressions of Thymic stromal lymphopoietin (TSLP), TGF-β and IL-10 by using TNBS colitis models. At the cellular level, the study uses LPS to stimulate Caco-2 cells to conduct inflammation models and then co-culture with R. intestinalis and detect changes of TSLP and TGF-β. The study then uses R. intestinalis to stimulate peripheral blood mononuclear cells, and the change of Treg cells was detected. RESULTS Genome sequencing of fecal samples from untreated CD patients (n = 10) revealed decreases in the abundance and diversity of intestinal microbiota, including R. intestinalis. Moreover, R. intestinalis reduced disease activity index scores, colon shortening, intestinal mucosal epithelial injury, and mucosal lymphocyte infiltration in a colitis mice model. It suppressed intestinal inflammation by increasing Treg cell numbers and expression of the anti-inflammatory cytokines TSLP, TGF-β, and interleukin-10 (P < 0.05). R. intestinalis also increased secretion of TSLP and TGF-β in lipopolysaccharide-treated Caco-2 cells. CONCLUSION These findings suggest that R. intestinalis suppresses CD pathogenesis by inducing anti-inflammatory responses.
Collapse
Affiliation(s)
- Zhaohua Shen
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Non-resolving Inflammation and Cancer, Changsha, Hunan, China
| | - Changxin Zhu
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Non-resolving Inflammation and Cancer, Changsha, Hunan, China
| | - Yongsheng Quan
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Non-resolving Inflammation and Cancer, Changsha, Hunan, China
| | - Jinming Yang
- Department of Pharmacology and The Penn State Cancer Institute, The Pennsylvania State University College of Medicine, and Milton S Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Wei Yuan
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Non-resolving Inflammation and Cancer, Changsha, Hunan, China
| | - Zhenyu Yang
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Non-resolving Inflammation and Cancer, Changsha, Hunan, China
| | - Shuai Wu
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Non-resolving Inflammation and Cancer, Changsha, Hunan, China
| | - Weiwei Luo
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Non-resolving Inflammation and Cancer, Changsha, Hunan, China
| | - Bei Tan
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Non-resolving Inflammation and Cancer, Changsha, Hunan, China
| | - Xiaoyan Wang
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Non-resolving Inflammation and Cancer, Changsha, Hunan, China
| |
Collapse
|
26
|
Thomsen M, Clarke S, Vitetta L. The role of adjuvant probiotics to attenuate intestinal inflammatory responses due to cancer treatments. Benef Microbes 2018; 9:899-916. [PMID: 30232908 DOI: 10.3920/bm2017.0172] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chemotherapy and radiotherapy treatment regimens for gastrointestinal, peritoneal and pelvic tumours can disrupt the intestinal microbiome and intestinal epithelia. Such disturbances can provoke symptoms such as diarrhoea, nausea and vomiting. Chemotherapy and radiotherapy induced gastrointestinal toxicity aggravating intestinal microbiome dysbiosis is postulated to adversely alter the intestinal microbiome, with a consequent induced pro-inflammatory effect that disrupts the intestinal microbiome-epithelia-mucosal immunity axis. Although not widely recognised, the intestinal mucosa is the largest and most densely and dynamically populated immune-environment. Cancer treatment adverse effects that affect intestinal and mucosal cells inadvertently target and disrupt resident intestinal macrophages, the cells that marshal immune activity in the intestinal mucosa by shaping pro-inflammatory and anti-inflammatory activities to control and eradicate infectious insults and maintain local homeostasis. Pathobionts (bacteria capable of pathogenic pro-inflammatory activity) and noxious environmental and bacterial antigens use the intestinal epithelia and gap junctions as a point of entry into the systemic circulation. This translocation movement promotes toxic sequelae that obstruct intestinal macrophage functions resulting in uncontrolled local and systemic pro-inflammatory activity, loss of phagocytic function and loss of expression of tight junction proteins. Probiotic bacteria as an adjunctive treatment shows efficacy in ameliorating enteropathies such as mucositis/diarrhoea resulting from chemotherapy or radiotherapy regimens. As such we posit that an important benefit that warrants a further focused research effort is the administration of adjuvant probiotics to help reduce the incidence of febrile neutropenia.
Collapse
Affiliation(s)
- M Thomsen
- 1 The University of Sydney, School of Medicine, Faculty of Medicine and Health, NSW 2006, Australia
| | - S Clarke
- 1 The University of Sydney, School of Medicine, Faculty of Medicine and Health, NSW 2006, Australia.,2 Northern Clinical School, Kolling Institute of Medical Research, Pacific Hwy, St Leonards NSW 2065, Australia
| | - L Vitetta
- 1 The University of Sydney, School of Medicine, Faculty of Medicine and Health, NSW 2006, Australia.,3 Medlab Clinical Ltd., 66 McCauley St., Sydney, 2006 NSW, Australia
| |
Collapse
|
27
|
Shen Y, Xie Y, Zhao Y, Long Y, Li L, Zeng Y. Pim-1 inhibitor attenuates trinitrobenzene sulphonic acid induced colitis in the mice. Clin Res Hepatol Gastroenterol 2018; 42:382-386. [PMID: 29551611 DOI: 10.1016/j.clinre.2018.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/28/2017] [Accepted: 01/02/2018] [Indexed: 02/04/2023]
Abstract
Pim-1 kinase has been implicated in inflammatory bowel disease (IBD). This study aimed to evaluate the application of Pim-1 inhibitor (PIM-Inh) for the treatment of IBD. Mouse model of IBD was established by the treatment with trinitrobenzene sulphonic acid (TNBS). The results showed that disease activity index score was significantly decreased, colon length was significantly increased while Wallace score and pathological score were significantly decreased after PIM-Inh treatment compared to TNBS model group. In addition, GATA3 and ROR-γt mRNA and protein levels significantly increased but Foxp3 mRNA and protein levels significantly decreased in mice with TNBS treatment compared to mice without TNBS treatment. Administration of PIM-Inh caused significant decreases in GATA3, T-bet and ROR-γt mRNA and protein levels as well as significant increases in FOXP3 mRNA and protein levels. In conclusion, our data suggest that Pim-1 kinase inhibitor could attenuate IBD by promoting T-cell differentiation into Foxp3+ regulatory T-cells and is a promising agent for IBD therapy.
Collapse
Affiliation(s)
- Yueming Shen
- Department of Digestive Diseases, Changsha Central Hospital, No. 163, Shaoshan Nanlu, Changsha 410004, China
| | - Yuanhong Xie
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, No. 145, Shandong Zhonglu, Shanghai 200001, China
| | - Yan Zhao
- Department of Digestive Diseases, Changsha Central Hospital, No. 163, Shaoshan Nanlu, Changsha 410004, China.
| | - Yan Long
- Department of Digestive Diseases, Changsha Central Hospital, No. 163, Shaoshan Nanlu, Changsha 410004, China
| | - Lingqian Li
- Department of Digestive Diseases, Changsha Central Hospital, No. 163, Shaoshan Nanlu, Changsha 410004, China
| | - Ya Zeng
- Department of Digestive Diseases, Changsha Central Hospital, No. 163, Shaoshan Nanlu, Changsha 410004, China
| |
Collapse
|
28
|
Liu J, Wang H, Li Y, Shi P, Gong J, Gu L, Zhu W, Li J. Anti-mouse CD52 Treatment Ameliorates Colitis through Suppressing Th1/17 Mediated Inflammation and Promoting Tregs Differentiation in IL-10 Deficient Mice. Biol Pharm Bull 2018; 41:1423-1429. [PMID: 29899181 DOI: 10.1248/bpb.b18-00261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent studies suggested that excessive T helper (Th)1/17 cells concomitant with regulatory T cell deficiency might play important roles in Crohn's disease. Anti-cluster of differentiation 52 (CD52) monoclonal antibody (mAb), which aims on CD52 antigen on mature immunocytes, has both T cell depletion and immunosuppressive activities. In this study, we evaluated the therapeutic effects and possible mechanisms of anti-CD52 treatment on interleukin-10 (IL-10) deficient mouse. Anti-mouse CD52 mAb was administered to C3H/HeJBir.IL-10-/- (C3H.IL-10-/-) mice intraperitoneally 20 µg per week for 2 weeks. The disease activity index, body weight, the histological grading of colitis, and levels of tumor necrosis factor (TNF)-α, interferon (IFN)-γ, IL-17 and IL-6 in colon were quantified after treatment. In addition, CD25, Forkhead box P3 (Foxp3) and transforming growth factor (TGF)-β gene as well as the percentage of CD25+Foxp3+ T cells in colon were also measured. The severity of colitis in IL-10-/- mice was significantly decreased by the treatment, with improvement of colon histological grade. The treatment also decreased the TNF-α, IFN-γ, IL-17 and IL-6 levels in colon. Furthermore, the treatment up-regulated the mRNA expression of CD25, Foxp3 and TGF-β gene as well as the percentage of CD25+Foxp3+ T cells in colon lamina propria mononuclear cells (LPMCs) of IL-10-/- mice. Our data might indicate that anti-CD52 treatment could ameliorate the colitis of C3H.IL-10-/- mice and it might be related to the suppression of Th1/17 related inflammation and the promotion of regulatory T cell differentiation. Thus, our data reveals that anti-CD52 treatment may hold potential for clinical applications for Crohn's disease treatment.
Collapse
Affiliation(s)
- Jianhui Liu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University.,Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University
| | - Honggang Wang
- Department of General Surgery, Taizhou People's Hospital, Taizhou Clinical Medical College of Nanjing Medical University
| | - Yi Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University
| | - Peiliang Shi
- Model Animal Research Center of Nanjing University
| | - Jianfeng Gong
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University
| | - Lili Gu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University
| | - Weiming Zhu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University
| | - Jieshou Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University
| |
Collapse
|
29
|
Shibata A, Uga K, Sato T, Sagara M, Igaki K, Nakamura Y, Ochida A, Kono M, Shirai J, Yamamoto S, Yamasaki M, Tsuchimori N. Pharmacological inhibitory profile of TAK-828F, a potent and selective orally available RORγt inverse agonist. Biochem Pharmacol 2018; 150:35-45. [DOI: 10.1016/j.bcp.2018.01.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 01/09/2018] [Indexed: 12/11/2022]
|
30
|
Anti-IL-23 receptor monoclonal antibody prevents CD4+ T cell-mediated colitis in association with decreased systemic Th1 and Th17 responses. Eur J Pharmacol 2018; 824:163-169. [DOI: 10.1016/j.ejphar.2018.01.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 12/19/2022]
|
31
|
The Brain-Intestinal Mucosa-Appendix- Microbiome-Brain Loop. Diseases 2018; 6:diseases6020023. [PMID: 29614774 PMCID: PMC6023457 DOI: 10.3390/diseases6020023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 02/28/2018] [Accepted: 03/29/2018] [Indexed: 12/20/2022] Open
Abstract
The brain and the gut are connected from early fetal life. The mother's exposure to microbial molecules is thought to exert in utero developmental effects on the fetus. These effects could importantly underpin the groundwork for subsequent pathophysiological mechanisms for achieving immunological tolerance and metabolic equilibrium post birth, events that continue through to 3-4 years of age. Furthermore, it is understood that the microbiome promotes cues that instruct the neonate's mucosal tissues and skin in the language of molecular and cellular biology. Post birth mucosal lymphoid tissue formation and maturation (most probably including the vermiform appendix) is microbiota-encouraged co-establishing the intestinal microbiome with a developing immune system. Intestinal mucosal tissue maturation loops the brain-gut-brain and is postulated to influence mood dispositions via shifts in the intestinal microbiome phyla. A plausible appreciation is that dysregulated pro-inflammatory signals from intestinal resident macrophages could breach the loop by providing adverse mood signals via vagus nerve afferents to the brain. In this commentary, we further suggest that the intestinal resident macrophages act as an upstream traffic controller of translocated microbes and metabolites in order to maintain local neuro-endocrine-immunological equilibrium. When macrophages are overwhelmed through intestinal microbiome and intestinal epithelial cell dysbiosis, pro-inflammatory signals are sustained, which may then lead to mood disorders. The administration of probiotics as an adjunctive medicine co-administered with antidepressant medications in improving depressed mood may have biological and clinical standing.
Collapse
|
32
|
Zhang B, Liu Y, Lan X, Xu X, Zhang X, Li X, Zhao Y, Li G, Du C, Lu S, Wang H. Oral Escherichia coli expressing IL-35 meliorates experimental colitis in mice. J Transl Med 2018; 16:71. [PMID: 29554971 PMCID: PMC5859778 DOI: 10.1186/s12967-018-1441-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 03/05/2018] [Indexed: 12/21/2022] Open
Abstract
Background Ulcerative colitis (UC) is a type of inflammatory bowel disease (IBD) characterized by chronic inflammation of colon. It is commonly believed that the imbalance of immune system and overwhelming production of cytokines are involved in the pathogenesis of UC. Recent studies demonstrated that interleukin-35 (IL-35), a key player in the regulation of inflammation, has been identified as potential therapeutic target to treat UC. However, conventional intravenous administration is costly and inconvenient. The present study was designed to establish a novel IL-35 delivery system and investigate its therapeutic effects on dextran sulfate sodium (DSS)-induced experimental colitis in mice for the first time. Methods An engineered Escherichia coli (E. coli/IL-35) expressing IL-35 was constructed. Adult male BALB/c mice randomly got the oral administration of E. coli/IL-35, empty plasmid-transformed E. coli (E. coli0) or PBS for treatment following ingestion of 3% DSS solution for 5 days. Normal mice were used as control group. Colonic and splenic tissues were collected on day 10 post-DSS-induction. Clinical signs, disease activity index (DAI), pathological and immunohistological changes, cytokine profiles and cell populations were evaluated. Results Intragastric administration of E. coli/IL-35 effectively protected the colitis mice from DSS assimilation including weight loss and colon shortening. Pathological analysis showed significantly lower DAI score and much less intra-colon infiltration of neutrophils and CD3+ cells in the IL-35 treated group. Moreover, E. coli/IL-35-treated mice demonstrated much less CD4+ IL-17A+ Th17 cells and a higher level of CD4+CD25+Foxp3+ Tregs in spleen and mesenteric lymph nodes, as well as increased colon and serum level of IL-10 and IL-35 and decreased levels of IL-6. Conclusions Our study showed that E. coli/IL-35 as a novel oral IL-35 delivery system alleviated inflammatory damage of colonic tissue in the colitic mice. Genetic therapeutic strategies using engineered E. coli encoding immunoregulatory cytokines may provide a potential approach for the treatment of IBD.
Collapse
Affiliation(s)
- Baoren Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Yi Liu
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xu Lan
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Xiaoxi Xu
- Department of Endocrinology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoning Zhang
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiang Li
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Yiming Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Guang Li
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Caigan Du
- Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada.,Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Shanzheng Lu
- Department of Anorectal Surgery, People's Hospital of Hunan Province, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China. .,Tianjin General Surgery Institute, Tianjin, China.
| |
Collapse
|
33
|
Saltzman ET, Palacios T, Thomsen M, Vitetta L. Intestinal Microbiome Shifts, Dysbiosis, Inflammation, and Non-alcoholic Fatty Liver Disease. Front Microbiol 2018; 9:61. [PMID: 29441049 PMCID: PMC5797576 DOI: 10.3389/fmicb.2018.00061] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 01/10/2018] [Indexed: 12/16/2022] Open
Abstract
Adverse fluctuations in the distribution of the intestinal microbiome cohort has been associated with the onset of intra- and extra-intestinal inflammatory conditions, like the metabolic syndrome (MetS) and it's hepatic manifestation, non-alcoholic fatty liver disease (NAFLD). The intestinal microbial community of obese compared to lean subjects has been shown to undergo configurational shifts in various genera, including but not limited to increased abundances of Prevotella, Escherichia, Peptoniphilus, and Parabacteroides and decreased levels of Bifidobacteria, Roseburia, and Eubacteria genera. At the phylum level, decreased Bacteroidetes and increased Firmicutes have been reported. The intestinal microbiota therefore presents an important target for designing novel therapeutic modalities that target extra-intestinal inflammatory disorders, such as NAFLD. This review hypothesizes that disruption of the intestinal-mucosal macrophage interface is a key factor in intestinal-liver axis disturbances. Intestinal immune responses implicated in the manifestation, maintenance and progression of NAFLD provide insights into the dialogue between the intestinal microbiome, the epithelia and mucosal immunity. The pro-inflammatory activity and immune imbalances implicated in NAFLD pathophysiology are reported to stem from dysbiosis of the intestinal epithelia which can serve as a source of hepatoxic effects. We posit that the hepatotoxic consequences of intestinal dysbiosis are compounded through intestinal microbiota-mediated inflammation of the local mucosa that encourages mucosal immune dysfunction, thus contributing important plausible insight in NAFLD pathogenesis. The administration of probiotics and prebiotics as a cure-all remedy for all chronic diseases is not advocated, instead, the incorporation of evidence based probiotic/prebiotic formulations as adjunctive modalities may enhance lifestyle modification management strategies for the amelioration of NAFLD.
Collapse
Affiliation(s)
- Emma T. Saltzman
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| | - Talia Palacios
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| | - Michael Thomsen
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| | - Luis Vitetta
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| |
Collapse
|
34
|
Lee SH, Kwon JE, Cho ML. Immunological pathogenesis of inflammatory bowel disease. Intest Res 2018; 16:26-42. [PMID: 29422795 PMCID: PMC5797268 DOI: 10.5217/ir.2018.16.1.26] [Citation(s) in RCA: 374] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory state of the gastrointestinal tract and can be classified into 2 main clinical phenomena: Crohn's disease (CD) and ulcerative colitis (UC). The pathogenesis of IBD, including CD and UC, involves the presence of pathogenic factors such as abnormal gut microbiota, immune response dysregulation, environmental changes, and gene variants. Although many investigations have tried to identify novel pathogenic factors associated with IBD that are related to environmental, genetic, microbial, and immune response factors, a full understanding of IBD pathogenesis is unclear. Thus, IBD treatment is far from optimal, and patient outcomes can be unsatisfactory. As result of massive studying on IBD, T helper 17 (Th17) cells and innate lymphoid cells (ILCs) are investigated on their effects on IBD. A recent study of the plasticity of Th17 cells focused primarily on colitis. ILCs also emerging as novel cell family, which play a role in the pathogenesis of IBD. IBD immunopathogenesis is key to understanding the causes of IBD and can lead to the development of IBD therapies. The aim of this review is to explain the pathogenesis of IBD, with a focus on immunological factors and therapies.
Collapse
Affiliation(s)
- Seung Hoon Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Jeong eun Kwon
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
35
|
Bian LQ, Bi Y, Zhou SW, Chen ZD, Wen J, Shi J, Mao L, Wang L. T cell responses in senior patients with community-acquired pneumonia related to disease severity. Exp Cell Res 2017; 361:56-62. [PMID: 28986067 DOI: 10.1016/j.yexcr.2017.09.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/27/2017] [Accepted: 09/30/2017] [Indexed: 12/19/2022]
Abstract
Senior individuals older than 65 years of age are at a disproportionally higher risk of developing pneumonia. Impaired capacity to defend against airway infections may be one of the reasons. It is generally believed that weaker regulatory T cell responses may be beneficial to host defense against pathogens. In senior patients with community-acquired bacterial pneumonia, we investigated the frequencies and functions of regulatory T cells. Interestingly, we found that compared to age- and sex-matched healthy controls, senior pneumonia patients presented lower frequencies of Foxp3-expressing and Helios-expressing CD4+ T cells. The quantity of Foxp3 and Helios being expressed, measured by their mRNA transcription levels, was also lower in CD4+ T cells from pneumonia patients. Furthermore, following TCR and TGF-β stimulation, pneumonia patients presented impaired capacity to upregulate Foxp3 and Helios. Functional analyses revealed that CD4+ T cells from pneumonia patients secreted lower amounts of IL-10 and TGF-β, two cytokines critical to regulatory T cell-mediated suppression. Also, the expression of granzyme B and perforin, which were cytolytic molecules potentially utilized by regulatory T cells to mediate the elimination of antigen-presenting cells and effector T cells, were reduced in CD4+CD25+ T cells from senior pneumonia patients. In addition, the CD4+CD25+ T cells from senior pneumonia patients presented reduced capacity to suppress effector CD4+ and CD8+ T cell proliferation. Moreover, the value of pneumonia severity index was inversely correlated with several parameters of regulatory T cell function. Together, our results demonstrated that senior pneumonia patients presented a counterintuitive impairment in regulatory T cell responses that was associated with worse prognosis.
Collapse
Affiliation(s)
- Lu-Qin Bian
- Department of Special Procurement Ward, The First Affiliated Hospital of Soochow University, Suzhou 215006 China; Department of Pneumoconiosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433 China
| | - Ying Bi
- Department of Pneumoconiosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433 China
| | - Shao-Wei Zhou
- Department of Pneumoconiosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433 China
| | - Zi-Dan Chen
- Department of Pneumoconiosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433 China
| | - Jun Wen
- Department of Pneumoconiosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433 China
| | - Jin Shi
- Department of Pneumoconiosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433 China
| | - Ling Mao
- Department of Pneumoconiosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433 China.
| | - Ling Wang
- Department of Special Procurement Ward, The First Affiliated Hospital of Soochow University, Suzhou 215006 China.
| |
Collapse
|
36
|
Zhang M, Zhou L, Zhang S, Yang Y, Xu L, Hua Z, Zou X. Bifidobacterium longum affects the methylation level of forkhead box P3 promoter in 2, 4, 6-trinitrobenzenesulphonic acid induced colitis in rats. Microb Pathog 2017; 110:426-430. [PMID: 28733028 DOI: 10.1016/j.micpath.2017.07.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/11/2017] [Accepted: 07/17/2017] [Indexed: 01/03/2023]
Abstract
Bifidobacterium longum (B. Longum) is a common probiotic colonized in the human gut and against the development of chronic inflammation including inflammatory bowel disease (IBD). But the underlying mechanism remains unknown. The aim of this study was to evaluate the affection of B. longum on the methylation levels of forkhead box P3 (Foxp3) promoter. 2, 4, 6-trinitrobenzenesulphonic acid (TNBS)-induced colitis rat models were treated with B. longum or medium, respectively. The genomic DNA of spleen peripheral blood mononuclear cells (PBMC) cells was extracted. After bisulphite treatment and pyrosequencing, the methylation levels of each CpG sites in the promoter of forkhead box protein P3 (Foxp3) were analyzed. B. Longum treatment changes the methylation level in Foxp3 promoter in TNBS-treated colitis rats, and significantly demethylates several CpG sites in Foxp3 promoter. The demethylation of Foxp3 promoter might be involved in the effectiveness of B. Longum treatment for IBD. Further research remains necessary to investigate the role of B. Longum in Foxp3 demethylation. Using B. Longum or its metabolic products is an option for further investigations on potential treatments for IBD.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Gastroenterology, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Clinical Medical Center of Digestive Disease, China
| | - Lixing Zhou
- Department of Gastroenterology, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Clinical Medical Center of Digestive Disease, China
| | - Shu Zhang
- Department of Gastroenterology, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Clinical Medical Center of Digestive Disease, China
| | - Yang Yang
- Department of Gastroenterology, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Clinical Medical Center of Digestive Disease, China
| | - Lei Xu
- Department of Gastroenterology, Nanjing Medical University Affiliated Drum Tower Clinical Medical College, Nanjing 210008, China
| | - Zichun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, China
| | - Xiaoping Zou
- Department of Gastroenterology, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing 210008, China; Jiangsu Clinical Medical Center of Digestive Disease, China.
| |
Collapse
|
37
|
Hirota T, Tsuboi H, Iizuka-Koga M, Takahashi H, Asashima H, Yokosawa M, Kondo Y, Ohta M, Wakasa Y, Matsumoto I, Takaiwa F, Sumida T. Suppression of glucose-6-phosphate-isomerase induced arthritis by oral administration of transgenic rice seeds expressing altered peptide ligands of glucose-6-phosphate-isomerase. Mod Rheumatol 2017; 27:457-465. [DOI: 10.1080/14397595.2016.1218598] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Tomoya Hirota
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan and
| | - Hiroto Tsuboi
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan and
| | - Mana Iizuka-Koga
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan and
| | - Hiroyuki Takahashi
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan and
| | - Hiromitsu Asashima
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan and
| | - Masahiro Yokosawa
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan and
| | - Yuya Kondo
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan and
| | - Masaru Ohta
- Functional Transgenic Crops Research Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Yuhya Wakasa
- Functional Transgenic Crops Research Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Isao Matsumoto
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan and
| | - Fumio Takaiwa
- Functional Transgenic Crops Research Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan and
| |
Collapse
|
38
|
Tarique M, Saini C, Naqvi RA, Khanna N, Sharma A, Rao DN. IL-12 and IL-23 modulate plasticity of FoxP3 + regulatory T cells in human Leprosy. Mol Immunol 2017; 83:72-81. [PMID: 28110210 DOI: 10.1016/j.molimm.2017.01.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 12/22/2016] [Accepted: 01/07/2017] [Indexed: 12/17/2022]
Abstract
Leprosy is a bacterial disease caused by M. leprae. Its clinical spectrum reflects the host's immune response to the M. leprae and provide an ideal model to investigate the host pathogen interaction and immunological dysregulation. Tregs are high in leprosy patients and responsible for immune suppression of the host by producing IL-10 and TGF-β cytokines. In leprosy, plasticity of Tregs remain unstudied. This is the first study describing the conversion of Tregs into Th1-like and Th17-like cells using in vitro cytokine therapy in leprosy patients. Peripheral blood mononuclear cells from leprosy patients were isolated and stimulated with M. leprae antigen (MLCwA), rIL-12 and rIL-23 for 48h. Expression of FoxP3 in CD4+CD25+ Tregs, intracellular cytokines IFN-γ, TGF-β, IL-10 and IL-17 in Tregs cells were evaluated by flow cytometry (FACS) after stimulation. rIL-12 treatment increases the levels of pStat4 in Tregs and IFN-γ production. In the presence of rIL-23, pStat3+ and IL-17A+ cells increase. rIL-12 and r-IL-23 treatment downregulated the FoxP3 expression, IL-10 and TGF-β production by Tregs and enhances the expression of co-stimulatory molecules (CD80, CD86). In conclusion rIL-12 converts Tregs into IFN-γ producing cells through STAT-4 signaling while rIL-23 converts Tregs into IL-17 producing cells through STAT-3 signaling in leprosy patients. This study may helpful to provide a new avenue to overcome the immunosuprression in leprosy patients using in vitro cytokine.
Collapse
Affiliation(s)
- Mohd Tarique
- Department of Biochemistry, All India Institute of Medical Sciences, AIIMS, New Delhi, 110029, India
| | - Chaman Saini
- Department of Biochemistry, All India Institute of Medical Sciences, AIIMS, New Delhi, 110029, India
| | - Raza Ali Naqvi
- Department of Biochemistry, All India Institute of Medical Sciences, AIIMS, New Delhi, 110029, India
| | - Neena Khanna
- Department of Dermatovenerology, All India Institute of Medical Sciences, AIIMS, New Delhi, 110029, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, AIIMS, New Delhi, 110029, India.
| | - D N Rao
- Department of Biochemistry, All India Institute of Medical Sciences, AIIMS, New Delhi, 110029, India; GITAM Institute of Medical Sciences and Research, GITAM University, Rushikonda, Visakhapatnam, 530045, India.
| |
Collapse
|
39
|
Stephen-Victor E, Sharma VK, Das M, Karnam A, Saha C, Lecerf M, Galeotti C, Kaveri SV, Bayry J. IL-1β, But Not Programed Death-1 and Programed Death Ligand Pathway, Is Critical for the Human Th17 Response to Mycobacterium tuberculosis. Front Immunol 2016; 7:465. [PMID: 27867382 PMCID: PMC5095489 DOI: 10.3389/fimmu.2016.00465] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/17/2016] [Indexed: 01/29/2023] Open
Abstract
The programed death-1 (PD-1)–programed death ligand-1 (PD-L1) and PD-L2 co-inhibitory pathway has been implicated in the evasion strategies of Mycobacterium tuberculosis. Specifically, M. tuberculosis-induced PD-L1 orchestrates expansion of regulatory T cells and suppression of Th1 response. However, the role of PD pathway in regulating Th17 response to M. tuberculosis has not been investigated. In the present report, we demonstrate that M. tuberculosis and M. tuberculosis-derived antigen fractions have differential abilities to mediate human monocyte- and dendritic cell (DC)-mediated Th17 response and were independent of expression of PD-L1 or PD-L2 on aforementioned antigen-presenting cells. Importantly, we observed that blockade of PD-L1 or PD-1 did not significantly modify either the frequencies of Th17 cells or the production of IL-17 from CD4+ T cells though IFN-γ response was significantly enhanced. On the contrary, IL-1β from monocytes and DCs were critical for the Th17 response to M. tuberculosis. Together, our results indicate that IL-1β, but not members of the programed death pathway, is critical for human Th17 response to M. tuberculosis.
Collapse
Affiliation(s)
- Emmanuel Stephen-Victor
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, France; UMR S 1138, Sorbonne Universités, UPMC Univ Paris, Paris, France; Centre de Recherche des Cordeliers, Equipe - Immunopathology and Therapeutic Immunointervention, Paris, France
| | - Varun Kumar Sharma
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, France; Centre de Recherche des Cordeliers, Equipe - Immunopathology and Therapeutic Immunointervention, Paris, France
| | - Mrinmoy Das
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, France; UMR S 1138, Sorbonne Universités, UPMC Univ Paris, Paris, France; Centre de Recherche des Cordeliers, Equipe - Immunopathology and Therapeutic Immunointervention, Paris, France
| | - Anupama Karnam
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, France; UMR S 1138, Sorbonne Universités, UPMC Univ Paris, Paris, France; Centre de Recherche des Cordeliers, Equipe - Immunopathology and Therapeutic Immunointervention, Paris, France
| | - Chaitrali Saha
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, France; Centre de Recherche des Cordeliers, Equipe - Immunopathology and Therapeutic Immunointervention, Paris, France
| | - Maxime Lecerf
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, France; UMR S 1138, Sorbonne Universités, UPMC Univ Paris, Paris, France; Centre de Recherche des Cordeliers, Equipe - Immunopathology and Therapeutic Immunointervention, Paris, France; UMR S 1138, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Caroline Galeotti
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, France; UMR S 1138, Sorbonne Universités, UPMC Univ Paris, Paris, France; Centre de Recherche des Cordeliers, Equipe - Immunopathology and Therapeutic Immunointervention, Paris, France
| | - Srinivas V Kaveri
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, France; UMR S 1138, Sorbonne Universités, UPMC Univ Paris, Paris, France; Centre de Recherche des Cordeliers, Equipe - Immunopathology and Therapeutic Immunointervention, Paris, France; UMR S 1138, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale Unité 1138, Paris, France; UMR S 1138, Sorbonne Universités, UPMC Univ Paris, Paris, France; Centre de Recherche des Cordeliers, Equipe - Immunopathology and Therapeutic Immunointervention, Paris, France; UMR S 1138, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
40
|
Sang L, Chang B, Zhu J, Yang F, Li Y, Jiang X, Sun X, Lu C, Wang D. Dextran sulfate sodium-induced acute experimental colitis in C57BL/6 mice is mitigated by selenium. Int Immunopharmacol 2016; 39:359-368. [PMID: 27533281 DOI: 10.1016/j.intimp.2016.07.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 07/02/2016] [Accepted: 07/29/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND Sodium selenite has been shown to have a protective role in experimental colitis. Th1 and Th17 responses are involved in the pathogenesis of dextran sulfate sodium (DSS)-induced colitis and inflammatory bowel disease. This study investigated whether sodium selenite can suppress Th1/Th17-mediated experimental colitis. METHODS Mice were administered sodium selenite (2μg/g body weight) by gavage daily for 30days. Beginning on day 21, mice were administered 2.5% oral DSS for 9days. The mice were sacrificed on day 31. Survival rates, clinical symptoms, colon lengths, and histological changes were determined. RESULTS Pretreatment with sodium selenite (2μg/g body weight) improved survival rates, colon shortening, body weight loss, disease activity index, and histopathological score in mice with DSS-induced colitis. Pretreatment with sodium selenite restored interleukin-10 and Foxp3 excretion, as well as reducing the levels of interferon-γ and interleukin-17A. CONCLUSIONS Pretreatment with sodium selenite showed therapeutic potential for preventing colitis in mice. This effect may be mediated by the immunomodulation of regulatory T cells, expressing anti-inflammatory genes that suppress Th1 and Th17 responses.
Collapse
Affiliation(s)
- Lixuan Sang
- Department of Immunology, China Medical University, Shenyang, China; Department of Geriatrics, First Affiliated Hospital, China Medical University, Shenyang, China.
| | - Bing Chang
- Department of Gastroenterology, First Affiliated Hospital, China Medical University, Shenyang, China.
| | - Junfeng Zhu
- Department of Immunology, China Medical University, Shenyang, China.
| | - Fangli Yang
- Department of Immunology, China Medical University, Shenyang, China.
| | - Yan Li
- Department of Immunology, China Medical University, Shenyang, China.
| | - Xuefeng Jiang
- Department of Immunology, China Medical University, Shenyang, China.
| | - Xun Sun
- Department of Immunology, China Medical University, Shenyang, China.
| | - Changlong Lu
- Department of Immunology, China Medical University, Shenyang, China.
| | - Danan Wang
- Department of Immunology, China Medical University, Shenyang, China.
| |
Collapse
|
41
|
Gu L, Deng WS, Sun XF, Zhou H, Xu Q. Rapamycin ameliorates CCl4-induced liver fibrosis in mice through reciprocal regulation of the Th17/Treg cell balance. Mol Med Rep 2016; 14:1153-61. [PMID: 27315465 PMCID: PMC4940054 DOI: 10.3892/mmr.2016.5392] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 04/11/2016] [Indexed: 01/04/2023] Open
Abstract
Previous investigations have suggested that the activation of Th17 cells and/or deficiency of regulatory T cells (Tregs) are involved in the pathogenesis of liver fibrosis. The aim of the present study was to investigate the effect of rapamycin on immune responses in a carbon tetrachloride (CCl4)-induced murine liver fibrosis model. Liver fibrosis was induced by intraperitoneal administration with CCl4. Following injection of CCl4, the mice were treated intraperitoneally with rapamycin (1.25 mg/kg/day) for 8 weeks. Hematoxylin and eosin staining and Masson's trichrome staining were used for histological examination. The protein levels of forkhead/winged helix transcription factor P3, retinoic-acid-related orphan receptor (ROR)-γt in liver tissue were determined by western blotting, the frequency of Th17 and Treg cells in the liver was evaluated by flow cytometry, and a suppression assay was measured by incorporating [3H]-thymidine. In addition, to explore the effect of Tregs expanded with rapamycin on hepatic stellate cells (HSC), HSCs were co-cultured with Tregs from rapamycin or phosphate-buffered saline-treated mice. It was found that rapamycin treatment led to a significant reduction in the number of Th17 cells and in the expression levels of ROR-γt in the liver tissues. Simultaneously, the results of the present study showed a significant increase in the frequency of Tregs and a marked enhancement in the expression of forkhead/winged helix transcription factor P3 in the rapamycin-treated mice. Furthermore, the Tregs in rapamycin-treated mice had significantly higher suppressive effects, compared with the cells from mice treated with phospphate-buffered saline. Consequently, rapamycin treatment prevented the development of CCl4-induced hepatic fibrosis, which was shown by its histological appearances. These results suggested that the immunosuppressive effect of rapamycin on liver fibrosis was associated with the suppression of hepatic fibrogenesis and regulation of the Th17/Treg cell balance.
Collapse
Affiliation(s)
- Lei Gu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Wen-Sheng Deng
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Xiao-Fei Sun
- Department of Gastrointestinal Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Hong Zhou
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Qing Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| |
Collapse
|
42
|
Lu HY, Lin BF. Wild bitter melon alleviates dextran sulphate sodium-induced murine colitis by suppressing inflammatory responses and enhancing intestinal regulatory T cells. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.03.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
43
|
Hu S, Chen M, Wang Y, Wang Z, Pei Y, Fan R, Liu X, Wang L, Zhou J, Zheng S, Zhang T, Lin Y, Zhang M, Tao R, Zhong J. mTOR Inhibition Attenuates Dextran Sulfate Sodium-Induced Colitis by Suppressing T Cell Proliferation and Balancing TH1/TH17/Treg Profile. PLoS One 2016; 11:e0154564. [PMID: 27128484 PMCID: PMC4851424 DOI: 10.1371/journal.pone.0154564] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/17/2016] [Indexed: 02/07/2023] Open
Abstract
It has been established that mammalian target of Rapamycin (mTOR) inhibitors have anti-inflammatory effects in models of experimental colitis. However, the underlying mechanism is largely unknown. In this research, we investigate the anti-inflammatory effects of AZD8055, a potent mTOR inhibitor, on T cell response in dextran sulfate sodium (DSS)-induced colitis in mice, a commonly used animal model of inflammatory bowel diseases (IBD). Severity of colitis is evaluated by changing of body weight, bloody stool, fecal consistency, histology evaluation and cytokine expression. We find that AZD8055 treatment attenuates DSS-induced body weight loss, colon length shortening and pathological damage of the colon. And AZD8055 treatment decreases colonic expression of genes encoding the pro-inflammatory cytokines interferon-γ, interleukin (IL)-17A, IL-1β,IL-6 and tumor necrosis factor(TNF)-a and increases colonic expression of anti-inflammatory cytokines IL-10. We show that AZD8055 treatment decreases the percentages of CD4+ T cells and CD8+ T cells in spleen, lymph nodes and peripheral blood of mice. We also find that AZD8055 treatment significantly reduces the number of T helper 1(TH1) cells and TH17 cells and increases regulatory T (Treg) cells in the lamina propria and mesenteric lymph nodes. Furthermore, we demonstrates that AZD8055 suppresses the proliferation of CD4+ and CD8+ T cells and the differentiation of TH1/TH17 cells and expands Treg cells in vitro. The results suggest that, in experimental colitis, AZD8055 exerts anti-inflammatory effect by regulating T helper cell polarization and proliferation.
Collapse
Affiliation(s)
- Shurong Hu
- Department of Gastroenterology, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Mengmeng Chen
- Department of Gastroenterology, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Yilin Wang
- Department of Surgery, Cancer hospital, Fudan University, Shanghai, PR China
| | - Zhengting Wang
- Department of Gastroenterology, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Yaofei Pei
- Department of Surgery, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Rong Fan
- Department of Gastroenterology, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Xiqiang Liu
- Department of Hepatobiliary-Pancreatic Surgery, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang Province, PR China
| | - Lei Wang
- Department of Gastroenterology, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Jie Zhou
- Department of Gastroenterology, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Sichang Zheng
- Department of Gastroenterology, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Tianyu Zhang
- Department of Gastroenterology, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Yun Lin
- Department of Gastroenterology, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Maochen Zhang
- Department of Gastroenterology, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | - Ran Tao
- Department of Hepatobiliary-Pancreatic Surgery, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang Province, PR China
| | - Jie Zhong
- Department of Gastroenterology, Ruijin hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| |
Collapse
|
44
|
The Th17/Treg Immune Imbalance in Ulcerative Colitis Disease in a Chinese Han Population. Mediators Inflamm 2016; 2016:7089137. [PMID: 26977120 PMCID: PMC4763012 DOI: 10.1155/2016/7089137] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/24/2015] [Accepted: 01/05/2016] [Indexed: 01/19/2023] Open
Abstract
Objective. To investigate the Th17/Treg immune balance in the ulcerative colitis (UC) patients in a Chinese Han population. Methods. Ninety UC patients and 30 healthy subjects were enrolled. The serum IL-17 and TGF-β1 levels of these participants were measured with ELISA; the percentage of Th17 and Treg cells in peripheral blood was determined with flow cytometry. Results. In UC patients, the levels of IL-17 and Th17 were significantly higher compared with healthy subjects; the percentage of Th17 and IL-17 level in moderate and severe subgroup was significantly higher than in mild subgroup; a positive correlation existed between these two indexes and clinical activity index and endoscopic evaluation. TGF-β1 level and Treg cells in UC patients were lower than healthy subjects. TGF-β1 level in moderate and severe subgroup was lower than in mild subgroup. There was a negative linear correlation between Treg cells and clinical activity index, endoscopic evaluation. A positive correlation was detected between Treg cells and TGF-β1 level. Conclusions. Th17/Treg immune imbalance might play a crucial role in the development of UC. To induce the production of Treg cells and TGF-β1, inhibit the level of Th17 and IL-17, and thus recover the Th17/Treg immune balance might imply new therapeutic targets in UC management.
Collapse
|
45
|
Sadhu S, Khaitan BK, Joshi B, Sengupta U, Nautiyal AK, Mitra DK. Reciprocity between Regulatory T Cells and Th17 Cells: Relevance to Polarized Immunity in Leprosy. PLoS Negl Trop Dis 2016; 10:e0004338. [PMID: 26751584 PMCID: PMC4709061 DOI: 10.1371/journal.pntd.0004338] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 12/08/2015] [Indexed: 12/17/2022] Open
Abstract
T cell defect is a common feature in lepromatous or borderline lepromatous leprosy (LL/BL) patients in contrast to tuberculoid or borderline tuberculoid type (TT/BT) patients. Tuberculoid leprosy is characterized by strong Th1-type cell response with localized lesions whereas lepromatous leprosy is hallmarked by its selective Mycobacterium leprae specific T cell anergy leading to disseminated and progressive disease. FoxP3+ Regulatory T cells (Treg) which are essential for maintaining peripheral tolerance, preventing autoimmune diseases and limiting chronic inflammatory diseases also dampen proinflammatory T cells that include T helper 17 (Th17) cells. This study is aimed at evaluating the role of Treg cells in influencing other effector T cells and its relationship with the cytokine polarized state in leprosy patients. Peripheral blood mononuclear cells from of BT/TT (n = 15) and BL/LL (n = 15) patients were stimulated with M. leprae antigen (WCL) in presence of golgi transport inhibitor monensin for FACS based intracellular cytokine estimation. The frequency of Treg cells showed >5-fold increase in BL/LL in comparison to BT/TT and healthy contacts. These cells produced suppressive cytokine, IL-10 in BL/LL as opposed to BT/TT (p = 0.0200) indicating their suppressive function. The frequency of Th17 cells (CD4, CD45RO, IL-17) was, however, higher in BT/TT. Significant negative correlation (r = -0.68, P = 0.03) was also found between IL-10 of Treg cells and IL-17+ T cells in BL/LL. Blocking IL-10/TGF-β restored the IL-17+ T cells in BL/LL patients. Simultaneously, presence of Th17 related cytokines (TGF-β, IL-6, IL-17 and IL-23) decreased the number of FoxP3+ Treg cells concomitantly increasing IL-17 producing CD4+ cells in lepromatous leprosy. Higher frequency of Programmed Death-1/PD-1+ Treg cells and its ligand, PDL-1 in antigen presenting cells (APCs) was found in BL/LL patients. Inhibition of this pathway led to rescue of IFN-γ and IL-17 producing T cells. Results indicate that Treg cells are largely responsible for the kind of immunosuppression observed in BL/LL patients. This study also proves that Treg cells are profoundly affected by the cytokine milieu and this property may be utilized for benefit of the host. Polarized T cell response (Th1/Th2 biased) to Mycobacterium leprae (M. leprae) is believed to be a critical element in the pathogenesis of leprosy and its varied clinical manifestations. However, immune response at the pathologic sites of leprosy is an extremely complex process, particularly in the light of recently evidenced heterogeneity of T cell subsets. FoxP3 positive regulatory T cells (Treg) are one of the most potent hierarchic cell types suppressing the effector T cell function with eventual regulation of immune response elicited by the host during intracellular infections. This study shows the recovery of the cell mediated response by CD4+ T cells by inhibiting the suppressive cytokines, IL-10 and TGF-β and also by blocking of the Programmed Death-1 pathway in cells isolated from lepromatous leprosy patients. Reversal of IL-17 immune response was also achieved by modulating the cytokine milieu of in vitro cell culture and hence provides us cues to counter the M. leprae unresponsiveness in leprosy patients.
Collapse
Affiliation(s)
- Soumi Sadhu
- Department of Transplant Immunology and Immunogenetics, AIIMS, New Delhi, India
| | | | - Beenu Joshi
- Immunology Division, National Jalma Institute for Leprosy and Other Mycobacterial Diseases, ICMR, Agra, India
| | - Utpal Sengupta
- Stanley Browne Research Laboratory, The Leprosy Mission, Shahdara, New Delhi, India
| | | | | |
Collapse
|
46
|
Yao J, Wei C, Wang JY, Zhang R, Li YX, Wang LS. Effect of resveratrol on Treg/Th17 signaling and ulcerative colitis treatment in mice. World J Gastroenterol 2015; 21:6572-6581. [PMID: 26074695 PMCID: PMC4458767 DOI: 10.3748/wjg.v21.i21.6572] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/23/2015] [Accepted: 03/12/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine the therapeutic efficacy of resveratrol on ulcerative colitis (UC) and its underlying mechanisms.
METHODS: The mouse UC model was developed using 5% dextran sulfate sodium. Mice were randomly divided into four groups: normal control, UC model group, resveratrol low-dose group (RLD; 50 mg/kg per day), and resveratrol high-dose group (RHD; 100 mg/kg per day).
RESULTS: The results showed that RLD regulates Treg/Th17 balance mainly through reducing the number of Th17 cells, whereas RHD regulates Treg/Th17 balance through both downregulating the number of Th17 cells and upregulating the number of Treg cells. Resveratrol can also regulate the level of plasma and intestinal mucosal cytokines including interleukin (IL)-10, transforming growth factor-β1, IL-6, and IL-17. The expressions of hypoxia inducible factor (HIF)-1α, mammalian target of rapamycin (mTOR), and signal transducer and activator of transcription 3 were significantly decreased in the intestinal tissues of mice treated with resveratrol.
CONCLUSION: The therapeutic efficacy of resveratrol in UC is dose dependent and closely associated with the regulation of Treg/Th17 balance and the HIF-1α/mTOR signaling pathway.
Collapse
|
47
|
Zhang D, Wei C, Yao J, Cai X, Wang L. Interleukin-10 gene-carrying bifidobacteria ameliorate murine ulcerative colitis by regulating regulatory T cell/T helper 17 cell pathway. Exp Biol Med (Maywood) 2015; 240:1622-9. [PMID: 25956685 DOI: 10.1177/1535370215584901] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/10/2015] [Indexed: 12/12/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease suggested to be closely related to the imbalance of regulatory T cell/T helper 17 cell (Treg/Th17) signaling. Previously, we constructed an interleukin-10 (IL-10) expression vector, BL-hIL-10, and proved that it ameliorates dextran sulfate sodium-induced intestinal inflammation in mice. In this study, we further explored the mechanisms underlying BL-hIL-10 treatment from the Treg/Th17 imbalance perspective. Our results showed that the oral administration of BL-hIL-10 reduced the UC inflammation in mice significantly, which was assessed by disease activity index, spleen index, and pathological changes in colon tissue. Moreover, the mice after BL-hIL-10 treatment had increased proportion of Treg cells while Th17 cells decreased greatly, leading to the reconstruction of Treg/Th17 balance. Furthermore, the Th17 cell-secreted factors, such as IL-6, IL-17, and IL-23, were reduced, but the Treg-related factors, IL-10 and Transforming growth factor-β1 (TGF-β1), were elevated accordingly. Finally, Western blot confirmed the inhibition of nuclear hypoxia-inducible factor-1α (HIF-1α) and cytoplasmic mechanistic target of rapamycin (mTOR) and signal transducer and activator of transcription 3 (STAT3) in intestinal tissues. In conclusion, oral administration of BL-hIL-10 can alleviate the inflammation responses of UC in murine model through the restoration of Treg/Th17 imbalance, which might be at least partially due to the inhibition of hypoxia-mTOR-HIF-1α-Th17 axis as well as IL-6-STAT3-HIF-1α-Th17 pathway.
Collapse
Affiliation(s)
- Dingguo Zhang
- Department of Gastroenterology, Jinan University of Medical Sciences, Shenzhen People's Hospital, Engineering Technology Research and Development Center of Shenzhen, Shenzhen, Guangdong 518020, ChinaThe first two authors contributed to this research equally
| | - Cheng Wei
- Department of Gastroenterology, Jinan University of Medical Sciences, Shenzhen People's Hospital, Engineering Technology Research and Development Center of Shenzhen, Shenzhen, Guangdong 518020, ChinaThe first two authors contributed to this research equally
| | - Jun Yao
- Department of Gastroenterology, Jinan University of Medical Sciences, Shenzhen People's Hospital, Engineering Technology Research and Development Center of Shenzhen, Shenzhen, Guangdong 518020, ChinaThe first two authors contributed to this research equally
| | - Xiaoyan Cai
- Department of Gastroenterology, Jinan University of Medical Sciences, Shenzhen People's Hospital, Engineering Technology Research and Development Center of Shenzhen, Shenzhen, Guangdong 518020, ChinaThe first two authors contributed to this research equally
| | - Lisheng Wang
- Department of Gastroenterology, Jinan University of Medical Sciences, Shenzhen People's Hospital, Engineering Technology Research and Development Center of Shenzhen, Shenzhen, Guangdong 518020, ChinaThe first two authors contributed to this research equally
| |
Collapse
|
48
|
Almeida CDS, Andrade-Oliveira V, Câmara NOS, Jacysyn JF, Faquim-Mauro EL. Crotoxin from Crotalus durissus terrificus is able to down-modulate the acute intestinal inflammation in mice. PLoS One 2015; 10:e0121427. [PMID: 25853847 PMCID: PMC4390225 DOI: 10.1371/journal.pone.0121427] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 02/15/2015] [Indexed: 12/30/2022] Open
Abstract
Inflammatory bowel diseases (IBD) is the result of dysregulation of mucosal innate and adaptive immune responses. Factors such as genetic, microbial and environmental are involved in the development of these disorders. Accordingly, animal models that mimic human diseases are tools for the understanding the immunological processes of the IBD as well as to evaluate new therapeutic strategies. Crotoxin (CTX) is the main component of Crotalus durissus terrificus snake venom and has an immunomodulatory effect. Thus, we aimed to evaluate the modulatory effect of CTX in a murine model of colitis induced by 2,4,6- trinitrobenzene sulfonic acid (TNBS). The CTX was administered intraperitoneally 18 hours after the TNBS intrarectal instillation in BALB/c mice. The CTX administration resulted in decreased weight loss, disease activity index (DAI), macroscopic tissue damage, histopathological score and myeloperoxidase (MPO) activity analyzed after 4 days of acute TNBS colitis. Furthermore, the levels of TNF-α, IL-1β and IL-6 were lower in colon tissue homogenates of TNBS-mice that received the CTX when compared with untreated TNBS mice. The analysis of distinct cell populations obtained from the intestinal lamina propria showed that CTX reduced the number of group 3 innate lymphoid cells (ILC3) and Th17 population; CTX decreased IL-17 secretion but did not alter the frequency of CD4+Tbet+ T cells induced by TNBS instillation in mice. In contrast, increased CD4+FoxP3+ cell population as well as secretion of TGF-β, prostaglandin E2 (PGE2) and lipoxin A4 (LXA4) was observed in TNBS-colitis mice treated with CTX compared with untreated TNBS-colitis mice. In conclusion, the CTX is able to modulate the intestinal acute inflammatory response induced by TNBS, resulting in the improvement of clinical status of the mice. This effect of CTX is complex and involves the suppression of the pro-inflammatory environment elicited by intrarectal instillation of TNBS due to the induction of a local anti-inflammatory profile in mice.
Collapse
Affiliation(s)
| | | | | | | | - Eliana L. Faquim-Mauro
- Laboratory of Immunopathology, Butantan Institute, São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
49
|
The th17/treg immune balance in ulcerative colitis patients with two different chinese syndromes: dampness-heat in large intestine and spleen and kidney yang deficiency syndrome. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:264317. [PMID: 25815027 PMCID: PMC4359821 DOI: 10.1155/2015/264317] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 10/07/2014] [Accepted: 10/08/2014] [Indexed: 01/14/2023]
Abstract
Objective. To investigate the Th17/Treg immune balance in the ulcerative colitis (UC) patients with two Chinese syndrome: dampness-heat in large intestine (DHLI) and spleen and kidney Yang deficiency (SKYD). Methods. Ninety UC patients (45 were diagnosed with DHLI and 45 with SKYD syndrome) and 23 healthy people were recruited. The serumIL-17 and TGF-β1 levels of these participants were measured with ELISA; the expression of IL-17 and TGF-β 1 in colonic mucosa tissue was determined with immunohistochemistry and the percentage of Th17 and Treg in peripheral blood with flow cytometry. Results. The levels of IL-17 and Th17 were significantly higher in both DHLI and SKYD groups than in healthy control group and higher in DHLI than in SKYD group (P < 0.05). The levels of TGF-β1 and Treg were significantly lower in the two UC patients groups than in healthy control group; and lower in SKYD group than in DHLI group (P < 0.05). Conclusions. UC with DHLI syndrome could be characterized by the elevation of Th17 and IL-17 levels, which indicated an accentuation of inflammatory reaction; UC with SKYD syndrome could be characterized by the reduction of serum Treg and TGF-β1 levels, which represented a depression of immune tolerance.
Collapse
|
50
|
Sánchez-Fidalgo S, Villegas I, Aparicio-Soto M, Cárdeno A, Rosillo MÁ, González-Benjumea A, Marset A, López Ó, Maya I, Fernández-Bolaños JG, Alarcón de la Lastra C. Effects of dietary virgin olive oil polyphenols: hydroxytyrosyl acetate and 3, 4-dihydroxyphenylglycol on DSS-induced acute colitis in mice. J Nutr Biochem 2015; 26:513-20. [PMID: 25736481 DOI: 10.1016/j.jnutbio.2014.12.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 11/26/2014] [Accepted: 12/02/2014] [Indexed: 01/16/2023]
Abstract
Hydroxytyrosol, a polyphenolic compound from extra virgin olive oil (EVOO) has exhibited an improvement in a model of DSS-induced colitis. However, other phenolic compounds present such as hydroxytyrosyl acetate (HTy-Ac) and 3,4-dihydroxyphenylglycol (DHPG) need to be explored to complete the understanding of the overall effects of EVOO on inflammatory colon mucosa. This study was designed to evaluate the effect of both HTy-Ac and DHPG dietary supplementation in the inflammatory response associated to colitis model. Six-week-old mice were randomized in four dietary groups: sham and control groups received standard diet, and other two groups were fed with HTy-Ac and DHPG, respectively, at 0.1%. After 30 days, all groups except sham received 3% DSS in drinking water for 5 days followed by a regime of 5 days of water. Acute inflammation was evaluated by Disease Activity Index (DAI), histology and myeloperoxidase (MPO) activity. Colonic expression of iNOS, COX-2, MAPKs, NF-kB and FOXP3 were determined by western blotting. Only HTy-Ac-supplemented group showed a significant DAI reduction as well as an improvement of histological damage and MPO. COX-2 and iNOS protein expression were also significantly reduced. In addition, this dietary group down-regulated JNK phosphorylation and prevented the DSS-induced nuclear translocation level of p65. However, no significant differences were observed in the FOXP3 expression. These results demonstrated, for the first time, that HTy-Ac exerts an antiinflammatory effect on acute ulcerative colitis. We concluded that HTy-Ac supplement might provide a basis for developing a new dietary strategy for the prevention of ulcerative colitis.
Collapse
Affiliation(s)
| | - Isabel Villegas
- Department of Pharmacology, Faculty of Pharmacy, University of Seville
| | | | - Ana Cárdeno
- Department of Pharmacology, Faculty of Pharmacy, University of Seville
| | | | | | - Azucena Marset
- Department of Organic Chemistry, Faculty of Pharmacy, University of Seville
| | - Óscar López
- Department of Organic Chemistry, Faculty of Pharmacy, University of Seville
| | - Inés Maya
- Department of Organic Chemistry, Faculty of Pharmacy, University of Seville
| | | | | |
Collapse
|